<<

Pharmacological Reports Copyright © 2013 2013, 65, 1611–1621 by Institute of ISSN 1734-1140 Polish Academy of Sciences

Review

Neuronalandimmunologicalbasisofaction ofinchronic–clinical andexperimentalstudies

JoannaMika,MagdalenaZychowska,WiolettaMakuch, EwelinaRojewska,BarbaraPrzewlocka

DepartmentofPainPharmacology,InstituteofPharmacology,Smêtna12,PL31-343Kraków,Poland

Correspondence: BarbaraPrzewlocka,e-mail:[email protected]

Abstract: The current knowledge of the pharmacological actions of the antidepressants (TCAs) has slowly evolved through their over 40-year history. represents one of the most important public health problems, and antidepressants are an essential part of the therapeutic strategy in addition to classical . This article reviews the available evidence on the efficacy and safety of antidepressants in chronic pain conditions; namely, , , , pain and especially . TCAs are traditionally the main type of depression used to treat chronic pain. Recently, new antidepressants were introduced into clinical use, with a significant reduction in side effects and equivalent efficacy on mood disorders. These new that are effective for chronic pain belong to the antidepressants (TeCAs) group (, ), the and noradrenaline inhibitors (SNRIs) group (, , ) and the atypical antidepressants group (, , , ). In this review, we present the available publications on TCAs (, , , , ), TeCAs (amoxapine, maprotiline), selective serotonin reuptake inhibitors (SSRIs) (, , ), SNRIs (duloxetine, venlafaxine, milnacipran) and atypical antidepressants (bupropion) for the treatment of neuropathic pain. We also review analgesics acting as both and also acting as aminergic reup- take inhibitors. Existing data are insufficient to conclude which of these new classes of antidepressants has the best clinical profile and will be the most effective in the treatment of neuropathic pain; in addition, a lower incidence of side effects should be considered. Increased experimental and translational research is a key for further improvement of the treatment of chronic pain with antidepres- sants. However, evidence from basic science is needed to improve our understanding of the mechanisms of action and their possible pharmacodynamicinteractions.

Keywords: antidepressants,chronicpain,neuropathy

Introduction by primary sensory neurons through the dorsal horn of the and then to many structures throughout the ascending pain pathway. In the spinal Chronic pain develops as a consequence of injury or cord, the descending fibres originating in the brain- affecting the somatosensory pathways in the stem suppress pain neurotransmission; this suppres- peripheral or (CNS). Pain sion functions as a homeostatic regulator. The neuro- stimulus that originates in the periphery is transported transmitters released by the descending fibres are se-

Pharmacological Reports, 2013, 65, 1611–1621 1611 rotonin and noradrenaline, and dysfunction of these shorter time than effect [20]. Amitrip- systems is likely to induce dysfunctional descending tyline, nortriptyline and desipramine have been estab- serotonin or noradrenaline antinociceptive pathways, lished as analgesics independent of their antidepres- which explains comorbid pain symptoms in some pa- sant effects [16]. However, other mechanisms have tients with depression [37]. Patients with chronic pain been suggested. For example, amitriptyline has been are prone to depression in addition to a continuous found to block N-methyl-D-aspartate (NMDA) recep- load of the -intensified pain. Because of the tors, to interact directly with receptors, to act shared nature of depression and pain, the presence of as a sodium and to inhibit the cellular both conditions in a patient may lead to similar thera- uptake of adenosine, which consequentially activates pies. The most universally used antidepressants for adenosine A1 receptors on sensory terminals. chronic pain are tricyclic antidepressants (TCAs); Several recently published papers have considered however, selective serotonin or noradrenaline reup- a neuroimmune influence on the pain-relieving me- take inhibitors and a few other antidepressants are chanisms of antidepressants. Accumulated evidence also effective against pain. People respond differently demonstrated that spinal glial cells and their immune to depression because each chronic pain factors play an important role in persistent pain devel- condition is unique. Antidepressants are commonly opment [34, 56]. Some proinflammatory cytokines, used to treat the following chronic pain conditions: ar- such as IL-6 and IL-1b derived from activated glia, thritis, central pain syndrome, fibromyalgia, low back are common mediators of pain [35, 56]. However, it is still unclear which type of glial cells plays the most pain, migraines, nerve damage from (diabetic significant role in chronic pain development and how neuropathy), and nerve damage from antidepressants affect these cells. Zhu et al. [61] re- (postherpetic ). Their effectiveness is the best ported that mirtazapine markedly reduced hippocam- documented for painful and herpe- pal cytokine production and glial activation, which neuralgia. On the other hand, some clinical data was blocked by both and an- suggest that in patients with depression but without tagonists. The new approaches to the treatment of pain symptoms after prolonged use of amitriptyline the chronic, especially neuropathic, pain are based on the can occur [52], and in contrast, we found more comprehensive knowledge of the interaction be- pronociceptive effect in naive animals after peripheral tween neuronal, glial and immune cells. Up to now, injection of doxepine and amitriptyline [in prepara- only a few studies on neuroimmune influence in the . tion] These observations mean that in the absence of pain-relieving mechanisms of antidepressants have pathological changes associated with depression or been carried out. The question has been posed as to chronic pain effects of antidepressant drugs may which of the antidepressants has the best clinical pro- change its character to pronociceptive. The explanation file and could be most effective in the treatment of of this phenomenon requires further research. chronic pain and especially neuropathic pain, which is The of antidepressant drugs has known to be related to changes in neuroimmune inter- been suggested to result from the inhibition of actions. monoamine reuptake in the CNS, which consequen- tially leads to increased activity of the antinociceptive descending pathways. The antidepressants have an analgesic effect that may be, at least partly, independent of their effect on Classificationofantidepressantsand depression. The dose at which it is necessary to admin- theiruseinthetreatmentofchronicpain ister them in order to achieve optimal analgesia is usu- (Tab.1) ally lower than that which is required for antidepres- sant therapy [18], which may suggest separation of Tricyclicandtetracyclicantidepressants analgesic and antidepressant effect. Another confirma- (TCAsandTeCAs) tion of this thesis may be the differences in analgesic effectiveness between different classes of antidepres- The TCAs were first discovered at the beginning of sants [33] as well as the fact that the delay in onset of the 1950s and consequently introduced later in the analgesic effects after administration appears after decade. They are named after their chemical structure,

1612 Pharmacological Reports, 2013, 65, 1611–1621 Antidepressantsinchronicpain Joanna Mika et al.

Tab. 1. Primary mechanism of antidepressants effective for chronic pain In addition, TCAs and TeCAs are traditionally the main type of depression medication used to treat Groups Drugs Mechanisms chronic pain. Some of the most common forms of chronic pain therapy are based on TCAs (amitrip- TCA Amitriptyline 5-HT>NEreuptake tyline, nortriptyline, desipramine, imipramine, doxe- Doxepin inhibition Imipramine pin, , and ) Desipramine NE5-HTreuptakeinhibition and on TeCAs (amoxapine and maprotiline). The Nortriptyline TCAs and/or TeCAs can influence analgesia by a TeCA Amoxapine NEand5-HTreuptake number of different mechanisms: the serotonergic inhibition system can be affected by interference of serotonin re- Maprotiline NE5-HTDAreuptake uptake and alteration of serotonin binding to recep- inhibitionblockingreceptors: tors; the noradrenergic system can be affected by in- H1;5-HT2, a1-NE,D2,mACh teraction with a2-adrenoreceptors; the SSRI Citalopram 5-HT>NEreuptake Fluoxetine inhibition system can be affected by modification of opioid re- Paroxetine ceptor densities in several brain structures; GABAB receptors can be affected by an increase of receptor function; glutamate receptors can be affected by bind- SNRI Duloxetine 5-HTNE>DAreuptake ing to NMDA and/or AMPA receptors; and adenosine Venlafaxine inhibition Milnacipran receptors can be affected by inhibition of adenosine MAOI – – uptake peripherally, especially by A1 receptor activa- tion. Other receptors can also be affected by mecha- Atypical Bupropion DANEreuptakeinhibition antidepressant nisms, such as inhibition of , choliner- Trazodone 5-HT2 receptorblockade 5-HTreuptakeinhibition gic, muscarinic and nicotinic receptors, blocking or activating voltage-dependent sodium, or po- Mirtazapine a2-NEand5-HT2 presynaptic ;5-HT2/3 receptor tassium channels, and by inhibition of blockade by decreased prostaglandin E2 and tumor necrosis fac-

Nefazodone 5-HT2 receptorblockade tor a production [9, 15, 21, 32]. In 2005, Sindrup et 5-HTreuptakeinhibition al. [48] reported that the TCAs and TeCAs have no ef- fect on reuptake, but they might have some indirect action by the adrenergic effect which contains three rings of atoms. The TeCAs, and desensitization of dopamine D2 receptors. The which contain four rings of atoms, are a closely re- classical TCAs differ in their effect on monoamine re- lated group of antidepressant compounds. These older uptake. Amitriptyline, imipramine and clomipramine antidepressants are effective but are usually not cause a balanced inhibition of serotonin and nor- a first-choice treatment for depression because of reuptake in vivo. The serotonin reuptake their numerous side effects, such as , se- inhibition is exerted by the compounds themselves, dation, dry mouth, difficulty urinating, weight gain whereas the noradrenaline reuptake inhibition comes and sexual side effects. It is now becoming apparent from their respective metabolites, nortriptyline, desi- that TCAs can have an analgesic effect when applied pramineanddesmethylclomipramine. topically and that this effect is produced by peripheral mechanisms rather than systemic uptake. In some cases, a low dose of a cyclic antidepressant may be ItiswelldocumentedthatTCAsrelievechronic added to another antidepressant, such as an selective pain: serotonin (SSRI), to increase the antidepressant effect [13]. These medications are not • Amitriptyline has been a first-line treatment for usually given to older adults or people who have low chronic pain for many years. Many studies suggest or certain heart problems. Some TCAs that amitriptyline should be used as part of the treat- also have properties, which may help regu- ment for chronic pain or fibromyalgia, however, only latesleep. somepatientsachievesatisfactorypainrelief[36].

Pharmacological Reports, 2013, 65, 1611–1621 1613 • Imipramine was shown to be effective in the man- SSRIsrelievechronicpain: agement of non-, especially pain due to diabetes[26;41]. • Citalopram: two small trials (20 and 15 patients) in- • Desipramine relieves pain from postherpetic neu- dicated its small but significant analgesic effect on ralgia,headacheanddiabetes[30]. diabetes [48], but no effect was observed for fibromy- • Doxepin relievedchronicpain[54]. algia[38]. • Nortriptyline was effective in diminishing chemo- • Fluoxetine: one large trial (46 patients) revealed no therapy-relatedpain[19]andorofacialpain[55]. apparent effect on diabetic nerve pain [30], but it did improvefibromyalgiapain[1]. • demonstrated efficacy and safety in ItisalsodocumentedthatTeCAsrelieve themanagementofchroniclowbackpain[31]. chronicpain: • Fluvoxamine showed a lack of unequivocal evi- denceforpainreliefofchronicpain. • Amoxapine, a second-generation antidepressant, • Paroxetine: two small trials (20 and 15 patients) in- wasusedincancerpainmanagement[39]. dicateditsweakanalgesiceffectondiabetes[48]. • Maprotiline, a second-generation antidepressant, • Sertraline showed a lack of unequivocal evidence relieved chronic pain, especially postherpetic neural- forthepainreliefofchronicpain. gia[57]. It seems that the older and the newer TCAs, which act as ‘balanced’ reuptake inhibitors, are more effica- cious in terms of providing chronic pain relief than Selectiveserotoninreuptakeinhibitors(SSRIs) SSRIs.

In the late 1980s, a new class of antidepressants with Serotoninandreuptake inhibitors(SNRIs) different chemical structures was introduced, the SSRIs, which are non-tricyclic drugs. They are char- SNRIs cause a balanced inhibition of serotonin and acterized by causing inhibition of serotonin reuptake, noradrenaline reuptake [3]. SNRIs are newer antide- but they do not block noradrenaline reuptake [3]. pressants that are used to treat several types of chronic SSRIs are safe and commonly used, and their effect pain. They are thought to have fewer unpleasant side on relieving depression is well established. Potential effects than TCAs, TeCAs and SSRIs, and they might side effects of SSRIs include decreased appetite and beeffectiveagainstchronicpain. , dry mouth, tremors, fatigue or drowsiness, in- creased and blood pressure and sexual prob- ItiswelldocumentedthatSNRIsrelieve lems. Nevertheless, they are thought to have fewer chronicpain: unpleasant side effects than the TCAs but are gener- ally not as effective at treating chronic pain; however, • Duloxetine might help relieve both chronic pain and they are used to treat certain types of headaches. The depression. Duloxetine is itself a potent balanced most common SSRIs used for chronic pain therapy inhibitor of serotonin and noradrenaline reuptake, are citalopram, escitalopram, fluoxetine, fluvoxa- with no significant effect on a range of postsynaptic mine, paroxetine and sertraline. Fluoxetine has been receptors or sodium channels [58]. Duloxetine dem- reported to block sodium channels, but the other onstrated efficacy and safety in the management of SSRIs are not known to block sodium channels [9]. chronic low back pain [31] but has some side effects The SSRIs’ pharmacological advantages result from (nausea, dry mouth and constipation). Current evi- the high selectivity for the serotonin receptor and the dence strongly suggests that treatment with duloxe- low or null affinity for other types of receptors, such tine may be beneficial in older adults with major de- as a-adrenergic, muscarinic, and hista- pressivedisordersandpain[10]. minic receptors [51]. Twenty trials available in the lit- • Venlafaxine can raise blood pressure; therefore, erature were analyzed by Smith et al. [49] to evaluate overdose can be dangerous or fatal. Venlafaxine at the efficacy of SSRIs in the treatment of chronic pain low doses has a very similar action to SSRIs, as the conditions;however,theresultswereconflicting. serotonin reuptake inhibition predominates; however,

1614 Pharmacological Reports, 2013, 65, 1611–1621 Antidepressantsinchronicpain Joanna Mika et al.

at high doses, noradrenaline reuptake inhibition is a few sexual side effects. Bupropion might also sup- prominent. Venlafaxine has no postsynaptic effects, press appetite, and it might help with smoking cessa- but it blocks sodium channels [24]. It was suggested tion. that venlafaxine might work for some people resistant • Trazodone is a serotonin antagonist and reuptake to other antidepressants. In 2009, treatment with ven- inhibitor, and it has anti- () and lafaxine was investigated in 186 patients with major sleep-inducing (hypnotic) effects. Trazodone has con- depressive disorders whose treatments had failed with siderably fewer prominent (dry mouth, an SSRI or TCA. Moreover, 12 patients receiving constipation and ) and sexual side effects, venlafaxine for one year demonstrated improvement but the clinical trials did not confer its beneficial ef- ofchronicpain[29]. fectsforchronicpain. • is a synthetic form of the major ac- • Mirtazapine has a unique pharmacological profile; tive metabolite of venlafaxine and causes similar ef- it increases 5-HT1A-mediated neurotransmission, has fects. a strong antagonism for a2 adrenergic and 5-HT2 and • Milnacipran has equal affinity for the serotonin and 5-HT3 serotoninergic receptors and a minimum effect noradrenaline reuptake sites, while duloxetine and on reuptake [8]. Mirtazapine can be used as an venlafaxine have a 10- and 30-fold selectivity, respec- optional treatment for headaches and nausea [28] and tively, for the serotonin reuptake site versus the nor- for resolving insomnia, anxiety and depressive symp- adrenaline reuptake site [50]. Milnacipran was exam- tomsincancerpainpatients[7]. ined in 125 patients with fibromyalgia and is now • Nefazodone antagonizes the action of 5-HT2 recep- consider as a possible second-line treatment for tors and inhibits noradrenaline and serotonin reup- fibromyalgia[17,54]. take. The 5-HT2-receptor antagonism increases 5-HT1A receptor binding [12]. Thus, the Monoamineoxidaseinhibitors(MAOIs) is believed to be due to a net effect of increased neu- rotransmission. Nefazodone has the double advantage MAOIs have no place in , and little of a weak affinity for a1- and b-adrenergic receptors evidence from experimental studies exists for their and no activity with histaminic, dopaminergic or mus- analgesic effect [32]. In depression treatment, MAOIs carinic cholinergic receptors, resulting in a signifi- are used as a last resort because of their numerous bothersome and potentially dangerous and serious side effects. The MAOIs that have no satisfactory ef- Tab.2. Antidepressantsusedinneuropathicpaintreatment fects on chronic pain, according to clinical data, are: , , , , Groups Drugs References , and tranylcy- promine. TCA Amitriptyline [36] Doxepin [54] Imipramine [41] Nortriptyline Atypicalantidepressants [19]

These medications are called “atypical” because they TeCA Amoxapine [39] Maprotiline do not fit into other categories, but they may also be [57] effective at treating chronic pain, including dopami- SSRI Citalopram [48] ne-noradrenaline reuptake inhibitors and serotonin re- Fluoxetine [1,30] Paroxetine ceptor modulators. Atypical antidepressants used for [48] chronicpaintreatmentinclude: • Bupropion, a second-generation non-tricyclic antide- Duloxetine [10,31] SNRI Venlafaxine [11] pressant that is a noradrenaline and dopamine reuptake Milnacipran inhibitor without the postsynaptic effects [3]. A study [17,54] of patients with various forms of chronic pain has MAOI – – shown that bupropion is effective [44]. Bupropion has several clinical contraindications, such as , eat- Atypical Bupropion [45] antidepressant ing disorder, activating effect for some patients and

Pharmacological Reports, 2013, 65, 1611–1621 1615 cantly lower incidence of side effects. However, - tients with both normal and depressed moods in zodone has been linked to dangerous problems. anumberofneuropathicpainconditions[27,30]. There are no published reports on nefazodone in pain The painkilling mechanism of these drugs under management, but experimental studies suggest its an- neuropathic pain conditions is still a matter of study. algesicproperties[40]. These studies have shown that doses necessary to at- tenuate pain are often lower than those used to treat depression, and the onset of activity is more rapid at these doses. Additionally, these studies have shown that analgesic efficacy is usually obtained in non- Usefulnessofantidepressantsfor depressed patients and does not correlate with im- attenuatingneuropathicpain(Tab.2) provement in mood in depressed patients, and the drugs are useful in acute and experimental pain. Ry- Antidepressantsinthetreatment der and Stannard [43] have published that almost 50% ofneuropathicpain of patients with pain have depression, but antidepres- sants are prescribed in the pain clinic for their specific Neuropathic pain is difficult to treat, however, several analgesic effects rather than their mood altering ef- treatment options with reasonable efficacy are avail- fects; however, the latter is also very important in able at the present time. Opioids and NSAIDs are chronic pain patients. Antidepressants might increase only partially effective, therefore, there is a need to in the spinal cord that reduce pain use adjuvant analgesics. Neuropathic pain affects signals, but it takes time to experience pain relief 6–8% of the general population, and its impact on (sometimes up to one or more weeks). Jain and Jain quality of life, mood and sleep exceeds the burden of [22] have stressed that neuropathic pain and depres- its causative pathology [48]. The characteristic syn- sion have a shared neurobiology and appear to have dromes of neuropathic pain ( and hyperalge- a shared neuroanatomy (in the brain and spinal cord) sia) develop as a result of damage to in conse- and neurochemistry (noradrenaline and serotonin), quence of tumors, , diabetes, herpes with similar hypothalamic-pituitary-adrenal axis, zoster, AIDS and many neurological , such as and inflammatory cyto- , radiculopathy, spinal cord in- kinedisturbances[22]. jury and [42, 48]. Antidepressants were used in TCAs remain one of the first-line therapies for neu- painful diabetic neuropathy based on empirical obser- ropathic pain, such as (amitrip- vations in 1977 by Davis and co-workers, but their tyline, nortriptyline, and desipramine) and painful potential analgesic properties were noted shortly after diabetic neuropathy (triptyline, desipramine, clomi- their use. Clinical studies have shown that TCAs tend pramine, imipramine, and nortriptyline) [32]. Com- to work better than an () parative studies show that TCAs (amitriptyline, doxe- and opioid ( and ) drugs, whereas pin, imipramine) with balanced noradrenergic and venlafaxine from the SNRI group appears to be serotonergic effects are more effective than TeCAs equally effective, and SSRIs appear to have lower ef- (maprotiline) with anticholinergic properties and se- ficacy [11, 46, 48]. Ryder and Stannard [43] have lective serotonin reuptake inhibitors from the SSRI shown that, from 100 patients with prescribed antide- group (citalopram, fluoxetine, paroxetine) [43, 47]. pressants for neuropathic pain, 30 patients will obtain TCAs might relieve neuropathic pain by their unique 50% pain relief, 30 patients will have minor adverse ability to block monoamine uptake in the CNS, spe- reactions and 4 patients will stop treatment because of cifically serotonin and/or noradrenaline, in addition to major adverse effects. Beneficial effects on sleep usu- other neurotransmitters. They might alter nociceptive ally appear within a few days of treatment, whereas processing by prolonging synaptic activity of these the improvement in pain will take a week or longer monoamines, thereby enhancing descending inhibi- [43]. Table 1 shows the antidepressants suitable for tory action in the spinal cord and having monoaminer- use in the treatment of neuropathic pain. The exact gic effects elsewhere in the CNS [23]. To a varying mechanism of the analgesic action of antidepressants degree, the antidepressants also block a number of is not yet clear; however, it is well established that re- other receptor types involved in pain processing, in- peatedly administered antidepressants work in pa- cluding a-adrenergic, H1-histaminergic and N-

1616 Pharmacological Reports, 2013, 65, 1611–1621 Antidepressantsinchronicpain Joanna Mika et al.

methyl-D-aspartate (NMDA) receptors. They also eral neuropathies, such as postherpetic neuralgia, in- block the effects on calcium and sodium channels and tercostal neuralgia, peripheral diabetic neuropathy are weak stimulators of µ-opioid receptors. A recent and . The use of venlafaxine is rec- study suggests that antidepressant treatment appears ommended as a second choice after other therapies to positively impact immune/cytokine deregulation (e.g., NSAIDs and other antidepressants) because of [22]. Research data indicate that antidepressants can either their inefficacy or the intolerable presence of reduce levels of inflammatory cytokines, such as side effects. In the treatment of post-herpetic neural- tumor necrosis factor a and interleukin-6 [5]. gia, venlafaxine was effective, especially when asso- The hyperexcitability of NMDA receptors is one of ciated with GABAergic drugs (e.g., gabapentin) due the factors in the genesis of neuropathic pain. Keta- toapossiblesynergybetweenthetwodrugs[29]. mine (an NMDA ) is useful for The MAOI antidepressants are not used to treat neu- treating neuropathic pain. Some studies show that ropathic pain in the clinic [32]. From the atypical anti- TCAs are NMDA receptor-like antagonists, although group, bupropion provided substantial neu- this effect might not be fully present when the drug is ropathic pain relief in a study by Semenchuck et al. used in therapeutic concentrations. Neuronal sponta- [44]. The results from this study suggest the possibility neous activity, caused by sodium and calcium chan- that a dopaminergic effect could enhance the peripheral nels, leads to neuropathic pain; therefore, the unspe- neuropathic pain-relieving effect obtained with seroto- cific relieves cen- nergic and noradrenergic mechanisms [32]. tral pain [2], and the anticonvulsant drug gabapentin clearly relieves neuropathic pain. The TCAs also Combinationofantidepressantswithother potently inhibit sodium and L-type calcium channels treatments and therefore act as sodium and calcium channel The effect of combining antidepressants with other blockers, respectively, in neuropathic pain [14]. In drugs in neuropathic pain needs additional research. summary, the effectiveness of TCAs in neuropathic Combining venlafaxine with gabapentin, if the latter pain is most likely multimodal, with contribution of provided insufficient pain relief in painful diabetic monoamine reuptake inhibition and blockade of neuropathy, resulted in a significant additional effect NMDA receptors, sodium channels and calcium chan- [46]. Thus, TCAs and venlafaxine could be combined nels. with gabapentin and with opioids, with the exception Several clinical studies have shown that the SSRI- of tramadol because it interferes with the monoamin- like drugs citalopram, fluoxetine and paroxetine can be ergic system and has an opioid effect, therefore, com- effective in painful diabetic neuropathy [1, 30, 48]; bination of different antidepressant classes should be however, the drugs from this group are generally less avoided with this drug. Clinical studies on the use of effective for neuropathy than TCAs, TeCAs and SNRI. nefazodone in pain treatment are not yet available. In A number of positive trials have been published on an experimental study on , nefazodone was shown the SNRIs group, including the use of duloxetine, to have analgesic properties, especially when associ- venlafaxine, and milnacipran for neuropathic pain ated with . In fact, nefazodone seems to be [54]. In the model of neuropathic pain, venlafaxine able to increase the analgesic effect on the µ-opioid and duloxetine both have a significant antinociceptive receptor, without effecting lethality or gastroenteric effect [32]. In addition, our results [in preparation] motility [29]. It is also very important to remember suggest that not only venlafaxine and duloxetine but that antidepressants administered together with other also milnacipran have similar antinociceptive effects drugs can participate in pharmacodynamic interac- in both and mouse neuropathic pain models. Ven- tions. Some drugs can add their own contribution to lafaxine and duloxetine should be second-choice antidepressant-related risks of treatments in painful diabetic neuropathy [54]. In the (e.g., antimigraine agents, , or elderly and in patients with cardiovascular risk fac- ecstasy and antibiotics, such as ), seizure in- tors, it is suggested that SNRIs should be preferred duction (e.g., , , , over TCAs. Venlafaxine is the most investigated of fluoroquinolone antibiotics, such as , and these new drugs in neuropathic pain management. antiasthmatics), ventricular arrhythmias (e.g., antiar- Many cases reports have shown that venlafaxine is ef- rhythmic agents, antiepileptics, antipsychotics, me- fective for relieving pain in different types of periph- thadone, and macrolide antibiotics) or gastrointestinal

Pharmacological Reports, 2013, 65, 1611–1621 1617 bleeding (e.g., anti-inflammatory agents and antico- tion, has an improved tolerability profile in agulants). In conclusion, antidepressants in the TCA the relevant animal models, particularly with regard and SNRI groups are fairly effective in peripheral to gastrointestinal and central side effects, despite its neuropathicpain,whereasSSRIshavealowefficacy. highpotency[53].

Analgesicsactingbybothopioidreceptors Experimentsonanimalmodelofneuropathic andaminergicreuptakeinhibitors pain

Neuropathic pain is generally considered to be diffi- Experimental findings demonstrated that the antide- cult to treat because of low affectivity of µ-opioid re- pressants used have an antinociceptive effect on rat ceptor agonists. Activation of the opioid receptors and and mice neuropathic pain conditions [60–63]. The norepinephrine and/or serotonin reuptake inhibition is analgesic effects of TCAs, such as amitriptyline, have currently suggested as an analgesic treatment in neu- been examined using a variety of animal models of ropathicpain. pain, and they have been shown to have antinocicep- Tramadol is an atypical, racemic opioid that com- tive and antiallodynic effects when administered sys- bines weak µ- activation with inhibi- temically, spinally, supraspinally and locally [6]. We, tion of norepinephrine and serotonin reuptake (the as well as others, have shown that subcutaneously, mechanisms common to antidepressants). Tramadol intraperitoneal and intrathecal administration of mil- also acts as an NMDA receptor antagonist, 5-HT2C re- nacipran and venlafaxine (SNRIs), fluoxetine (an ceptor antagonist, (a7)5 nicotinic re- SSRI), and amitriptyline and doxepin (TCAs) reduce ceptor antagonist, TRPV1 receptor agonist, and an M1 pain syndromes in a rat model of neuropathic pain and M3 muscarinic antagonist. [43, 48, 59]. Milnacipran is thus similar to the TCAs, This combination of complementary mechanisms of which also inhibit the reuptake of both serotonin and action results in potent analgesic activity in neuro- noradrenaline. A major difference, however, between pathic pain. The contribution of non-opioid activity is the actions of milnacipran and the TCAs is that the demonstrated by the fact that the analgesic effect of former compound is devoid of interactions at neuro- tramadol is not fully antagonized by the µ-opioid re- transmitter receptors and is thus devoid of the trouble- ceptor antagonist – . Tramadol is metabo- some adverse effects that these interactions cause. In lized to O-desmethyltramadol, a significantly more rats, milnacipran has been reported to have antiallo- potent µ-opioid agonist. Tramadol and its major me- dynic and antihyperalgesic effects on neuropathic tabolite are distinguished from other more potent pain. These effects are similar to duloxetine, which opioid agonists by their relative selectivity for has been shown to significantly attenuate inflamma- µ-opioidreceptors. tory and diabetic pain by intraperitoneal, intrathecal, Tapentadol (3-[(1R, 2R)-3-(dimethylamino)-1-ethyl- and intracisternal administration. Korzeniewska- 2-methylpropyl]phenol) is a centrally acting analgesic Rybicka and P³aŸnik [25] examined the influence of of a new substance class for the treatment of severe imipramine, amitriptyline, citalopram and maprotiline nociceptive and neuropathic pain. Tapentadol com- on the nociceptive response. Their results indicate bines µ-opioid receptor agonism and selective norepi- that there is a disparate sensitivity to antidepressant nephrine reuptake inhibition into one molecule. treatment of differently evoked behavioral reactions Because of its combined mechanisms of action, tap- to the nociceptive stimuli. They also showed that the entadol offers a broad therapeutic spectrum for noci- most potent effects of administered antidepressants ceptive and neuropathic pain. In different animal are in the model of visceral pain, and there is no rela- models, its high efficacy was shown in acute nocicep- tionship between the analgesic and antidepressant- tive, acute and chronic inflammatory as well as like effects of the antidepressants compounds that chronic neuropathic pain. Using several preclinical theyexamined. approaches, it was shown that the noradrenergic com- In the rat model of neuropathic pain, animals de- ponent of tapentadol interacts with the opioid compo- veloped and allodynia in parallel with nent and that both synergistically contribute to the microglial activation [35, 56]. Our results of western high analgesic effect of the substance. In comparison blot analysis [in preparation] are interesting because to known drugs with only one of the two modes of ac- antidepressants, such as milnacipran, venlafaxine,

1618 Pharmacological Reports, 2013, 65, 1611–1621 Antidepressantsinchronicpain Joanna Mika et al.

fluoxetine, amitriptyline and doxepin, reduce similar 3. BaldessariniRJ:Drugsforthetreatmentofpsychiatric pain syndromes; however, these drugs demonstrate disorders.In:Goodman&Gilman’sThePharmacologi- calBasisofTherapeutics,10th edn.,Eds.HardmanJG, a different influence on microglial activation under LimbirdLE,McGrawHill,NewYork,USA,2001, neuropathic pain conditions. In the lumbar part of the 447–483. spinal cord, we observed that microglial changes are 4. BansalD,BhansaliA,HotaD,ChakrabartiA,DuttaP: enhanced in neuropathic pain after single administra- Amitriptylinevs.inpainfuldiabeticneuropa- tion of amitriptyline, doxepin and milnacipran, and thy:arandomizeddoubleblindclinicaltrial.Diabet Med,2009,26,1019–1026. the opposite effect was observed for venlafaxine and 5. BasterziAD,AydemirC.KisaC:IL-6levelsdecrease fluoxetine. Simultaneously, we did not observe withSSRItreatmentinpatientswithmajordepression. changes in astroglial cell activation. Therefore, we HumPsychopharmacol,2005,20,473–476. verified the influence of chronic administration of 6. BenbouzidM,Gavériaux-RuffC,YalcinI,Waltisperger (microglial inhibitor) on the effective- E,TessierLH,MullerA,KiefferBL etal.:Delta-opioid ness of selected antidepressants. The results showed receptorsarecriticalfortricyclicantidepressanttreat- mentofneuropathicallodynia.BiolPsychiatry,2008,63, that a single administration of fluoxetine, milnacipran 633–636. and amitriptyline after chronic administration of 7. CankurtaranES,OzalpE,SoygurH,AkbiyikDI,Turhan minocycline increased its effectiveness, measured by L,AlkisN:Mirtazapineimprovessleepandlowersanxi- von Frey and cold plate tests. This result might sug- etyanddepressionincancerpatients:superiorityover gest involvement of microglial cells in the effective- imipramine.SupportCareCancer,2008,16,1291–1298. 8. deBoerT:Thepharmacologicprofileofmirtazapine. ness of these drugs. The results are interesting and JClinPsychiatry,1996,57,Suppl4,19–25. needtobestudiedfurther. 9. DeffoisA,FageD,CarterC:Inhibitionofsynaptosomal veratridine-inducedsodiuminfluxbyantidepressantsand neurolepticsusedinchronicpain.NeurosciLett,1996, 220,117–1120. 10. DhillonS:Duloxetine:areviewofitsuseinthemanage- Summary mentofmajordepressivedisorderinolderadults.Drugs Aging,2013,30,59–79. 11. EardleyW,TothC:Anopen-label,non-randomized In summary, more preclinical and controlled clinical comparisonofvenlafaxineandgabapentinasmonother- trials might establish the safety and efficacy of antide- apyoradjuvanttherapyinthemanagementofneuro- pathicpaininpatientswithperipheralneuropathy.JPain pressant use in neuropathic pain. There is an experi- Res,2010,3,33–49. mental basis for the use of antidepressants in chronic 12. EisonAS,EisonMS,TorrenteJR,WrightRN,Yocca pain, but there is still a need for future preclinical be- FD:Nefazodone:preclinicalpharmacologyofanew havioral and biochemical studies due to their different antidepressant.PsychopharmacolBull,1990,26, mechanismofaction. 311–315. 13. FergusonJM:SSRIantidepressantmedications:Adverse effectsandtolerability.PrimCareCompanionJClin Psychiatry,2001,3,22–27. Acknowledgment: 14. FinnerupNB,Biering-SørensenF,JohannesenIL,Terkel- Thisstudywassupportedbystatutoryfundsandgrant senAJ,JuhlGI,KristensenAD,SindrupSHetal.: Demeter-1.1.2.5.4. Intravenouslidocainerelievesspinalcordinjurypain: arandomizedcontrolledtrial.Anesthesiology,2005,102, 1023–1030. 15. FryerJD,LukasRJ:Antidepressantsnoncompetitively References: inhibitnicotinicacetylcholinereceptorfunction.JNeu- rochem,1999,72,1117–1124. 16. GalerBS:Neuropathicpainofperipheralorigin:ad- 1. ArnoldLM,HessEV,HudsonJI,WelgeJA,BernoSE, vancesinpharmacologictreatment.,1995,45, KeckPEJr:A randomized,-controlled,double- Suppl9,S17–25. blind,flexible-dosestudyoffluoxetineinthetreatment 17. GendreauRM,ThornMD,GendreauJF,KranzlerJD, ofwomenwithfibromyalgia.AmJMed,2002,112, RibeiroS,GracelyRH,WilliamsDA etal.:Efficacyof 191–197. milnacipraninpatientswithfibromyalgia.JRheumatol, 2. AttalN,GaudéV,BrasseurL,DupuyM,GuirimandF, 2005,32,1975–1985. ParkerF,BouhassiraD:Intravenouslidocaineincentral 18. GoldsteinDJ,LuY,DetkeMJ,LeeTC,IyengarS: pain:adouble-blind,placebo-controlled,psychophysical Duloxetinevs.placeboinpatientswithpainfuldiabetic study.Neurology,2000,54,564–574. neuropathy.Pain,2005,116,109–118.

Pharmacological Reports, 2013, 65, 1611–1621 1619 19. HammackJE,MichalakJC,LoprinziCL,SloanJA, adults.CochraneDatabaseSystRev,2012,12, NovotnyPJ,SooriGS,TironaMT etal.:PhaseIII CD008242. evaluationofnortriptylineforalleviationofsymptoms 37. MorrisDW,TrivediMH,FavaM,WisniewskiSR, ofcis-platinum-inducedperipheralneuropathy.Pain, BalasubramaniGK,KhanAY,JainS,RushAJ:Diurnal 2002,98,195–203. moodvariationinoutpatientswithmajordepressivedis- 20. HirschfeldRM,MallinckrodtC,LeeTC,DetkeMJ: order.DepressAnxiety,2009,26,851–863. Timecourseofdepression-symptomimprovementdur- 38. NørregaardJ,VolkmannH,Danneskiold-SamsøeB: ingtreatmentwithduloxetine.DepressAnxiety,2005, Arandomizedcontrolledtrialofcitalopraminthetreat- 21,170–177. mentoffibromyalgia.Pain,1995,61,445–449. 21. IgnatowskiTA,SudR,ReynoldsJL,KnightPR, 39. OnishiH,YamamotoW,WadaM,NishidaT,WadaM, SpenglerRN:Thedissipationofneuropathicpainpara- SunagawaY,MiyaT etal.:Detectionandtreatmentof doxicallyinvolvesthepresenceoftumornecrosis inadvancedcancerpatientsduringadjuvantan- factor-alpha(TNF).Neuropharmacology,2005,48, algesictherapywithtricyclicantidepressants:casere- 448–460. portsandreviewoftheliterature.PalliatSupportCare, 22. JainR,JainS:Anti-inthetreatmentof 2007,5,411–414. chronicpain.PractPainManag,2011,44–50. 40. PickCG,PaulD,EisonMS,PasternakGW:Potentiation 23. JasminL,TienD,JanniG,OharaP:Isnoradrenaline ofopioidanalgesiabytheantidepressantnefazodone. asignificantfactorintheanalgesiceffectofantidepres- EurJPharmacol,1992,211,375–381. sants?Pain,2003,106,3–8. 41. 24. KhalifaM,DaleauP,TurgeonJ:Mechanismofsodium RasmussenPV,JensenTS,SindrupSH,BachFW: channelblockbyvenlafaxineinguineapigventricular TDM-basedimipraminetreatmentinneuropathicpain. myocytes.JPharmacolExpTher,1999,291,280–284. TherDrugMonit,2004,26,352–360. 42. 25. Korzeniewska-RybickaI,P³aŸnikA:Analgesiceffectof RowbothamDJ:Neuropathicpainandqualityoflife. antidepressantdrugs.PharmacolBiochemBehav,1998, EurJPain,2002,6,SupplB,19–24. 59,331–338. 43. RyderSA,StannardCF:Treatmentofchronicpain:anti- 26. KvinesdalB,MolinJ,FrolandA,GramLF:Imipramine depressant,antiepilepticandantiarrhythmicdrugs.Con- treatmentofpainfuldiabeticneuropathy.J.Amer.Med. tinEducAnaesthCritCarePain,2005,5,18–21. Assoc,1984,251,1727–1730. 44. SemenchuckMR,ShermanS,DavisB:Double-blind, 27. LeijonG,BoivieJ:Centralpost-strokepain–acon- randomizedtrialofbupropionSRforthetreatmentof trolledtrialofamitriptylineand.Pain, neuropathicpain.Neurology,2001,57,1583–1588. 1989,36,27–36. 45. ShahTH,MoradimehrA:Bupropionforthetreatmentof 28. LiTC,ShiahIS,SunCJ,TzangRF,HuangKC, neuropathicpain.AmJHospPalliatCare,27,333–336. LeeWK:Mirtazapinerelievespost-electroconvulsive 46. SimpsonDA:Gabapentinandvenlafaxineforthetreat- therapyheadachesandnausea:acaseseriesandreview mentofpainfuldiabeticneuropathy.JClinNeuromusc oftheliterature.JECT,2011,27,165–167. Dis,2001,3,53–62. 29. MattiaC,PaolettiF,ColuzziF,BoanelliA:Newantide- 47. SindrupSH,JensenTS:Efficacyofpharmacological pressantsinthetreatmentofneuropathicpain.A review. treatmentsofneuropathicpain:anupdateandeffectre- MinervaAnestesiol,2002,68,105–114. latedtomechanismofdrugactionPain,1999,83, 30. MaxMB,LynchSA,MuirJ,ShoafSE,SmollerB,Dub- 389–400. nerR:Effectsofdesipramine,amitriptyline,andfluoxet- 48. SindrupSH,OttoM,FinnerupNB,JensenTS:Antide- ineonpainindiabeticneuropathy.NewEnglJMed, pressantsinthetreatmentofneuropathicpain.BasicClin 1992,326,1250–1256. PharmacolToxicol,2005,96,399–409. 31. MazzaM,MazzaO,PazzagliaC,PaduaL,MazzaS: 49. Smith AJ: The analgesic effects of selective serotonin re- Escitalopram20mgversusduloxetine60mgforthe uptake inhibitors. J Psychopharmacol, 1998, 12, 407–413. treatmentofchroniclowbackpain.ExpertOpinPhar- 50. macother,2010,11,1049–1052. StahlSM,GradyMM,MoretC,BrileyM:SNRIs:their 32. McCleaneG:Antidepressantsasanalgesics.CNSDrugs, pharmacology,clinicalefficacy,andtolerabilityincom- 2008,22,139–156. parisonwithotherclassesofantidepressants.CNS 33. MicóJA,ArdidD,BerrocosoE,EschalierA:Antide- Spectr,2005,10,732–747. pressantsandpain.TrendsPharmacolSci,2006,27, 51. StokesPE:Fluoxetine:afiveyearsreview.ClinTher, 348–354. 1993,15,216–243. 34. MikaJ,OsikowiczM,RojewskaE,KorostynskiM, 52. SuterMR,KirschmannG,LaedermannCJ,AbrielH, Wawrzczak-BargielaA,PrzewlockiR,PrzewlockaB: DecosterdI:Rufinamideattenuatesmechanicalallodynia Differentialactivationofspinalmicroglialandastroglial inamodelofneuropathicpaininthemouseandstabi- cellsinamousemodelofperipheralneuropathicpain. lizesvoltage-gatedsodiumchannelinactivatedstate. EurJPharmacol,2009,623,65–72. Anesthesiology,2013,118,160–172. 35. MikaJ,ZychowskaM,Popiolek-BarczykK,Rojewska 53. TzschentkeTM,ChristophT,SchröderW,Englberger E,PrzewlockaB:Importanceofglialactivationinneuro- W,DeVryJ,JahnelU,KögelBY:Tapentadol:withtwo pathicpain.EurJPharmacol,2013,716,106–119. mechanismsofactioninonemoleculeeffectiveagainst 36. MooreRA,DerryS,AldingtonD,ColeP,WiffenPJ: nociceptiveandneuropathicpain.Preclinicaloverview. Amitriptylineforneuropathicpainandfibromyalgiain Schmerz,2011,25,19–25.

1620 Pharmacological Reports, 2013, 65, 1611–1621 Antidepressantsinchronicpain Joanna Mika et al.

54. VerduB,DecosterdI,BuclinT,StiefelF,BerneyA: 60. ZhuJ,WeiX,FengX,SongJ,HuY,XuJ:Repeatedad- Antidepressantsforthetreatmentofchronicpain.Drugs, ministrationofmirtazapineinhibitsdevelopmentofhy- 2008,68,2611–2632. peralgesia/allodyniaandactivationofNF-kBinarat 55. VickersER,CousinsMJ:Neuropathicorofacialpain. modelofneuropathicpain.NeurosciLett,2008,433, Part2–Diagnosticprocedures,treatmentguidelinesand 33–37. casereports.AustEndodJ,2000,26,53–63. 61. ZhuJ,WeiX,LiuJ,HuY,XuJ:Interactionofgliaacti- 56. WatkinsLR,HutchinsonMR,JohnstonIN,MaierSF: vationandneurotransmissioninhippocampusofneuro- Glia:novelcounter-regulatorsofopioidanalgesia. pathicratstreatedwithmirtazapine.ExpClinPsycho- TrendsNeurosci,2005,28,661–669. pharmacol,2009,17,198–203. 57. WatsonCP,ChipmanM,ReedK,EvansRJ,BirkettN: 62. ZieglerD:Painfuldiabeticneuropathy:advantageof Amitriptylineversusmaprotilineinpostherpeticneural- noveldrugsoverolddrugs?DiabetesCare,2009,32, gia:arandomized,double-blind,crossovertrial.Pain, Suppl2,S414–S419. 199,48,29–36. 63. ZieglerD:Painfuldiabeticneuropathy:treatmentandfu- 58. WongDT,BymasterFP:Dualserotoninandnoradrena- tureaspects.DiabetesMetabResRev,2008,24,Suppl1, lineuptakeinhibitorclassofantidepressantspotentialfor S52–S57. greaterefficacyorjusthype?ProgDrugRes,2002,58, 169–222. 59. WrzosekA,ObaraI,WordliczekJ,PrzewlockaB:Effi- cacyoftramadolincombinationwithdoxepinorvenla- faxineininhibitionofnociceptiveprocessintherat modelofneuropathicpain:anisobolographicanalysis. Received: July15,2013; intherevisedform: October30,2013; JPhysiolPharmacol,2009,60,71–78. accepted: November8,2013.

Pharmacological Reports, 2013, 65, 1611–1621 1621