<<

Neurotherapeutics: The Journal of the American Society for Experimental NeuroTherapeutics

Targeting Voltage-Gated Channels for Neuropathic

Danielle Perret*‡ and Z. David Luo*†

*Department of Anesthesiology & Perioperative Care, †Department of , and ‡Department of Physical & Rehabilitation, School of Medicine, University of California Irvine, Irvine, California 92697

Summary: Voltage-gated calcium channels (VGCC) play lief is limited in some patients, and their long-term use is obligatory roles in diverse physiological functions. Pathologi- limited by their side-effect profiles. Certainly, there is room cal conditions leading to changes in their biophysical properties for improvement in developing more subtype-specific and expression levels may cause malfunctions of VGCC-me- VGCC blockers or modulators for chronic pain conditions. diated activities, resulting in disease states. It is believed that In this review, we summarized the most recent preclinical changes in VGCC properties under pain-inducing conditions and clinical studies related to chronic pain act- may play a causal role in the development of chronic pain, ing on the VGCC. We also included clinical trials aiming to including nerve injury-induced pain or . For expand the application of approved VGCC to different the past several decades, preclinical and clinical research in pain states derived from various pathological conditions, as developing VGCC blockers or modulators for chronic pain well as combination therapies trying to improve the management has been fruitful, leading to some U.S. Food and efficacies and side-effect profiles of current pain medica- Drug Administration-approved drugs currently available for tions. Key Words: Chronic, neuropathic pain, voltage-gated chronic pain management. However, their efficacy in pain re- calcium channels, .

INTRODUCTION derive from different pathological conditions that each mediate chronic pain states by unique mechanisms. A recent survey has indicated that at least 50 million 1 Thus, directing our treatment toward a limited number of people in the United States suffer from chronic pain. It is predicted that this number will increase dramatically targets by medications in our current toolbox could ren- due to advances in health care that will continue to der partial pain relief in some patients only. Combina- prolong the lifespan of patients. In addition to adversely tional treatment is an option to improve efficacy and affecting quality of life, inadequate management of reduce side effects. Unfortunately, most of the currently chronic pain also has profound social, economical, and available medications act through targets that psychological consequences. Current pain medications, are not only important in pain processing, but also crit- both and nonopioid, at best, cause partial pain ical in mediating normal physiological functions, which relief in some, but not all, patients. In addition, long-term therefore lead to intolerable side effects, especially after usage of these medications is often associated with in- long-term usage. Individualized pain management based tolerable side effects, some of which can be life-threat- on pain-inducing pathological conditions would have the ening. Therefore, there is an urgent need for safer and least interference with normal physiological functions, more specific analgesic medications for chronic pain and is therefore an ideal approach in chronic pain man- management. agement. Even though different etiologies of chronic pain may Voltage-gated calcium channels (VGCC) or their sub- have similar clinical manifestations, chronic pain can units are considered one family of molecules with ther- apeutic potentials in chronic pain management. The VGCC are assembled through interactions of different Address correspondence and reprint requests to: Z. David Luo, M.D., 2 ␣ ␣ ␤ ␤ ␣ ␦ ␣ ␦ subunits, namely 1 (CaV 1), (CaV ) 2 (CaV 2 ) Ph.D., Department of Anesthesiology & Perioperative Care, University ␥ ␥ ␣ of California, Irvine Medical Center, Bldg 53, Room 227, 101 The City and (CaV ). So far, 10 channel-forming CaV sub- Dr. South, Orange, CA 92868. E-mail: [email protected]. units, encoded by distinct genes, have been identified.2

Vol. 6, 679–692, October 2009 © The American Society for Experimental NeuroTherapeutics, Inc. 679 680 PERRET AND LUO

␣ These CaV subunits consist of four homologous trans- pain management, especially neuropathic pain manage- membrane regions, and each is composed of six trans- ment. For more generalized reviews about the biophysical membrane domains linked by intracellular loops and properties, and plasticity of VGCC on other disease states, amino and carboxy termini. The diversified physiological the readers are referred to other recent reviews.46,47 and pharmacological properties of VGCC are mainly ␣ derived from the existence of these CaV subunits. Four ␤ DRUGS FOR CaV subunits have been identified so far. They all have 3 ␤ PAIN MANAGEMENT alternative splicing variants. The CaV subunits are en- tirely intracellular, phosphorylated by multiple protein The diversities in biophysical properties and tissue- kinases, including protein kinase C and cAMP-depen- specific expression of VGCC thus become an important dent protein kinase, and they play a critical role in cell issue in drug specificity and safety in developing anal- surface expression and modulating the gating properties gesic drugs for pain management. Data from preclinical ␣ 4 ␣ ␦ of the CaV subunit. Four distinctive CaV 2 genes, studies have indicated that most , including sen- ␣ ␦ ␣ ␦ ␣ ␦ ␣ ␦ 48 49–51 Cav 2 1,Cav 2 2,Cav 2 3, and Cav 2 4 have been sory neurons and spinal dorsal horn neurons, ex- 5,6 identified. Their products and splice variants have spe- press multiple types of VGCC. Several types of VGCC 6–9 ␣ ␦ cific tissue distribution patterns. The CaV 2 subunit are considered potential targets for analgesics based on ␣ ␦ consists of two disulfide-linked ( 2 and ) that their distribution, biophysical/pathological roles, and 10,11 are encoded by the same gene. It is highly glycosy- plasticity under pain-inducing conditions.52 We aim lated and mainly extracellular, with a single transmem- here to highlight both preclinical and clinical advances brane domain and five intracellular carboxyl terminal in VGCC drug use, including combinational therapies, 12-14 amino acids. Data from in vitro studies have shown in pain medicine. A summary of the current VGCC ␣ ␦ that co-expression of CaV 2 with other calcium channel medications that are commonly used for pain manage- 13,15-20 subunits increases and stabilizes current amplitude, ment is available in Table 1. The U.S. Food and Drug channel binding sites, and binding affinity for N-type Administration (FDA)-approved indications for each 19,21 VGCC ligands. Three-dimensional structural analy- are noted. A summary of ongoing clinical trials, as sis of the L-type VGCC by electron cryo-microscopy has listed on clinicaltrials.gov, is available in Table 2, ␣ indicated that the extracellular CaV 2 subunit protrudes which provides the reader with information regarding from the membrane in close proximity to the channel studies using these common VGCC drugs to treat non- ␣ 22 forming CaV subunit. Recent findings have indicated FDA-approved pain etiologies and indications. ␣ ␦ that the CaV 2 subunit is also involved in cellular traf- ficking of the calcium channel complex.23,24 These find- N-type VGCC blockers ␣ ␦ ings suggest that the CaV 2 subunit is likely involved in N-type VGCC are highly expressed in dorsal root gan- ␣ VGCC assembly and stabilization, modulation of CaV glion (DRG) cell bodies and at the presynaptic termi- ␥ subunit functions, and binding. The CaV 1 sub- nals where afferent sensory fibers form with unit is a structural component for the postsynaptic dorsal horn neurons,47,53–55 implying an im- L-type calcium channels.25–27 Whether other identified portant role of these calcium channels in mediating nor- ␥ ␥ CaV or CaV -like subunits associate with other types of mal sensory excitability and re- VGCC remained to be confirmed.28–32 The functional lease. Changes in the biophysical properties and enhanced ␥ role of the CaV subunit is not well understood. expression of these VGCC under pain-inducing patho- Based on their physiological and pharmacological prop- logical conditions would likely enhance synaptic vesicle erties, VGCC can be subdivided into low voltage-activated release of pain-inducing transmitters, such as glutamate,

T-type (Cav3.1, Cav3.2, and Cav3.3), and high voltage- substance P, and calcitonin gene-related on stim- activated L- (Cav1.1 through Cav1.4), N-(Cav2.2), P/Q- ulation that could activate interneurons and projection (Cav2.1), and R-(Cav2.3) types, depending on the channel- neurons, altering sensory excitability and leading to pain ␣ 2,33 forming CaV subunits. All of these five subclasses of sensations. In addition, N-type VGCC are unique in that calcium channels are found in the central and peripheral they are a target for descending activation of adrenergic nervous systems.33 Regulation of intracellular calcium pathways by norepinephrine56 and for inhibition by opi- through activation of these VGCC plays obligatory roles oid pathways.57,58 A role for N-type VGCC in neuro- in: 1) neurotransmitter release, 2) membrane depolariza- pathic pain is solidified by findings indicating that spi- tion and hyperpolarization, 3) enzyme activation and nally delivered N-type calcium channel antagonist can inactivation, and 4) gene regulation.34–40 A large body block nerve injury-induced tactile allodynia,59 and dorsal of data has clearly indicated that VGCC are implicated in horn neuronal responses.50 Evidently, blocking the N- mediating various disease states,38,41 including pain pro- type VGCC at the levels of spinal cord and sensory cessing.40,42–45 This review focuses on recent preclinical neurons results in inhibition of stimulus-evoked release and clinical studies regarding VGCC as targets for chronic of pain-inducing peptides, such as substance P, calcito-

Neurotherapeutics, Vol. 6, No. 4, 2009 CALCIUM CHANNEL DRUGS IN TREATING CHRONIC PAIN 681

Table 1. Summary of Current Calcium-Channel Drugs in Common Clinical Use for Pain Management

Drug FDA Indications Adverse Reactions Reference

Gabapentin 2002: Postherpetic ; , , , http://www.accessdata.fda.gov/ 1994: (partial nausea, dyspepsia, increased appetite, drugsatfda_docs/label/2009/ seizures), pediatric partial 020235s041,020882s028, seizures 021129s027lbl.pdf 2004: Neuropathic pain Dizziness, somnolence, dry mouth, http://www.accessdata.fda.gov/ associated with diabetic edema, blurred vision, weight gain, drugsatfda_docs/label/2009/ ; abnormal thinking (difficulty with 021446s013s014lbl.pdf ; concentration/attention) adjunctive therapy for adult patients with partial onset seizures; fibromyalgia 2004: Management of severe Vertigo, vision blurred, asthenia, http://www.accessdata.fda.gov/ chronic pain in patients for abnormal gait, pyrexia, rigors, sinusitis, drugsatfda_docs/label/2007/ whom intrathecal therapy is anorexia, muscle spasms, pain in limb, 021060s003lbl.pdf warranted, and who are amnesia, , dizziness, dysarthria, intolerant or refractory to other dysgeusia, headache, memory treatment, such as systemic impairment, nausea, , analgesics, adjunctive somnolence, , anxiety, confusion, therapies, or IT , , pruritus, increased sweating

FDA ϭ U.S. Food and Drug Administration; IT ϭ intrathecal.

nin gene-related peptide, and excitatory neurotransmit- VGCC by this toxin leads to diminished neurotransmitter ter, glutamate.43,60–62 Nerve or tissue injury-induced tac- release in the spinal cord,73 and diminished postopera- tile allodynia and thermal , but not acute tive, inflammatory, and neuropathic pain states in animal pain states, are suppressed in mice lacking the N-type models.59,74–76 The analgesic effect of intrathecal zi- ␣ 63–65 calcium channel-forming CaV subunit. This im- conotide is more potent and longer lasting than intrathe- plies that the N-type VGCC are more directly involved in cal morphine without tolerance or cross-tolerance to chronic, rather than acute, nociception. This is consistent morphine analgesia.74,75 with data from direct blockade of N-type VGCC by cone Clinical trials of intrathecal application of this peptide snail peptides ␻-conotonix-GVIA and ␻-conotonix- drug result in significant pain relief for patients with MVIIA (ziconotide or Prialt) that leads to inhibition of severe chronic pain (Table 2), including neuropathic pain neuropathic and inflammatory pain, but not acute pain, in and pain secondary to or AIDS. Several reviews 52 animal models. Interestingly, splicing variants of the summarizing the details of some early clinical trials were N-type VGCC have been identified in sensory neu- 77–79 66–68 recently published. Briefly, the first randomized, rons. With validation of their functional contribu- double-blind, placebo-controlled pilot study for pain re- tion to pain processing, these subtypes of N-type VGCC lief with intrathecal ziconotide was in patients undergo- could be potential targets for specific analgesic drugs. ing surgery.80 Patients received either a placebo or one of ␮ ␮ Peptide N-type VGCC blockers two intrathecal ziconotide doses (0.7 g/h and 7.0 g/h) The FDA approval of ziconotide, or the synthetic ver- postoperatively during the following 48 h. The mean sion of ␻-conotonix-MVIIA (also called SNX-111, or daily morphine consumption from patient-controlled ad- Prialt (Elan Pharmaceuticals, Inc., San Diego, CA), for ministration in the 24- to 48-h period was significantly the treatment of chronic severe pain refractory to other lower in patients receiving ziconotide than in placebo- current pain medications in December 2004 in the United controlled patients. Pain scores in both ziconotide groups States and in Europe69 (Table 1) marked the first clinical were lower than in the placebo group. Adverse events application of N-type VGCC peptide blockers in chronic were more frequent on the higher ziconotide dose. In an pain management. ␻-conotonix-MVIIA is a 25-amino open-label clinical study,81 31 male patients with chronic acid peptide isolated from the marine fish-hunting cone pain who had failed opioid management received con- snail, Conus magus.70 Due to the peptidergic nature of tinuous intrathecal ziconotide infusions. The average this drug, it is only approved for intrathecal application. pain reduction was 43% in 19 out of 24 patients who Ziconotide can block N-type VGCC currents with high completed the study. The concomitant use of was potency, but in a reversible manner.71,72 Data from pre- reduced by at least 50% in 15 patients. Two multicenter, clinical studies have indicated that blocking N-type randomized, double-blind, placebo-controlled trials in

Neurotherapeutics, Vol. 6, No. 4, 2009 682 PERRET AND LUO

Table 2. Summary of Clinical Trials by Pain Etiology*

Study Drug Study Indication

Ziconotide, , pregabalin AIDS/HIV Pregabalin Anxiety, preoperative Ziconotide, , , pregabalin Cancer Amlodopine, gabapentin, ziconotide, , Chronic pain syndromes (pelvic, chronic prostatitis, abdominal, pregabalin, chronic pain secondary to trauma, chronic , chronic pancreatitis, vulvodynia) Pregabalin Complex regional pain syndrome Pregabalin Lamotrigine, pregabalin Facial pain (excluding postherpetic neuralgia) Gabapentin, pregabalin Hemorrhoidectomy Gabapentin, pregabalin Gabapentin, gabapentin XR, pregabalin, lamotrigine Peripheral neuropathy (, radiation, , diabetic, vascular, idiopathic) Gabapentin pain Gabapentin, gabapentin XR, lamotrigine, pregabalin Postherpetic neuralgia Gabapentin, pregabalin Postoperative pain (sternotomy, thoracotomy, hip surgery, replacement, cesarean section, tonsillectomy, keratectomy, inguinal hernia repair, spinal fusion, knee arthroscopy, scoliosis surgery, CABG, hysterectomy, cholecystectomy, mastectomy, axillary node dissection, bunionectomy) Pregabalin Post-traumatic pain Pregabalin Radiculopathy Pregabalin Spinal cord injury Pregabalin Spinal stenosis Pregabalin Stroke Gabapentin Stump pain Gabapentin Lamotrigine

*Source: clinicaltrials.gov. patients with chronic cancer or AIDS-associated pain and severity of adverse effects were observed in patients (“malignant pain”)82 or with nonmalignant pain83 were with low initial doses and gradual titration to achieve conducted. In the “malignant,” chronic cancer/AIDS-re- analgesia. In a randomized, double-blind, placebo-con- lated pain trial,82 ziconotide improved the mean visual trolled study,85 slower titration of intrathecal ziconotide analog scale of pain intensity (VASPI) scores by 53%. In to a lower maximum dose was associated with a signif- addition, 53% patients in the ziconotide group had pain icant improvement in pain relief and was better tolerated relief classified as moderate to complete compared with compared with faster titrations and higher maximum 18% patients in the placebo group. Five patients in the mean dose reported in two earlier placebo-controlled ziconotide group had achieved “complete” pain relief. In trials. It was suggested that administration of intrathecal the “nonmalignant” pain trial,83 240 patients were ran- ziconotide at a low starting (maximum) dose of 0.5 ␮g/ domized to ziconotide versus a placebo. Ziconotide re- day and a limitation of dose escalations to no more than duced the mean VASPI scores by 31% (compared with 0.5 ␮g/day may limit adverse effects.86 6% for the placebo group). In addition, more than 43% of A recent case report highlighted the use of ziconotide patients in the ziconotide group had moderate-to-com- in a patient with recalcitrant pain post-spinal cord injury, plete analgesia, compared with 17% in the placebo with both at-level and below-the-neurological-level of group. neuropathic pain syndromes. When intrathecal hydro- Several additional studies published in 2006 assessed morphone was used as a treatment modality, the patient’s the ideal effective dose and tolerability of ziconotide. In at-level, but not the below-level, pain was reduced. Con- three well-designed trials with up to 21-day durations,84 versely, when intrathecal ziconotide was administered, titration of ziconotide resulted in significant improve- analgesia was positive for the below-level, but only min- ment in the VASPI scores for chronic malignant or non- imally for the at-level, pain. When combination therapy malignant pain compared with a placebo. The analgesic with intrathecal and ziconotide was effects of ziconotide remained for up to 12 months in used, analgesia was sufficient for both pain components. long-term, open-label trials. In addition, lower incidence This study implies that central pain due to spinal cord

Neurotherapeutics, Vol. 6, No. 4, 2009 CALCIUM CHANNEL DRUGS IN TREATING CHRONIC PAIN 683

injury may be an indication for intrathecal ziconotide, Nevertheless, searching for safer analgesic peptide an- particularly in combination with intrathecal opioids.87 tagonists against the N-type VGCC continues. It has The usage of this peptide drug is limited by the route been reported that ␻-conotonix-GVIA, a 27- of administration and undesirable side effects, including peptide isolated from the cone snail, Conus geographus, sedation, dizziness, nausea, emesis, somnolence, head- also exhibits analgesic properties in animal models.52 In ache, confusion, memory impairment, slurred speech, contrast to ziconotide, ␻-conotonix-GVIA is an irrevers- nystagmus, double or blurry vision, urinary retention, ible inhibitor of N-type VGCC,96 which precludes its , elevated creatine kinase levels, and gait clinical application due to potential severe side effects. abnormality.78,88–90 It is believed that the serious side However, recent studies have indicated that binding of effects of ziconotide are derived from complete blockade this toxin peptide to the N-type VGCC can also modulate of the N-type VGCC that would affect the normal bio- (inhibit) N-type VGCC gating properties so that approx- physical functions of widely distributed N-type VGCC.91 imately 50%, instead of a complete blockade, of the However, it is argued that more convincing data are calcium influx during an is inhibited,91 in needed to make definitive conclusions because similar ad- addition to the primary pore blocking mechanism. This verse effects are not detectable in N-type VGCC knockout would provide a means to normalize elevated (activated) mice, which have no detectable evidence of developmental N-type VGCC activities in a disease condition, such as compensation from other types of VGCC.89 It is possible chronic pain, but preserve the biophysical functions of that some of these adverse effects may be secondary to N-type VGCC in maintaining normal physiological func- effects at other receptors besides the N-type VGCC. tions, and thus reduce adverse side effects. High- Most adverse events occur during the first week of treat- throughput screening using a scintillation proximity as- ment. After 6 months of treatment, the incidence of say has been used to search for small molecules that can ␻ adverse events is noted to be 0 to 35% of that reported in displace -conotonix-GVIA binding to the N-type VGCC the first month of treatment.84 Potential contamination of in an attempt to find N-type VGCC modulators with high affinity to the ␻-conotonix-GVIA binding sites on the intrathecal pump devices used to administer ziconotide 97 (or any other intrathecal agent) may increase the risk of N-type VGCC. ␻ meningitis.77 The neurological side effects of ziconotide Another 27-amino acid peptide -conotonix-CVID, also Conus catus dictate that the drug should be used with caution and called AM336, from the cone snail, , is perhaps the most selective of all N-type VGCC peptide blockers careful dose titration. Particular symptoms may correlate with greater than 24 hours of analgesia observed in rats with the rate of infusion.90 Because and toler- from subnanomolar intrathecal doses.73,98,99 Intrathe- ance are not detectable with the analgesic effects of cal administration of this peptide drug into rat pain ziconotide,88 but are often observed with the use of opi- models results in inhibition of spinal release of pain- oid analgesics, intrathecal ziconotide could be an option inducing peptides and potent dose-dependent anti-no- for replacing intrathecal morphine for chronic severe ciception.73,98 In addition, AM336 showed a greater ratio pain relief. However, the combination of opioid with- of analgesic efficacy to behavioral than zi- drawal symptoms and the cognitive/psychiatric adverse conotide,73,98 probably due to its greater selectivity for effects of ziconotide could make the conversion chal- N-type in comparison with P/Q type VGCC.71 This pep- lenging. Successful treatment with ziconotide mono- tide drug was assessed in a phase I in on- therapy has been suggested to include and cology patients with severe pain100 and its efficacy was psychological supports to decrease possible adverse psy- 92 established in a small phase IIa clinical trial for patients chological and physiological complications. with severe cancer pain.40 Unfortunately, the side effects Overall, although pain relief by ziconotide is accom- were undesirable and dose limiting. The adverse effect panied with some adverse effects, use of this analgesic profile is surprising given the 106-fold binding selectivity has several benefits. Ziconotide is less mood-altering of this peptide for N-type in comparison with P/Q-type than morphine for comparable analgesia and has the VGCC,71 and it may be secondary to supraspinal effects. advantage of lacking addiction, opioid-induced hyperal- gesia, and other systematic effects commonly observed Small molecule N-type VGCC blockers with opioids.93 For these reasons, the Polyanalgesic Con- The potency of ziconotide in chronic pain relief, and sensus Conference of 2007 puts ziconotide in the first its association with an undesirable administration route line of intrathecal therapy management, along with mor- and an undesirable side-effect profile, has prompted a phine and hydromorphone, in its recommended algo- race in searching for small molecules that would be rithm for nociceptive, mixed, and neuropathic pain.94 As orally active and have an acceptable therapeutic window summarized in a recent review, the evidence for short- for chronic pain management. Using a high-throughput

term improvement of -related pain or neuro- fluorescence-based in vitro assay to compare IC50 values pathic pain with intrathecal ziconotide is also strong.95 of blocking both N-type and L-type VGCC in a human

Neurotherapeutics, Vol. 6, No. 4, 2009 684 PERRET AND LUO

neuroblastoma cell line for identifying specific N-type Peptide T-type VGCC blockers VGCC blockers from its corporate compound library, Low-voltage T-type VGCC are found in DRG primary scientists in Ionix Pharmaceuticals Ltd. have developed a afferent cell bodies and in free nerve endings. They series of new N-type VGCC blockers derived from struc- contribute to the initiation of the action potential in these tural modifications of “hit” structures.101 After structural locations by lowering the required threshold for activa- and activity relationship analysis, compounds with up to tion.108 By promoting burst activity and synaptic excita- 30-fold of N-type/L-type VGCC selectivity and up to 0.2 tion, enhanced T-type VGCC activity favors the devel- ␮ opment of pain.51,109 T-type VGCC density has been MinIC50 values are identified. Unfortunately, analge- sic efficacies in chronic pain models were not included in increased in rat neuropathic pain models of diabetic neu- 110,111 the study. Further investigations for the analgesic effi- ropathy and chronic constriction nerve injury. The cacy and toxicity in animal pain models may prove the facts that T-type VGCC knockout animals have hypo- usefulness of these compounds for further development sensitivity to pain and that intrathecal injection with T- as therapeutic agents for chronic pain management. type VGCC blockers or antisense oligonucleotides re- sults in reduced excitability of the primary afferents and Indirect inhibitors of N-type channels therefore reduced nociceptive responses all attest to their established role in the processing of pain, especially N-type VGCC can be modulated by morphine, an im- 51,112–118 portant analgesic. Binding of morphine to ␮-opioid re- neuropathic pain. However, the exact mecha- nisms underlying the role of T-type VGCC in nocicep- ceptors activates G␤␥ which translocates to the mem- tion still remain elusive. Several regulatory mechanisms brane and binds to the N-type VGCC. A massive of T-type VGCC in pain processing have been proposed. G-protein-dependent inhibition of calcium currents re- These include redox modulation of T-type VGCC in rat sults in an inhibition of neurotransmitter release,2,45,102 peripheral nociceptors,119 and the selective enhancement and reduces the ability of the DRG sensory neurons to of T-type VGCC in nociceptive DRG neurons by reduc- propagate pain signals. This process is at least partially ing agents, such as L-cysteine, synthetic, and endoge- responsible for morphine’s analgesic effect, but can also nous chelators of zinc.119 The latter is supported by the contribute to tolerance development. Combination of zi- findings that peripherally injected reducing agents pro- ␮ conotide with -opioids shows synergistic analgesia duce thermal hyperalgesia in wild-type but not Cav3.2 74,98,103 when administered intrathecally. This combina- knockout mice, indicating that T-type VGCC may have a tion therapy may help to reduce opioid tolerance, a poor special role in peripheral sensitization.119 Currently, side-effect profile, and the potential for opioid-induced is the only T-type VGCC blocker approved hyperalgesia, all of which limit the long-term usefulness for human use.120 of ␮-opioid in pain medicine. Spinal noradrenaline can reduce VGCC-mediated transmission L-type VGCC blockers by presynaptic and postsynaptic inhibition, and by ␣1- Evidence from recent clinical studies shows that L- adrenoceptor-mediated activation of inhibitory interneu- type VGCC blockers, such as , are efficacious rons.40 These may explain the effectiveness of ␣2-adren- for neuropathic pain management, and leads to the sug- ergic agonists (such as ) and nonselective small gestion that this should be a third-line agent 121 molecule norepinephrine transporter (NET) inhibitors for neuropathic pain treatment. In one study, topira- (such as ) in pain relief. However, these med- mate has been shown to cause statistically significant 104 reduction in pain intensity compared to a placebo, as ications also have limiting side-effect profiles. Highly measured on a 100-mm visual analog scale, for up to 12 selective NET inhibitors, ␹-conopeptides, have been iso- weeks in diabetic peripheral neuropathy patients.122 lated from the cone snail, Conus marmoreus.105 It was However, three other randomized, double-blind, place- demonstrated that the ␹-conopeptide NET-binding site bo-controlled trials with more than 1,200 diabetic neu- partially overlaps the tricyclic NET-bind- 106 ropathy patients failed to show statistically significant ing site. Based on studies in rat neuropathic pain mod- reductions in pain intensity after 18 to 22 weeks of ␹ els, the -conopeptide compound MrIA (Xen2174) was treatment. In addition, these three studies also resulted in found to produce strong anti-allodynic effects without topiramate drop out rates ranging from 16% to 31% 107 significant side effects after intrathecal administration. compared with 8% for a placebo. The most commonly A phase I/IIa clinical trial involving intrathecal evalua- reported adverse events were , , nau- tion of Xen2174 in cancer patients with sea, somnolence, and diminished appetite.123 Another shows initial promising results.40 This line of compounds study evaluated the efficacy of topiramate for pain in may represent an important option in the search to iden- patients associated with chronic lumbar radiculopathy tify a potentially different therapeutic approach in target- and found modest pain relief in the topiramate-treated ing VGCC for chronic pain management. group, but with a 24% drop out rate.124 At least one

Neurotherapeutics, Vol. 6, No. 4, 2009 CALCIUM CHANNEL DRUGS IN TREATING CHRONIC PAIN 685

␣ ␦ recent case report has suggested that topiramate may be cleotides against the Cav 2 1 subunit can block nerve ␣ ␦ a useful pharmacologic modality for pain relief in pa- injury-induced Cav 2 1 upregulation and prevent the on- tients with postherpetic neuralgia who have failed other set of allodynia152 and reserve established allodynia.136 agents.125 It has been reported that topiramate has other Both gabapentin and pregabalin are structural deriva- pharmacologic actions including blockage of voltage- tives of the inhibitory neurotransmitter GABA although gated sodium channels in a dose-dependent manner,125 they do not bind to GABAA, GABAB, or potentiation of GABA inhibition, and AMPA receptor receptors, or alter GABA regulation in animal blockade,122–124,126 which may contribute to both its an- preparations.153 Binding of gabapentin and pregabalin to ␣ ␦ algesic potential and its adverse side effects. the Cav 2 1 subunit of VGCC results in a reduction in the calcium-dependent release of multiple neurotransmit- Other types of VGCC blockers ters,121,154 leading to efficacy and tolerability for neuro- Contribution of other types of VGCC to pain process- pathic pain management.121 ing have been suggested in some preclinical studies. Missense mutations in the Cav2.1 P/Q-type calcium Gabapentin channels lead to familial hemiplegic , an inher- Gabapentin is approved by the FDA for postherpetic 127–129 ited form of migraine with aura and hemiparesis. neuralgia, neuropathic pain, and partial seizures155–157 In addition, R-type VGCC have been implicated in the (Table 1). Several studies also suggest a clinical role for 130 processing of neuropathic pain and other pain states. restless leg syndrome,158 general anxiety,159 and general However, few clinical researches evaluating the effica- neuropathic pain.141 In common practice, gabapentin is cies of specific blockers to these VGCC in pain relief are used as a first-line agent to treat neuropathic pain from available; thus, these are not covered in this review. central origin (such as stroke or spinal cord injury) or from peripheral origin (such as peripheral neuropathy or radiculopathy). Despite its role and ubiquity, several CALCIUM CHANNEL ␣2␦1 LIGANDS FOR controlled trials demonstrate a low responder rate of PAIN MANAGEMENT approximately 32%.155 ␣ ␦ The unique features of the Cav 2 subunit and a large Gabapentin has inherent pharmacokinetic limitations; ␣ ␦ body of recent findings have suggested that the Cav 2 1 the half-life of gabapentin is short, and therefore admin- subunit may play an important role in neuropathic pain istration must be frequent. In addition, gabapentin is development. Biochemical data have indicated a significant absorbed actively via absorption pumps located in the ␣ ␦ ␣ ␦ Cav 2 1, but not Cav 2 2, subunit upregulation in the spi- upper . Therefore, conventional sus- nal dorsal horn, and DRG after nerve injury23,131–135 that tained-release formulations, where drug release is pro- correlates with neuropathic pain development.131,132,136 longed and occurs throughout the gastrointestinal tract, In addition, blocking axonal transport of injury-induced would simply decrease the of the drug.160 ␣ ␦ DRG Cav 2 1 subunit to the central presynaptic termi- Therapeutic doses and responses vary among patients nals diminishes tactile allodynia in nerve injured ani- and prediction of the ultimate desired doses is exceed- ␣ ␦ mals, suggesting that elevated DRG Cav 2 1 subunit ingly difficult. Furthermore, most gabapentin absorption contributes to neuropathic allodynia, even though a is nonlinear (i.e., as the dose is increased, the bioavail- postsynaptic mechanism can not be completely ruled ability decreases).160–162 23,24,136 ␣ ␦ out. Interestingly, the Cav 2 1 subunit (and the The FDA has approved gabapentin at a dose of up to ␣ ␦ ␣ ␦ ␣ ␦ Cav 2 2, but not Cav 2 3 and Cav 2 4, subunits) is the 1,800 mg/day. This drug is frequently used off-label for binding site for gabapentin,6,7,137 which has anti-allo- the treatment of other neuropathic pain conditions, in- dynic/hyperalgesic properties in patients and animal cluding painful diabetic peripheral neuropathy and radic- models with unknown mechanisms.131,132,138–143 Be- ulopathy, at higher doses up to 3,600 mg/day.141,163,164 It ␣ ␦ cause injury-induced Cav 2 1 expression correlates with has been suggested that gabapentin may be effective for neuropathic pain development and maintenance, and var- the treatment of chronic pain of any etiology, including ious calcium channels are known to contribute to spinal musculoskeletal headache, cervical pain, neuropathic synaptic neurotransmission50,144–149 and DRG neuron pain, lumbar pain, or multiple pains, such as fibromyal- 66,149–151 ␣ ␦ 165,166 excitability, injury-induced Cav 2 1 subunit up- gia (Table 2). Although gabapentin is well toler- regulation may contribute to the initiation and mainte- ated in studies, a statistically significant incidence of nance of neuropathic pain by altering the properties sedation, lightheadedness, and dizziness is noted when and/or distribution of VGCC in the subpopulation of compared to control patients.166 DRG neurons and their central terminals, therefore mod- Combinational treatment of gabapentin with other pain ulating excitability and/or synaptic neuroplasticity in the medications for pain management has been tested in dorsal horn. This is supported by findings from preclin- numerous clinical trials. Combination treatment of gaba- ical studies indicating that intrathecal antisense oligonu- pentin and morphine in an animal model of neuropathic

Neurotherapeutics, Vol. 6, No. 4, 2009 686 PERRET AND LUO pain results in a significant increase in the inhibitory twice-daily group (28.8%) was comparable with that re- effect of morphine on the dorsal horn neural response ported in the immediate-release gabapentin studies to peripheral stimulation,167 providing preclinical evi- (32.2%)156 and in the pregabalin studies (150 mg/day, dences to support that combination therapies with gaba- three times a day [26%], or 300 mg/day three times a day pentin may have clinical benefits in chronic pain man- [28%]).172 The incidence of adverse events was not sta- agement. A crossover study involving 57 patients with tistically different between the placebo group and either neuropathic pain demonstrated improved clinical out- of the gabapentin ER groups (once- or twice-daily), and comes when patients received a combination of mor- it did not appear to be dose-dependent.173 phine and gabapentin versus monotherapy.168 A larger study with 338 patients demonstrated that co-administra- XP13512: tion of gabapentin and prolonged-release of A novel pro-drug of gabapentin, XP13512/GSK has a clinically meaningful effect in patients with painful 1838262 ((Ϯ)-1-([(␣-isobutanoyloxyethoxy)carbonyl]- (diabetic) neuropathy.169 aminomethyl)-1-cyclohexane acetic acid) was recently developed. This drug has enhanced absorption in the Gabapentin extended-release large intestine, allowing an extended-release formula- A gabapentin extended-release (ER) has been devel- tion.174 Four clinical studies (two immediate-release [IR] oped. Gabapentin ER was constructed using polymer- formulation studies and two extended-release [XR] for- based AcuForm technology (Depomed, Inc., Menlo mulation studies) have evaluated the bioavailability of Park, CA). When taken with a meal, the tablet is retained the drug. In the first IR study, XP13512 was adminis- in the stomach for up to 8 h and the drug is gradually tered to healthy adults. Five groups of 10 subjects were released over 10 h to the , its optimal site randomized to receive the pro-drug or placebo in a dou- of absorption.162,170 This prolonged release was designed ble-blind fashion with five ascending pro-drug/placebo to provide similar or improved systemic exposure as doses (200, 400, 800, 1,200, and 1,400 mg). Maximum compared with the immediate release formulation. gabapentin concentrations were noted at 2.5 h In an exploratory study, the daily exposure provided postadministration. Bioavailability was consistently high by less frequent gabapentin extended-release dosing (Ͼ67%) across all doses. In the second IR study, twice (once- and twice-daily) was found not to be statistically daily dosing (350 mg–2,100 mg) was given. Steady state different from that provided by gabapentin immediate- concentrations of XP13512 were found proportional to release, administered more frequently (three times a day the oral dose range. In addition, the bioavailability [TID]). Gabapentin ER once-daily dosing was found to of gabapentin from XP13512 was consistently high produce higher maximum plasma concentrations com- (Ͼ72%) across the dose range. Blood concentrations of pared with the TID gabapentin immediate-release regi- the intact pro-drug after oral dosing were low. In the first men and twice-daily gabapentin ER dosing was found to XR study, plasma exposure to gabapentin was higher result in less fluctuation in plasma concentrations.171 post-XR treatment than after an equimolar dose of oral A recent randomized, double-blinded, placebo-con- gabapentin. The time to maximum concentration (T- trolled study evaluated gastric-retentive gabapentin in max) of gabapentin was substantially greater in the sub- 158 individuals who had chronic pain from post-herpetic jects given XP13512 than in subjects given oral gabap- neuralgia for at least 3 months. Patients were given 1,800 entin. In another XR study, plasma gabapentin was mg gabapentin ER once daily in the afternoon or 600 mg proportional to the XP13512 dose over the range studied in the morning with 1,200 mg in the afternoon (twice- (300 mg–1200 mg); bioavailability was increased in the daily dosing) or placebo. Efficacy outcomes included presence of food. Sustained delivery was noted at all changes from baseline on the pain intensity numeric dose levels. Overall, both IR and XR formulations of rating scale (average daily pain) and average daily sleep XP13512 were well tolerated without serious adverse interference score. The authors found statistically signif- events. Minor adverse events reported included dizzi- icant reductions in pain scores at all time points and as ness, headache, and sedation. Minor event reporting was early as week 1 post-treatment in the gabapentin ER similar for oral gabapentin, except for increased dizzi- twice-daily group compared with the placebo group. But ness in the IR formulation of XP13512, perhaps due to there was no significant reduction in pain scores in the rapid absorption. The XR formulation, although with once-daily gabapentin ER group compared with the pla- higher peak gabapentin concentrations than oral gabap- cebo control, although a trend was noted. It was con- entin, actually had similar adverse events compared with cluded that although ideal, once-daily gabapentin ER oral gabapentin.174 dosing may result in plasma levels that fall below the Given its increased absorption and more predictable therapeutic range before the end of the 24-hour dosing gabapentin exposure, its reduced inter-patient variability, period. Overall, the proportion of responders with at least and its reduced-dosing frequency, XP13512 may become a 50% reduction in pain score in the gabapentin ER an important option in our toolbox of upcoming neuro-

Neurotherapeutics, Vol. 6, No. 4, 2009 CALCIUM CHANNEL DRUGS IN TREATING CHRONIC PAIN 687 pathic agents. This novel extended-release pro-drug was approved by the FDA in 2007 as the first drug for the would be ideal for patients requiring prolonged clinical treatment of fibromyalgia181 (Table 1). exposure, such as patients with restless leg syndrome, Pregabalin is well tolerated, has predictable absorption using a single daily dose. Use of the XR formulation may across the gastrointestinal tract, and has minimal drug- dramatically improve treatment compliance. drug interactions.182 Clinical trials have been conducted GlaxoSmithKline announced results from a phase II to examine the efficacy of pregabalin in other chronic clinical trial of XP13512 for painful diabetic neurop- pain conditions (Table 2). At least seven published pro- athy in adults in April 2009 (Phase II Results for spective, randomized clinical trials have documented its GSK1838262 [XP13512] Reported for Neuropathic Pain efficacy in postherpetic neuralgia and painful diabetic Associated with Diabetic Peripheral Neuropathy; Medi- peripheral neuropathy with improvements in pain inten- cal News Today; article date, 4/29/2009; http://www. sity scores, decreased sleep interference, and secondary 182 medicalnewstoday.com/articles/148026.php). There were outcome improvements. The effects of pregabalin on 421 patients who were enrolled in a 14-week, double- patients with fibromyalgia are a current focus. These 183 blind, placebo-controlled study and were randomized to were first published in 2005 in an 8-week multicenter receive either 1,200 mg/day, 2,400 mg/day, or 3,600 efficacy and safety clinical trial. Pain intensity, sleep, mg/day of XP13512 in divided doses twice-daily; 300 fatigue, and quality of life were measured outcomes. mg/day pregabalin as an active control in divided doses Pregabalin (at 450 mg/day) significantly reduced pain three times daily, or a placebo. The primary endpoint intensity ona0to10point scale as compared with a placebo (p Յ 0.001). Pregabalin (at doses of 300 mg/day was a change from baseline on the Pain Intensity-Nu- and 450 mg/day) was reported to statistically improve merical Rating Scale (PI-NRS). Both the pregabalin ac- sleep, fatigue, and global measures of change. At 450 tive control and XP13512 failed to show a statistically mg/day, pregabalin also improved other secondary out- significant benefit when compared with a placebo. These comes associated with health-related quality of life as results may be due to an unusually high placebo re- measured in the Short Form-36 (SF-36) Health Survey. sponse. Therefore, efficacy conclusions are difficult to Adverse events were mostly mild, and the most fre- draw. XP13512, at all doses, was well tolerated in the quently reported adverse events were dizziness and som- study. Dizziness and somnolence were the most common nolence, although weight gain and peripheral edema reported adverse events. were also reported.183 The efficacy of XP13512 was also recently tested in a Pregabalin was also studied in combination therapy for randomized, double-blind, placebo-controlled study in a pain management. It was used in a combinational treat- population of patients with moderate-to-severe primary 184 175 ment of fibromyalgia in an open-label, 12-week study restless leg syndrome. There were 222 patients who in 19 female patients already receiving therapy with were randomized to 1,200 mg/day of XP13512 once- (76 mg/day). At the dose range from 75 mg/ daily or a placebo, and 192 patients completed the study. day to 300 mg/day, depending on the patient’s tolerabil- At week 12, there was a greater improvement in inter- ity, pregabalin was found to statistically improve the national restless leg syndrome scores. Significant treat- physical component of the SF-12 Health Survey, as well ment effects were noted as early as 1 week, the earliest as the pain and tiredness in the awakening subscales of time point measured, in the treatment group. More pa- the fibromyalgia impact questionnaire. Six of 19 patients tients treated with XP13512 (76%) were responders withdrew from the study, including 3 of them due to side compared with placebo (39%) (p Ͻ 0.0001). The medi- effects.184 cation was generally well tolerated; mild-to-moderate A multicenter study involving more than 700 patients side effects, including somnolence and dizziness were evaluated the efficacy and safety of pregabalin in patients reported.175 with fibromyalgia.185 Patients were given 300, 450, or 600 mg/day in twice-daily dosing schedules for 13 Pregabalin weeks. All patients treated with pregabalin had statisti- Pregabalin, S-enantiomer of racemic 3-isobutyl GABA, cally improved pain scores and patient global impression is a second-generation and structurally of change scores as compared with placebo treatments. similar to gabapentin. The binding affinity of pregabalin Statistically significant improvements in secondary out- ␣ ␦ for the Cav 2 1 subunit, however, is 6 times greater than comes measuring sleep were also reported. Side effects that of gabapentin, which makes pregabalin more clini- were mild to moderate and the most frequently reported cally effective at lower doses.176 Pregabalin is approved side effects were dizziness, somnolence, headache, infec- in the United States and Europe for the treatment of tion, and weight gain. This efficacy and safety was similarly neuropathic pain associated with diabetic peripheral neu- documented in another recently published trial.186 A recent ropathy and postherpetic neuralgia, and epilepsy as an long-term multicenter double-blind, placebo-controlled ran- add-on agent177–180 (Table 1). In addition, pregabalin domized discontinuation trial (FREEDOM) in the fibro-

Neurotherapeutics, Vol. 6, No. 4, 2009 688 PERRET AND LUO myalgia population demonstrated the durability (mainte- 13. Gurnett CA, De Waard M, Campbell KP. Dual function of the ϩ nance of response with pregabalin treatment relative to voltage-dependent Ca2 channel alpha 2 delta subunit in current 187 stimulation and subunit interaction. Neuron 1996;16:431–440. placebo) of pregabalin for this indication. In this trial, 14. Wiser O, Trus M, Tobi D, Halevi S, Giladi E, Atlas D. The alpha individually-determined optimal pregabalin doses were 2/delta subunit of voltage sensitive Ca2ϩ channels is a single determined with the hypothesis that the therapeutic re- transmembrane extracellular protein which is involved in regu- lated secretion. Febs Letters 1996;379:15–20. sponse would persist longer. The trial included a 6-week, 15. Mikami A, Imoto K, Tanabe T, et al. Primary structure and open-label pregabalin-treatment phase followed by a 26- functional expression of the cardiac dihydropyridine-sensitive week, double-blinded treatment with pregabalin or a pla- calcium channel. Nature 1989;340:230–233. 16. Mori Y, Friedrich T, Kim MS, et al. Primary structure and func- cebo. Patients treated with pregabalin had significantly tional expression from complementary DNA of a brain calcium delayed time to loss of therapeutic response, defined as channel. Nature 1991;350:398–402. Ͻ30% reduction in pain from open-label baseline, versus 17. Hullin R, Singer-Lahat D, Freichel M, et al. Calcium channel beta subunit heterogeneity: functional expression of cloned cDNA patients receiving a placebo (p Ͻ 0.0001). Half of the from , aorta and brain. Embo J 1992;11:885–890. placebo group had loss of therapeutic response by day 18. Williams ME, Feldman DH, McCue AF, et al. Structure and 19, but half of the pregabalin group retained therapeutic functional expression of alpha 1, alpha 2, and beta subunits of a novel human neuronal calcium channel subtype. Neuron 1992;8: effect and had not lost response by the end of the trial. At 71–84. the end of the 26-week, double-blind treatment phase, 19. Brust PF, Simerson S, McCue AF, et al. Human neuronal voltage- 61% of the placebo patients met loss of therapeutic re- dependent calcium channels: studies on subunit structure and role in channel assembly. Neuropharmacology 1993;32:1089–1102. sponse criteria versus 32% of pregabalin-treated patients. 20. Kang MG, Felix R, Campbell KP. Long-term regulation of volt- One contemporary commentary proposes a fibromyalgia age-gated Ca(2ϩ) channels by gabapentin. FEBS Lett 2002;528: treatment strategy combining pregabalin with memantine 177–182. 188 21. Gurnett CA, Felix R, Campbell KP. Extracellular interaction of in the clinical trial. The latter is proposed as an agent the voltage-dependent Ca2ϩ channel alpha2delta and alpha1 sub- that may slow down the loss of cephalic gray matter units. J Biol Chem 1997;272:18508–18512. 22. Wolf M, Eberhart A, Glossmann H, Striessnig J, Grigorieff N. commonly observed as a comorbidity in chronic pain ϩ 189–194 Visualization of the domain structure of an L-type Ca2 channel states. using electron cryo-microscopy. J Mol Biol 2003;332:171–182. 23. Bauer CS, Simerson S, McCue AF, et al. The increased traffick- ing of the calcium channel subunit alpha2delta-1 to presynaptic Acknowledgements: Supported in part by the National In- terminals in neuropathic pain is inhibited by the alpha2delta stitutes of Health, grant no. DE019298 (Z. D. Luo). ligand pregabalin. J Neurosci 2009;29:4076–4088. 24. Hendrich J, Van Minh AT, Heblich F, et al. Pharmacological REFERENCES disruption of calcium channel trafficking by the alpha2delta li- gand gabapentin. Proc Natl Acad SciUSA2008;105:3628– 1. Mitka M. “Virtual textbook” on pain developed: effort seeks to 3633. remedy gap in medical education. JAMA 2003;290:2395. 25. Takahashi M, Seagar MJ, Jones JF, Reber BF, Catterall WA. 2. Catterall WA. Structure and regulation of voltage-gated Ca2ϩ Subunit structure of dihydropyridine-sensitive calcium channels channels. Annu Rev Cell Dev Biol 2000;16:521–555. from skeletal muscle. Proc Natl Acad SciUSA1987;84:5478– 3. Dolphin AC. Beta subunits of voltage-gated calcium channels. 5482. J Bioenerg Biomembr 2003;35:599–620. 26. Catterall WA. Structure and function of voltage-sensitive ion 4. Dolphin AC. Beta subunits of voltage-gated calcium channels. channels. Science 1988;242:50–61. J Bioenerg Biomembr 2003;35:599–620. 27. Catterall WA. Functional subunit structure of voltage-gated cal- 5. Klugbauer N, Lacinova L, Marais E, Hobom M, Hofmann F. cium channels. Science 1991;253:1499–1500. Molecular diversity of the calcium channel a2d subunit. J Neu- 28. Black JL 3rd, Lennon VA. Identification and cloning of putative rosci 1999;19:684–691. human neuronal voltage-gated calcium channel gamma-2 and 6. Qin N, Yagel S, Momplaisir ML, Codd EE, D’Andrea MR. gamma-3 subunits: neurologic implications. Mayo Clin Proc Molecular cloning and characterization of the human voltage- 1999;74:357–361. gated calcium channel alpha(2)delta-4 subunit. Mol Pharmacol 29. Kang MG, Chen CC, Felix R, et al. Biochemical and biophysical 2002;62:485–496. evidence for gamma 2 subunit association with neuronal voltage- 7. Marais E, Klugbauer N, Hofmann F. Calcium channel activated Ca2ϩ channels. J Biol Chem 2001;276:32917–32924. alpha2delta subunits-structure and gabapentin binding. Mol Phar- 30. Klugbauer N, Dai S, Specht V, et al. A family of gamma-like macol 2001;59:1243–1248. calcium channel subunits. FEBS Lett 2000;470:189–197. 8. Kim HL, Kim H, Lee P, King RG, Chin H. Rat brain expresses an 31. Letts VA, Felix R, Biddlecome GH, et al. The mouse stargazer alternatively spliced form of the dihydropyridine-sensitive L-type gene encodes a neuronal Ca2ϩ-channel gamma subunit. Nat calcium channel alpha 2 subunit. Proc Natl Acad SciUSA Genet 1998;19:340–347. 1992;89:3251–3255. 32. Moss FJ, Viard P, Davies A, et al. The novel product of a 9. Angelotti T, Hofmann F. Tissue-specific expression of splice five-exon stargazin-related gene abolishes Ca(V)2.2 calcium variants of the mouse voltage-gated calcium channel alpha2/delta channel expression. Embo J 2002;21:1514–1523. subunit. Febs Letters 1996;397:331–337. 33. Ertel EA, Campbell KP, Harpold MM, et al. Nomenclature of 10. Ellis SB, Williams ME, Ways NR, et al. Sequence and expression voltage-gated calcium channels. Neuron 2000;25:533–535. of mRNAs encoding the alpha 1 and alpha 2 subunits of a DHP- 34. Jan LY, Jan YN. Voltage-sensitive ion channels. Cell 1989;56: sensitive calcium channel. Science 1988;241:1661–1664. 13–25. 11. De Jongh KS, Warner C, Catterall WA. Subunits of purified 35. Spedding M, Lepagnol J. Pharmacology of sodium and calcium calcium channels. Alpha 2 and delta are encoded by the same channel modulation in neurons: implications for neuroprotection. gene. J Biol Chem 1990;265:14738–14741. Biochem Soc Trans 1995;23:633–636. 12. Brickley K, Campbell V, Berrow N, et al. Use of site-directed 36. Tsien RW, Ellinor PT, Horne WA. Molecular diversity of volt- antibodies to probe the topography of the alpha 2 subunit of age-dependent Ca2ϩ channels. Trends Pharmacol Sci 1991;12: voltage-gated Ca2ϩ channels. Febs Letters 1995;364:129–133. 349–354.

Neurotherapeutics, Vol. 6, No. 4, 2009 CALCIUM CHANNEL DRUGS IN TREATING CHRONIC PAIN 689

37. Finkbeiner S, Greenberg ME. Ca2ϩ channel-regulated neuronal tivity induced by electrical field stimulation from rat spinal gene expression. J Neurobiol 1998;37:171–189. afferents is mediated by -sensitive calcium channels. 38. Belardetti F, Zamponi GW. Linking calcium-channel isoforms to Neurosci Lett 1992;136:161–164. potential therapies. Curr Opin Investig Drugs 2008;9:707–715. 62. Evans AR, Nicol GD, Vasko MR. Differential regulation of 39. Davies JN, Zamponi GW. Old proteins, developing roles: the evoked peptide release by voltage-sensitive calcium channels in regulation of calcium channels by synaptic proteins. Channels rat sensory neurons. Brain Res 1996;712:265–273. (Austin) 2008;2:130–138. 63. Saegusa H, Kurihara T, Zong S, et al. Suppression of inflamma- 40. Zamponi GW, Lewis RJ, Todorovic SM, Arneric SP, Snutch TP. tory and neuropathic pain symptoms in mice lacking the N-type Role of voltage-gated calcium channels in ascending pain path- Ca2ϩ channel. Embo J 2001;20:2349–2356. ways. Brain Res Rev 2009;60:84–89. 64. Saegusa H, Matsuda Y, Tanabe T. Effects of ablation of N- and 41. Catterall WA, Dib-Hajj S, Meisler MH, Pietrobon D. Inherited R-type Ca(2ϩ) channels on pain transmission. Neurosci Res neuronal ion channelopathies: new windows on complex neuro- 2002;43:1–7. logical diseases. J Neurosci 2008;28:11768–11777. 65. Hatakeyama S, Wakamori M, Ino M, et al. Differential nocicep- 42. Yaksh TL. Calcium channels as therapeutic targets in neuropathic tive responses in mice lacking the alpha(1B) subunit of N-type pain. J Pain 2006;7:S13–30. Ca(2ϩ) channels. Neuroreport 2001;12:2423–2427. 43. McGivern JG, McDonough SI. Voltage-gated calcium channels 66. Bell TJ, Thaler C, Castiglioni AJ, Helton TD, Lipscombe D. as targets for the treatment of chronic pain. Curr Drug Targets Cell-specific alternative splicing increases calcium channel cur- CNS Neurol Disord 2004;3:457–478. rent density in the pain pathway. Neuron 2004;41:127–138. 44. Bourinet E, Zamponi GW. Voltage gated calcium channels as 67. Lin Y, McDonough SI, Lipscombe D. Alternative Splicing in the targets for analgesics. Curr Top Med Chem 2005;5:539–546. voltage-sensing region of N-type CaV2.2 channels modulates 45. Schroeder CI, Doering CJ, Zamponi GW, Lewis RJ. N-type cal- channel kinetics. J Neurophysiol 2004;92:2820–2830. cium channel blockers: novel therapeutics for the treatment of 68. Lin Z, Haus S, Edgerton J, Lipscombe D. Identification of func- pain. Med Chem 2006;2:535–543. tionally distinct isoforms of the N-type Ca2ϩ channel in rat 46. Kisilevsky AE, Zamponi GW. Presynaptic calcium channels: sympathetic ganglia and brain. Neuron 1997;18:153–166. structure, regulators, and blockers. Handb Exp Pharmacol 2008: 69. Miljanich GP. In: Venoms to drugs. Heron Island, Australia. 45–75. 2005. Available at: http://www.venomstodrugs.com/index.html? 47. Catterall WA, Few AP. Calcium channel regulation and presyn- pageϭ107301&pidϭ0. aptic plasticity. Neuron 2008;59:882–901. 70. Hillyard DR, Monje VD, Mintz IM, et al. A new Conus peptide 48. Fuchs A, Rigaud M, Sarantopoulos CD, Filip P, Hogan QH. ligand for mammalian presynaptic Ca2ϩ channels. Neuron 1992; Contribution of calcium channel subtypes to the intracellular 9:69–77. calcium signal in sensory neurons: the effect of injury. Anesthe- 71. Lewis RJ, Nielsen KJ, Craik DJ, et al. Novel omega- siology 2007;107:117–127. from Conus catus discriminate among neuronal calcium channel 49. Diaz A, Dickenson AH. Blockade of spinal N- and P-type, but not subtypes. J Biol Chem 2000;275:35335–35344. L-type, calcium channels inhibits the excitability of rat dorsal horn neurones produced by subcutaneous formalin inflammation. 72. Nadasdi L, Yamashiro D, Chung D, Tarczy-Hornoch K, Adri- Pain 1997;69:93–100. aenssens P, Ramachandran J. Structure-activity analysis of a Co- 50. Matthews EA, Dickenson AH. Effects of spinally delivered N- nus peptide blocker of N-type neuronal calcium channels. Bio- and P-type voltage-dependent calcium channel antagonists on chemistry 1995;34:8076–8081. dorsal horn neuronal responses in a rat model of neuropathy. Pain 73. Smith MT, Cabot PJ, Ross FB, Robertson AD, Lewis RJ. The 2001;92:235–246. novel N-type calcium , AM336, produces potent 51. Matthews EA, Dickenson AH. Effects of ethosuximide, a T-type dose-dependent antinociception after intrathecal dosing in rats Ca(2ϩ) channel blocker, on dorsal horn neuronal responses in and inhibits substance P release in rat spinal cord slices. Pain rats. Eur J Pharmacol 2001;415:141–149. 2002;96:119–127. 52. Vanegas H, Schaible H. Effects of antagonists to high-threshold 74. Wang YX, Gao D, Pettus M, Phillips C, Bowersox SS. Interac- calcium channels upon spinal mechanisms of pain, hyperalgesia tions of intrathecally administered ziconotide, a selective blocker and allodynia. Pain 2000;85:9–18. of neuronal N-type voltage-sensitive calcium channels, with mor- 53. Westenbroek RE, Hell JW, Warner C, Dubel SJ, Snutch TP, phine on nociception in rats. Pain 2000;84:271–281. Catterall WA. Biochemical properties and subcellular distribution 75. Wang YX, Pettus M, Gao D, Phillips C, Scott Bowersox S. of an N-type calcium channel alpha 1 subunit. Neuron 1992;9: Effects of intrathecal administration of ziconotide, a selective 1099–1115. neuronal N-type , on mechanical allo- 54. Kerr LM, Filloux F, Olivera BM, Jackson H, Wamsley JK. Autora- dynia and heat hyperalgesia in a rat model of postoperative pain. diographic localization of calcium channels with [125I]omega-cono- Pain 2000;84:151–158. toxin in rat brain. Eur J Pharmacol 1988;146:181–183. 76. Sluka KA. Blockade of N- and P/Q-type calcium channels re- 55. Gohil K, Bell JR, Ramachandran J, Miljanich GP. Neuroanatomi- duces the secondary heat hyperalgesia induced by acute inflam- cal distribution of receptors for a novel voltage-sensitive calcium- mation. J Pharmacol Exp Ther 1998;287:232–237. channel antagonist, SNX-230 (omega-conopeptide MVIIC). 77. Lynch SS, Cheng CM, Yee JL. Intrathecal ziconotide for refrac- Brain Res 1994;653:258–266. tory chronic pain. Ann Pharmacother 2006;40:1293–1300. 56. Pertovaara A. Noradrenergic pain modulation. Prog Neurobiol 78. Klotz U. Ziconotide—a novel neuron-specific calcium channel 2006;80:53–83. blocker for the intrathecal treatment of severe chronic pain—a 57. Bourinet E, Soong TW, Stea A, Snutch TP. Determinants of the short review. Int J Clin Pharmacol Ther 2006;44:478–483. G protein-dependent opioid modulation of neuronal calcium 79. Wermeling DP. Ziconotide, an intrathecally administered N-type channels. Proc Natl Acad SciUSA1996;93:1486–1491. calcium channel antagonist for the treatment of chronic pain. 58. Altier C, Zamponi GW. Targeting Ca2ϩ channels to treat pain: Pharmacotherapy 2005;25:1084–1094. T-type versus N-type. Trends Pharmacol Sci 2004;25:465–470. 80. Atanassoff PG, Hartmannsgruber MW, Thrasher J, et al. Zi- 59. Chaplan SR, Pogrel JW, Yaksh TL. Role of voltage-dependent conotide, a new N-type calcium channel blocker, administered calcium channel subtypes in experimental tactile allodynia. intrathecally for acute postoperative pain. Reg Anesth Pain Med J Pharmacol Exp Ther 1994;269:1117–1123. 2000;25:274–278. 60. Maggi CA, Tramontana M, Cecconi R, Santicioli P. Neurochem- 81. Mathur VS. Ziconotide: a new pharmacological class of drug for ical evidence for the involvement of N-type calcium channels in the management of pain. Sem Anesth Periop Med Pain 2000; transmitter secretion from peripheral endings of sensory nerves in 19:67–75. guinea pigs. Neurosci Lett 1990;114:203–206. 82. Staats PS, Yearwood T, Charapata SG, et al. Intrathecal zi- 61. Santicioli P, Del Bianco E, Tramontana M, Geppetti P, Maggi conotide in the treatment of refractory pain in patients with cancer CA. Release of calcitonin gene-related peptide like-immunoreac- or AIDS: a randomized controlled trial. JAMA 2004;291:63–70.

Neurotherapeutics, Vol. 6, No. 4, 2009 690 PERRET AND LUO

83. Wallace MS, Charapata SG, Fisher R, et al. Intrathecal Ziconotide 105. Sharpe IA, Gehrmann J, Loughnan ML, et al. Two new classes of in the treatment of chronic non-malignant pain: a randomized, conopeptides inhibit the alpha1-adrenoceptor and noradrenaline double-blind, placebo-controlled clinical trial. Neuromodulation transporter. Nat Neurosci 2001;4:902–907. 2006;9:75–86. 106. Paczkowski FA, Sharpe IA, Dutertre S, Lewis RJ. chi-Conotoxin 84. Lyseng-Williamson KA, Perry C. Ziconotide. CNS Drugs 2006; and interactions at the norepinephrine 20:331–341. transporter define a new transporter model. J Biol Chem 2007; 85. Rauck RL, Wallace MS, Leong MS, et al. A randomized, double- 282:17837–17844. blind, placebo-controlled study of intrathecal ziconotide in adults 107. Nelson MT, Joksovic PM, Perez-Reyes E, Todorovic SM. The with severe chronic pain. J Pain Symptom Manage 2006;31:393– endogenous redox agent L-cysteine induces T-type Ca2ϩ chan- 406. nel-dependent sensitization of a novel subpopulation of rat pe- 86. Fisher R, Hassenbusch S, Krames E, et al. A consensus statement ripheral nociceptors. J Neurosci 2005;25:8766–8775. regarding the present suggested titration for prialt (ziconotide). 108. Todorovic SM, Jevtovic-Todorovic V. The role of T-type calcium Neuromodulation 2005;8:153–154. channels in peripheral and central pain processing. CNS Neurol 87. Saulino M. Successful reduction of neuropathic pain associated Disord Drug Targets 2006;5:639–653. with spinal cord injury via of a combination of intrathecal hydro- 109. Sekizawa SI, French AS, Torkkeli PH. Low-voltage-activated morphone and ziconotide: a case report. Spinal Cord 2007;45: calcium current does not regulate the firing behavior in paired 749–752. mechanosensory neurons with different adaptation properties. 88. Staats PS, Yearwood T, Charapata SG, et al. Intrathecal zi- J Neurophysiol 2000;83:746–753. conotide in the treatment of refractory pain in patients with cancer 110. Jagodic MM, Pathirathna S, Nelson MT, et al. Cell-specific al- or AIDS: a randomized controlled trial. JAMA 2004;291:63–70. terations of T-type calcium current in painful 89. Snutch TP. Targeting chronic and neuropathic pain: the N-type enhance excitability of sensory neurons. J Neurosci 2007;27: calcium channel comes of age. NeuroRx 2005;2:662–670. 3305–3316. 90. Wermeling DP, Berger JR. Ziconotide infusion for severe chronic 111. Jagodic MM, Pathirathna S, Joksovic PM, et al. Upregulation of pain: case series of patients with neuropathic pain. Pharmacother- the T-type calcium current in small rat sensory neurons after apy 2006;26:395–402. chronic constrictive injury of the sciatic nerve. J Neurophysiol 91. Yarotskyy V, Elmslie KS. Omega-conotoxin GVIA alters gating 2008;99:3151–3156. charge movement of N-type (CaV2.2) calcium channels. J Neu- 112. Dogrul A, Gardell LR, Ossipov MH, Tulunay FC, Lai J, Porreca rophysiol 2009;101:332–340. F. Reversal of experimental neuropathic pain by T-type calcium 92. Thompson JC, Dunbar E, Laye RR. Treatment challenges and channel blockers. Pain 2003;105:159–168. complications with ziconotide monotherapy in established pump 113. Flatters SJ, Bennett GJ. Ethosuximide reverses paclitaxel- and vin- patients. Pain Physician 2006;9:147–152. cristine-induced painful peripheral neuropathy. Pain 2004;109: 93. Vitale V, Battelli D, Gasperoni E, Monachese N. Intrathecal 150–161. therapy with ziconotide: clinical experience and considerations 114. Todorovic SM, Meyenburg A, Jevtovic-Todorovic V. Redox on its use. Minerva Anestesiol 2008;74:727–733. modulation of peripheral T-type Ca2ϩ channels in vivo: alter- 94. Deer T, Krames ES, Hassenbusch SJ, et al. Polyanalgesic Con- ation of nerve injury-induced thermal hyperalgesia. Pain 2004; sensus Conference 2007: recommendations for the management 109:328–339. of pain by intrathecal (intraspinal) drug delivery: report of an 115. Todorovic SM, Meyenburg A, Jevtovic-Todorovic V. Mechanical interdisciplinary expert panel. Neuromodulation 2007;10:300– and thermal antinociception in rats following systemic adminis- 328. tration of , a T-type calcium channel blocker. Brain Res 95. Smith HS, Deer TR, Staats PS, Singh V, Sehgal N, Cordner H. 2002;951:336–340. Intrathecal drug delivery. Pain Physician 2008;11:S89–S104. 116. Choi S, Na HS, Kim J, et al. Attenuated pain responses in mice 96. Pin JP, Bockaert J. Omega-conotoxin GVIA and dihydropyri- lacking Ca(V)3.2 T-type channels. Genes Brain Behav 2007;6: dines discriminate two types of Ca2ϩ channels involved in 425–431. GABA release from striatal neurons in culture. Eur J Pharmacol 117. Bourinet E, Alloui A, Monteil A, et al. Silencing of the Cav3.2 1990;188:81–84. T-type calcium channel gene in sensory neurons demonstrates its 97. Zhang SP, Kauffman J, Yagel SK, Codd EE. High-throughput major role in nociception. Embo J 2005;24:315–324. screening for N-type calcium channel blockers using a scintilla- 118. Shin HS, Cheong EJ, Choi S, Lee J, Na HS. T-type Ca2ϩ tion proximity assay. J Biomol Screen 2006;11:672–677. channels as therapeutic targets in the . Curr Opin 98. Scott DA, Wright CE, Angus JA. Actions of intrathecal omega- Pharmacol 2008;8:33–41. conotoxins CVID, GVIA, MVIIA, and morphine in acute and 119. Todorovic SM, Jevtovic-Todorovic V, Meyenburg A, et al. Re- neuropathic pain in the rat. Eur J Pharmacol 2002;451:279–286. dox modulation of T-type calcium channels in rat peripheral 99. Malmberg AB, Yaksh TL. Effect of continuous intrathecal infu- nociceptors. Neuron 2001;31:75–85. sion of omega-conopeptides, N-type calcium-channel blockers, 120. Todorovic SM, Jevtovic-Todorovic V. Regulation of T-type cal- on behavior and antinociception in the formalin and hot-plate cium channels in the peripheral pain pathway. Channels (Austin) tests in rats. Pain 1995;60:83–90. 2007;1:238–245. 100. Cousins M, Goucke R, Cher L, Brooker C. A phase I clinical trial 121. Dworkin RH, O’Connor AB, Backonja M, et al. Pharmacologic of AM336, a novel N-type calcium channel blocker. IASP Press: management of neuropathic pain: evidence-based recommenda- 10th World Congress on Pain 2002;200:615–P249. tions. Pain 2007;132:237–251. 101. Knutsen LJ, Hobbs CJ, Earnshaw CG, et al. Synthesis and SAR 122. Raskin P, Donofrio PD, Rosenthal NR, et al. Topiramate vs of novel 2-arylthiazolidinones as selective analgesic N-type cal- placebo in painful diabetic neuropathy: analgesic and metabolic cium channel blockers. Bioorg Med Chem Lett 2007;17:662– effects. Neurology 2004;63:865–873. 667. 123. Thienel U, Neto W, Schwabe SK, Vijapurkar U. Topiramate in 102. Herlitze S, Garcia DE, Mackie K, Hille B, Scheuer T, Catterall painful diabetic polyneuropathy: findings from three double-blind WA. Modulation of Ca2ϩ channels by G-protein beta gamma placebo-controlled trials. Acta Neurol Scand 2004;110:221–231. subunits. Nature 1996;380:258–262. 124. Khoromi S, Patsalides A, Parada S, Salehi V, Meegan JM, Max 103. Pirec V, Laurito CE, Lu Y, Yeomans DC. The combined effects MB. Topiramate in chronic lumbar radicular pain. J Pain 2005; of N-type calcium channel blockers and morphine on A delta 6:829–836. versus C fiber mediated nociception. Anesth Analg 2001;92: 125. Fowler JA, Shen JY, Bettinger TL. Successful use of topiramate 239–243. in a patient with severe postherpetic neuralgia. Ann Pharmacother 104. Martin TJ, Eisenach JC. Pharmacology of opioid and nonopioid 2009;43:139–142. analgesics in chronic pain states. J Pharmacol Exp Ther 2001; 126. Chong MS, Libretto SE. The rationale and use of topiramate for 299:811–817. treating neuropathic pain. Clin J Pain 2003;19:59–68.

Neurotherapeutics, Vol. 6, No. 4, 2009 CALCIUM CHANNEL DRUGS IN TREATING CHRONIC PAIN 691

127. Ophoff RA, Terwindt GM, Vergouwe MN, et al. Familial hemi- 148. Reid CA, Bekkers JM, Clements JD. Presynaptic Ca2ϩ channels: plegic migraine and episodic ataxia type-2 are caused by muta- a functional patchwork. Trends Neurosci 2003;26:683–687. tions in the Ca2ϩ channel gene CACNL1A4. Cell 1996;87:543– 149. Talley EM, Cribbs LL, Lee JH, Daud A, Perez-Reyes E, Bayliss 552. DA. Differential distribution of three members of a gene family 128. Pietrobon D, Striessnig J. Neurobiology of migraine. Nat Rev encoding low voltage-activated (T-type) calcium channels. J Neu- Neurosci 2003;4:386–398. rosci 1999;19:1895–1911. 129. Pietrobon D. Familial hemiplegic migraine. Neurotherapeutics 150. Lambert RC, McKenna F, Maulet Y, et al. Low-voltage-activated 2007;4:274–284. Ca2ϩ currents are generated by members of the CavT subunit 130. Murakami M, Nakagawasai O, Suzuki T, et al. Antinociceptive family (alpha1G/H) in rat primary sensory neurons. J Neurosci effect of different types of calcium channel inhibitors and the 1998;18:8605–8613. distribution of various calcium channel alpha 1 subunits in the 151. McCallum JB, Kwok WM, Mynlieff M, Bosnjak ZJ, Hogan QH. dorsal horn of spinal cord in mice. Brain Res 2004;1024:122–129. Loss of T-type calcium current in sensory neurons of rats with 131. Luo ZD, Chaplan SR, Higuera ES, et al. Upregulation of dorsal neuropathic pain. Anesthesiology 2003;98:209–216. root ganglion (alpha)2(delta) calcium channel subunit and its 152. Boroujerdi A, Kim HK, Lyu YS, et al. Injury discharges regulate correlation with allodynia in spinal nerve-injured rats. J Neurosci calcium channel alpha-2-delta-1 subunit upregulation in the dor- 2001;21:1868–1875. sal horn that contributes to initiation of neuropathic pain. Pain 132. Luo ZD, Calcutt NA, Higuera ES, et al. Injury type-specific 2008. calcium channel alpha 2 delta-1 subunit up-regulation in rat neu- 153. Zareba G. New treatment options in the management of fibromy- ropathic pain models correlates with antiallodynic effects of gaba- algia: role of pregabalin. Neuropsychiatr Dis Treat 2008;4:1193– pentin. J Pharmacol Exp Ther 2002;303:1199–1205. 1201. 133. Newton RA, Bingham S, Case PC, Sanger GJ, Lawson SN. 154. Stahl SM. and the relief of chronic pain: pre- Dorsal root ganglion neurons show increased expression of the gabalin and gabapentin as alpha(2)delta ligands at voltage-gated calcium channel alpha2delta-1 subunit following partial sciatic calcium channels. J Clin Psychiatry 2004;65:596–597. nerve injury. Brain Res Mol Brain Res 2001;95:1–8. 155. Rowbotham M, Harden N, Stacey B, Bernstein P, Magnus-Miller 134. Wang H, Sun H, Della Penna K, et al. Chronic neuropathic pain L. Gabapentin for the treatment of postherpetic neuralgia: a ran- is accompanied by global changes in gene expression and shares domized controlled trial. JAMA 1998;280:1837–1842. pathobiology with neurodegenerative diseases. Neuroscience 156. Rice AS, Maton S. Gabapentin in postherpetic neuralgia: a ran- 2002;114:529–546. domised, double blind, placebo controlled study. Pain 2001;94: 135. Valder CR, Liu JJ, Song YH, Luo ZD. Coupling gene chip 215–224. analyses and rat genetic variances in identifying potential target 157. Anhut H, Ashman P, Feuerstein TJ, Sauermann W, Saunders M, genes that may contribute to neuropathic allodynia development. Schmidt B. Gabapentin (Neurontin) as add-on therapy in patients J Neurochem 2003;87:560–573. with partial seizures: a double-blind, placebo-controlled study. 136. Li CY, Song YH, Higuera ES, Luo ZD. Spinal dorsal horn The International Gabapentin Study Group. Epilepsia 1994;35: calcium channel alpha2delta-1 subunit upregulation contributes 795–801. to peripheral nerve injury-induced tactile allodynia. J Neurosci 158. Garcia-Borreguero D, Larrosa O, de la Llave Y, Verger K, Mas- 2004;24:8494–8499. ramon X, Hernandez G. Treatment of with 137. Gee NS, Brown JP, Dissanayake VU, Offord J, Thurlow R, gabapentin: a double-blind, cross-over study. Neurology 2002;59: Woodruff GN. The novel anticonvulsant drug, gabapentin (Neu- 1573–1579. rontin), binds to the alpha2delta subunit of a calcium channel. J Biol Chem 1996;271:5768–5776. 159. Pollack MH, Matthews J, Scott EL. Gabapentin as a potential 138. Hwang JH, Yaksh TL. Effect of subarachnoid gabapentin on treatment for anxiety disorders. Am J Psychiatry 1998;155:992– tactile-evoked allodynia in a surgically induced neuropathic pain 993. model in the rat. Regional Anesthesia 1997;22:249–256. 160. Bockbrader H. Clinical of gabapentin. Drugs 139. Abdi S, Lee DH, Chung JM. The anti-allodynic effects of ami- Today 1995;31:613–619. triptyline, gabapentin, and in a rat model of neuropathic 161. McLean MJ. Clinical pharmacokinetics of gabapentin. Neurology pain. Anesth Analg 1998;87:1360–1366. 1994;44:S17–22; discussion S31–12. 140. Field MJ, McCleary S, Hughes J, Singh L. Gabapentin and pre- 162. Stewart BH, Kugler AR, Thompson PR, Bockbrader HN. A sat- gabalin, but not morphine and , block both static and urable transport mechanism in the intestinal absorption of gaba- dynamic components of mechanical allodynia induced by strep- pentin is the underlying cause of the lack of proportionality tozocin in the rat. Pain 1999;80:391–398. between increasing dose and drug levels in plasma. Pharm Res 141. Backonja M, Beydoun A, Edwards KR, et al. Gabapentin for the 1993;10:276–281. symptomatic treatment of painful neuropathy in patients with 163. Nicholson B. Gabapentin use in neuropathic pain syndromes. diabetes mellitus: a randomized controlled trial. JAMA 1998;280: Acta Neurol Scand 2000;101:359–371. 1831–1836. 164. Hemstreet B, Lapointe M. Evidence for the use of gabapentin in 142. Rosner H, Rubin L, Kestenbaum A. Gabapentin adjunctive ther- the treatment of diabetic peripheral neuropathy. Clin Ther 2001; apy in neuropathic pain states. Clin J Pain 1996;12:56–58. 23:520–531. 143. Rosenberg JM, Harrell C, Ristic H, Werner RA, de Rosayro AM. 165. Todorov AA, Kolchev CB, Todorov AB. and gabap- The effect of gabapentin on neuropathic pain. Clin J Pain 1997; entin for the management of chronic pain. Clin J Pain 2005;21: 13:251–255. 358–361. 144. Nudler S, Piriz J, Urbano FJ, Rosato-Siri MD, Renteria ES, 166. Arnold LM, Goldenberg DL, Stanford SB, et al. Gabapentin in Uchitel OD. Ca2ϩ channels and synaptic transmission at the the treatment of fibromyalgia: a randomized, double-blind, pla- adult, neonatal, and P/Q-type deficient neuromuscular junction. cebo-controlled, multicenter trial. Arthritis Rheum 2007;56: Ann N Y Acad Sci 2003;998:11–17. 1336–1344. 145. Perrier JF, Alaburda A, Hounsgaard J. Spinal plasticity mediated 167. Matthews EA, Dickenson AH. A combination of gabapentin and by postsynaptic L-type Ca2ϩ channels. Brain Res Brain Res Rev morphine mediates enhanced inhibitory effects on dorsal horn 2002;40:223–229. neuronal responses in a rat model of neuropathy. Anesthesiology 146. Westenbroek RE, Hoskins L, Catterall WA. Localization of 2002;96:633–640. Ca2ϩ channel subtypes on rat spinal motor neurons, interneu- 168. Gilron I, Bailey JM, Tu D, Holden RR, Weaver DF, Houlden RL. rons, and nerve terminals. J Neurosci 1998;18:6319–6330. Morphine, gabapentin, or their combination for neuropathic pain. 147. Kato A, Ohkubo T, Kitamura K. Algogen-specific pain process- N Engl J Med 2005;352:1324–1334. ing in mouse spinal cord: differential involvement of voltage- 169. Hanna M, O’Brien C, Wilson MC. Prolonged-release oxycodone dependent Ca(2ϩ) channels in synaptic transmission. Br J Phar- enhances the effects of existing gabapentin therapy in painful macol 2002;135:1336–1342. diabetic neuropathy patients. Eur J Pain 2008;12:804–813.

Neurotherapeutics, Vol. 6, No. 4, 2009 692 PERRET AND LUO

170. Gu R, Gusler GM, Hou SYE, et al. Extended release and dose- double-blind, placebo-controlled trial. Arthritis Rheum proportionality of a novel gabapentin formulation. The 88th An- 2005;52:1264–1273. nual Meeting of the Endocrine Society; 2006:319 (abstract). 184. Calandre EP, Morillas-Arques P, Rodriguez-Lopez CM, Rico- 171. Gordi T, Hou E, Kasichayanula S, Berner B. Pharmacokinetics Villademoros F, Hidalgo J. Pregabalin augmentation of quetiap- of gabapentin after a single day and at steady state following ine therapy in the treatment of fibromyalgia: an open-label, pro- the administration of gastric-retentive- extended-release and spective trial. Pharmacopsychiatry 2007;40:68–71. immediate-release tablets: a randomized, open-label, multiple-dose, 185. Mease PJ, Russell IJ, Arnold LM, et al. A randomized, double- three-way crossover, exploratory study in healthy subjects. Clin Ther blind, placebo-controlled, phase III trial of pregabalin in the 2008;30:909–916. treatment of patients with fibromyalgia. J Rheumatol 2008;35: 172. Sabatowski R, Gálvez R, Cherry DA, et al. Pregabalin reduces 502–514. pain and improves sleep and mood disturbances in patients with 186. Arnold LM, Russell IJ, Diri EW, et al. A 14-week, randomized, post-herpetic neuralgia: results of a randomised, placebo-controlled double-blinded, placebo-controlled monotherapy trial of pregaba- clinical trial. Pain 2004;109:26–35. lin in patients with fibromyalgia. J Pain 2008;9:792–805. 173. Irving G, Jensen M, Cramer M, et al. Efficacy and tolerability of gastric-retentive gabapentin for the treatment of postherpetic neu- 187. Crofford LJ, Mease PJ, Simpson SL, et al. Fibromyalgia re- ralgia: results of a double-blind, randomized, placebo-controlled lapse evaluation and efficacy for durability of meaningful re- clinical trial. Clin J Pain 2009;25:185–192. lief (FREEDOM): a 6-month, double-blind, placebo-controlled 174. Cundy KC, Sastry S, Luo W, Zou J, Moors TL, Canafax DM. trial with pregabalin. Pain 2008;136:419–431. Clinical pharmacokinetics of XP13512, a novel transported pro- 188. Recla JM, Sarantopoulos CD. Combined use of pregabalin and drug of gabapentin. J Clin Pharmacol 2008;48:1378–1388. memantine in fibromyalgia syndrome treatment: a novel analge- 175. Kushida CA, Becker PM, Ellenbogen AL, Canafax DM, Barrett sic and neuroprotective strategy? Med Hypotheses 2009;73:177– RW. Randomized, double-blind, placebo-controlled study of 183. XP13512/GSK1838262 in patients with RLS. Neurology 2009; 189. De Lange FP, Knoop H, Bleijenberg G, Van der Meer JW, 72:439–446. Hagoort P, Toni I. Gray matter volume reduction in the chronic 176. Gajraj NM. Pregabalin: its pharmacology and use in pain man- fatigue syndrome. Neuroimage 2005;26:777–781. agement. Anesth Analg 2007;105:1805–1815. 190. Kuchinad A, Schweinhardt P, Seminowicz DA, Wood PB, Chizh 177. Dworkin RH, Kirkpatrick P. Pregabalin. Nat Rev Drug Discov BA, Bushnell MC. Accelerated brain gray matter loss in fibro- 2005;4:455–456. myalgia patients: premature aging of the brain? J Neurosci 2007; 178. Frampton JE, Foster RH. Pregabalin: in the treatment of posther- 27:4004–4007. petic neuralgia. Drugs 2005;65:111–120. 191. Okada T, Tanaka M, Kuratsune H, Watanabe Y, Sadato N. Mech- 179. Gray P. Pregabalin in the management of central neuropathic anisms underlying fatigue: a voxel-based morphometric study of pain. Expert Opin Pharmacother 2007;8:3035–3041. chronic fatigue syndrome. BMC Neurol 2004;4:14. 180. Zareba G. Pregabalin: a new agent for the treatment of neuro- 192. Schmidt-Wilcke T, Luerding R, Weigand T, et al. Striatal grey pathic pain. Drugs Today 2007;41:509–516. matter increase in patients suffering from fibromyalgia--a voxel- 181. Owen RT. Pregabalin: its efficacy, safety and tolerability profile based morphometry study. Pain 2007;132(suppl 1):S109–116. in fibromyalgia syndrome. Drugs Today (Barc) 2007;43:857– 193. Schmidt-Wilcke T, Leinisch E, Straube A, et al. Gray matter 863. decrease in patients with chronic tension type headache. Neurol- 182. Stacey BR, Swift JN. Pregabalin for neuropathic pain based on ogy 2005;65:1483–1486. recent clinical trials. Curr Pain Headache Rep 2006;10:179–184. 194. Villarreal G, Hamilton DA, Petropoulos H, et al. Reduced hip- 183. Crofford LJ, Rowbotham MC, Mease PJ, et al. Pregabalin for the pocampal volume and total white matter volume in posttraumatic treatment of fibromyalgia syndrome: results of a randomized, stress disorder. Biol Psychiatry 2002;52:119–125.

Neurotherapeutics, Vol. 6, No. 4, 2009