Abstract
Migraine is a complex disorder that is characterized by an assortment of neurological and systemic effects. While headache is the most prominent feature of migraine, a host of symptoms affecting many physiological functions are also observed before, during, and after an attack. Furthermore, migraineurs are heterogeneous and have a wide range of responses to migraine therapies. The recent approval of calcitonin gene related peptide (CGRP) based therapies has opened up the treatment of migraine and generated a renewed interest in migraine research and discovery. Ongoing advances in migraine research have identified a number of other promising therapeutic targets for this disorder. In this review we highlight emergent treatments within the following biological systems: pituitary adenylate cyclase activating protein (PACAP), two non-mu opioid receptors that have low abuse liability – the delta and kappa opioid receptors, orexin, and nitric oxide-based therapies. Multiple mechanisms have been identified in the induction and maintenance of migraine symptoms; and this divergent set of targets have highly distinct biological effects. Increasing the mechanistic diversity of the migraine tool box will lead to more treatment options and better patient care.
Keywords: delta opioid receptor, kappa opioid receptor, PACAP, orexin, soluble guanylyl cyclase, NOS
Introduction
Headache has a high global prevalence and has been identified as a leading cause of disability worldwide1, 2. However, until recently the only pharmacotherapy specifically targeted for migraine was the triptans. The recent approval of anti-calcitonin gene related peptide (CGRP) therapies appear to be a breakthrough in migraine prevention, and they have created a great deal of excitement in the field. The development of CGRP exemplifies how translational research can result in targeted strategies specific for the treatment of migraine. The success of CGRP based therapies has also spurred the investigation for new therapies to encourage the expansion of the migraine toolbox. Increasing the variety of migraine-selective treatments is especially beneficial as not all patients respond to CRGP-based treatments, and patients dosed on CGRP antibodies might still need rescue therapies. In addition, the consequences of long acting CGRP based antibodies on pregnancy are still unclear3 thus further supporting the need for mechanistically diverse treatment strategies.
Migraine is a brain disorder that produces a wide array of neurological and systemic symptoms. The pathophysiology of migraine is correspondingly complex and includes changes in the vasculature, central and peripheral pain processing, and inflammation. Migraine can be treated by a number of different drug classes which supports the idea that divergent mechanisms can regulate the migraine brain state4. Anti-convulsants, beta blockers, tricyclic antidepressants, and botulinum toxin have all been used as migraine preventives with varying levels of efficacy5, 6. Prior to the approval of anti-CGRP therapies, most of these therapies were poorly tolerated or ineffective, and over 50% of chronic migraine patients were dissatisfied with their treatment7. Migraineurs are a heterogeneous population, and have shown a wide range of response rates to all of these therapies, including CGRP targeting drugs6, 8. Ongoing advances in migraine have provided novel insight into the diverse mechanisms regulating this disorder. In this review we will highlight the following emerging therapies: pituitary adenylate cyclase activating peptide (PACAP), two non-μ opioid receptors - δ and κ receptors, orexin, and nitric oxide (NO) based therapies. All of these targets have compelling evidence supporting their role in migraine; and their physiological diversity highlights the range of mechanisms that can regulate headache disorders. The authors used PubMed as their primary source of information, and only peer review articles were considered. For data on clinical trials, ClinicalTrials.gov and PubMed were utilized.
Pituitary Adenylate Cyclase Activating Peptide
Pituitary adenylate cyclase activating peptide (PACAP) has garnered attention in recent years as a key peptide in migraine pathophysiology9–11. PACAP is a vasoactive intestinal peptide (VIP) and as such is part of the glucagon-growth hormone releasing factor-secretin superfamily12. PACAP is found in two forms, the 38 amino acid peptide, PACAP38; and a truncated PACAP2713. PACAP38 is the predominant form and accounts for 90% of the PACAP in mammalian tissue14. PACAP38 is commonly found in both the peripheral and central nervous system. In humans, PACAP38 is expressed in the otic and sphenopalatine ganglia15. These neurons in turn project to the perivascular parasympathetic nerves to the cranial circulation14. PACAP38 is also expressed in a number of other regions that regulate headache, including the pituitary, trigeminal ganglia, trigeminal nucleus caudalis, dorsal horn of the spinal cord, brain stem, hypothalamus, and cortex16. Given its wide spread distribution, PACAP38 has been implicated in a variety of physiological functions including vasodilation, neurotransmission, neuroprotection, and neuromodulation12. As part of the VIP family PACAP38 can activate three different Gs G protein coupled receptors (GPCRs): VPAC1, VPAC2, and PAC1; and activation of these receptors results in stimulation of adenylyl cyclase and increased levels of cyclic adenosine monophosphate (cAMP). PACAP and its related peptide, VIP, can bind with the same affinity to both VPAC1 and 2, but PACAP alone has up to a 1000X higher affinity for the PAC1 receptor17, 18.
The first clear link between PACAP and headache was made through human provocation studies. In an initial study, infusion of PACAP38 into healthy volunteers resulted in 83% of participants developing headache19, which is in line with the expression of the PACAPergic system in head pain processing regions and its vasodilatory effects. Subsequent studies compared the effects of PACAP on migraine without aura patients versus healthy controls. Although PACAP38 evoked headache in most participants, it specifically evoked a delayed migraine attack in 58% of migraineurs20. Furthermore, PACAP38 infusion resulted in mild to moderate dilation of both the middle cerebral artery (MCA) and the superficial temporal artery (STA)20. In a follow up study, the same group used high resolution magnetic resonance angiography to examine the middle meningeal artery (MMA) and found significant and sustained dilation after PACAP38 that coincided with headache in healthy volunteers21. Interestingly, when patients were administered sumatriptan, PACAP38-induced dilation of the MMA was reduced as was headache severity21. This work demonstrates that PACAP38 is sufficient to produce vasodilation of meningeal arteries and evoke headache.
Although there is considerable physiological overlap between PACAP and VIP, including their affinity for VPAC receptors, PACAP appears to be more closely tied to migraine pathophysiology. In human provocation studies comparing the two peptides, the infusion of PACAP38 produced migraine in 73% of migraine without aura subjects as compared to 18% by VIP22. Interestingly both PACAP38 and VIP produced dilation of the MMA22, and this study highlights that vasodilation alone is not sufficient to produce migraine. The effects of PACAP38 and VIP on brain connectivity was also examined using resting state functional magnetic resonance imaging. Migraine without aura participants showed altered functional connectivity of the salience, sensorimotor, and default networks following PACAP38 infusion, effects not observed with VIP23. Interestingly, infusion of PACAP38 into migraineurs does not result in increased blood levels of calcitonin gene related peptide (CGRP)24, suggesting that PACAP38 can produce migraine independent of this endogenous pro-migraine neuropeptide. Together, these human provocation studies support the role of PACAP38 in migraine development.
There is also evidence that PACAP38 levels are also altered in spontaneous headache conditions. PACAP38 plasma levels were found to be upregulated in migraineurs during the ictal period25. A separate group found a similar increase during migraine, and showed that treatment with sumatriptan correspondingly decreased PACAP38 levels26. Furthermore, elevated PACAP38 levels were also observed during the attack phase in episodic cluster headache patients27. Interestingly, PACAP38 levels were also found to be decreased during the interictal period relative to a healthy control group25, 27, suggestive of a broader dysregulation of the PACAPergic system in headache sufferers.
Preclinical studies have further explored the mechanism by which PACAP38 may be modulating headache. Infusion of PACAP38 in mice produces photophobia, meningeal dilation, and increased c-fos expression (a marker of cellular activation) in trigeminal nucleus caudalis28. More broadly, PACAP also appears to regulate the photophobia and meningeal effects induced by the human migraine trigger, nitroglycerin, as these endpoints are reduced in PACAP knockout mice28. In rats, PACAP38 but not VIP resulted in delayed activation and sensitization of central trigeminovascular neurons10. In this study PACAP38 caused an increase in spontaneous firing and hypersensitivity to both central and peripheral somatosensory stimulation, which occurred at a distinctly different time point from the meningeal dilation10. Further, dural meningeal vasodilation induced by PACAP38 was mediated through VPAC2 receptors, while the delayed activation and sensitization of trigeminocervcial neurons was mediated by PAC110; another example of nociceptive and vascular effects being regulated separately. PACAP38 was also shown to produce degranulation of peritoneal and dural mast cells which in turn could be responsible for PACAP-induced vasodilation of MMA29, 30 , and this effect may be mediated by a non-canonical PACAP signaling pathway31.
In addition to the role of PACAP in migraine, PACAP38 and PAC1 are also heavily implicated in stress and emotional processing. In rodents, administration of PACAP38 increases behaviors associated with anxiety and depression32; and correspondingly, PACAP knockout results in decreased anxiety and depression-related phenotypes33, 34. In addition, studies in humans indicate that polymorphism of the PAC1 gene confers increased risk of post-traumatic stress disorder (PTSD)35, and increased PACAP38 levels were also associated with PTSD36. Considering the high co-morbidity between headache and emotional disorders makes the PACAPergic system appears to be particularly attractive as a novel therapeutic target.
Currently there is no clinically available PACAP-specific treatment. However, a Phase IIa randomized, double blind, placebo-controlled trial is underway to examine the efficacy of AMG-301, a PAC1 receptor targeting antibody, on migraine prevention37 (Table 1). In preclinical studies, this antibody inhibited nociceptive activity in the trigeminocervical complex to the same extent as sumatriptan38. Although it has not yet entered clinical trial, a monoclonal antibody targeting PACAP38, ALD1910, is also in development for the treatment of migraine patients 39.
Table 1:
Clinical Trials and Pipeline for Emerging Therapeutic Targets for Migraine
| Target | Drug Name | Mechanism | Condition | Status | Phase | Primary Endpoint Met? | Manufacturer | ClinicalTrials.gov identifier |
|---|---|---|---|---|---|---|---|---|
| PACAP | AMG-301 | Monoclonal Antibody PAC1 Receptor | Migraine | Completed in 2019 | Phase II | Results not available | Amgen | |
| ALD1910 | Monoclonal Antibody PACAP38 | Migraine | Ongoing | Pre-clinical | N/A | Alder Biopharmaceutics | Company website | |
| Delta Opioid Receptor | ADL5859 | δ receptor agonist | Osteoarthritis of the knee | Completed in 2010 | Phase II | No | Adolor Corporation; Pfizer | |
| ADL 5747 | δ receptor agonist | Osteoarthritis of the knee and postherpetic neuralgia | Completed in 2010 | Phase II | No | Adolor Corporation; Pfizer |
|
|
| AZD7268 | δ receptor agonist | Major Depressive Disorder | Completed in 2010 | Phase II | No | AstraZeneca | ||
| AZD2327 | δ receptor agonist | Major Depressive Disorder | Completed in 2011 | Phase II | No | AstraZeneca | ||
| TRV 250 | δ receptor agonist | Migraine | Completed in 2018 | Phase I | Yes | Trevena | Company website | |
| Kappa Opioid Receptor | CR845 | peripherally restricted κ agonist | Hysterectomy Bunionectomy Osteoarthritis |
Completed in 2012 2013 2017 |
Phase II | Yes Yes Not available |
Cara Therapeutics |
|
| CERC501 (LY-2456302) | κ-opioid receptor antagonist | Anxiety Disorder | Completed 2017 | Phase II | Yes | Eli Lilly | ||
| Orexin | Filorexant | Dual orexin receptor antagonist | Migraine preventative | Completed in 2013 | Phase II | No | Merck Sharp & Dohme Corp. | |
| Suvorexant (DORA-12) | Dual orexin receptor antagonist | Post-Traumatic Stress Disorder | Ongoing | Phase IV | Results not available | Merck Sharp & Dohme Corp | ||
| Nitric Oxide Synthase (NOS) |
GW274150 |
iNOS inhibitor | Migraine | Completed in 2010 | Phase II | No | GlaxoSmithKline | |
| NXN-188 | nNOS inhibitor +5HT1B/1D agonist | Migraine with aura | Completed in 2010 | Phase II | Results not available No |
NeurAxon |
|
|
| NXN-188 | nNOS inhibitor +5HT1B/1D agonist | Migraine without aura | Completed in 2010 | Phase II | Yes | NeurAxon |
Non-Mu Opioid Receptors - Delta Opioid Receptor
Opioid analgesics in current use (e.g. morphine, hydrocodone, oxycodone, meperidine) act primarily at the μ opioid receptor40 and are still commonly prescribed for headache41. However, while opioids may provide acute relief, regular use of μ receptor agonists can result in increased headache severity, and progression of headache from an episodic to a chronic state42. This form of medication overuse headache43 is not only difficult to treat, but can also result in opioid dependence and abuse which has resulted in a significant public health crisis44–46. Evidence from preclinical studies indicate that other members of the opioid receptor family may provide effective alternatives to μ-based approaches. The opioid receptor family is comprised of μ, δ, κ opioid receptors, and the nociceptin/ORL-1 receptor. All four receptors share homology but are encoded by different genes and produce highly distinct physiological effects. Opioid receptors are activated by endogenous opioid peptides. Although these peptides can bind across opioid receptors there is some selectivity; and enkephalins have the highest affinity for δ receptors, endorphins for μ receptors, dynorphins for κ receptors, and nociceptin for nociception/ORL1 receptor47. The δ and κ receptors have been identified as novel therapeutic targets for migraine48, 49 and are discussed below.
δ receptors are expressed in a number of regions involved in headache, including trigeminal and dorsal root ganglia, trigeminal nucleus caudalis, and the cortex50–52. δ receptors are found on a small subset of dorsal root ganglia that express the pro-migraine peptide CGRP53. Further, its expression in limbic regions such as the striatum, hippocampus, and amygdala support its role in emotional regulation50, 54. Relative to μ receptors, δ receptors have low abuse liability. In rodent and non-human primate models, δ agonists do not facilitate rewarding behaviors, nor do they cause physical dependence55–58. In addition, δ agonists produce less respiratory depression and have fewer effects of gastrointestinal transit59, 60 relative to μ agonists. In animal model, δ receptor agonists can produce pro-convulsant effects. However, not all δ agonists show this adverse effect, and it may be possible to separate out this consequence from pain-relieving effects through drug development61.
Although δ agonists are not highly effective in acute pain, they have shown efficacy in a number of chronic pain states, including models of inflammatory and neuropathic pain62. δ agonists appear to be particularly effective in models of headache48. In a behavioral model of migraine-associated pain, nitroglycerin, a known human migraine trigger63, caused severe mechanical hyperalgesia in mice that was prevented by three different δ agonists48. Another lab also found that heat hyperalgesia induced by nitroglycerin was also blocked by the δ agonist, SNC8064. Further, more recent evidence suggests that δ agonists could be effective for a number of different types of headache. SNC80 effectively inhibited cephalic allodynia in mouse models of chronic migraine, and medication overuse headache associated with triptans65. In a model of post-traumatic headache, acute SNC80 blocked established cephalic allodynia65, and long term treatment with this δ agonist also prevented the chronification of post-traumatic headache66. In a model of opioid-induced hyperalgesia, chronic morphine treatment produced a severe cephalic and peripheral allodynia that was inhibited by treatment with a δ agonist65. This latter finding particularly supports the distinction between the μ and δ receptors, as chronic μ receptor stimulation by morphine resulted in a correlate of mediation overuse headache which could still be blocked by δ receptor activation. Preclinical studies indicate that δ opioid receptors are functionally upregulated during chronic peripheral pain states62, 67–69, and may serve as an endogenous protective mechanism to counteract increased pain signaling. Ongoing studies are underway to determine if this upregulation is also observed in headache models. Furthermore, moderate to high expression of δ receptors has been observed in trigeminal ganglia, trigeminal nucleus caudalis52 and the dura70 suggesting that it may be particularly important for the regulation of headache specifically.
The δ receptor was also found to be effective in preclinical studies examining other symptoms of migraine. Cortical spreading depression (CSD) is the slowly propagated wave of depolarization followed by inhibition of brain activity thought to be the underlying physiologic correlate of migraine aura71. Migraine preventives have been shown to decrease CSD72, 73, and it is therefore used as a screening tool for novel migraine therapies. The δ agonist, SNC80, significantly decreased the number of CSD events evoked by KCl74 suggesting that δ receptors may also have potential as a preventive treatment. Chronic δ activation has also been examined for the development of medication overuse headache; and compared to sumatriptan or morphine, SNC80 produced a limited form of overuse allodynia65, further supporting its development as a headache therapy.
The δ receptor has been shown to play an important role in emotional regulation54, and δ agonists have been developed for the treatment of anxiety and depression. Knockout of the δ receptor results in anxiogenic and depressive-like behaviors75, and δ agonists have anxiolytic and anti-depressant effects in animal models54, 76. This positive effect of emotional modulation may be particularly beneficial considering the high co-morbidity between headache and emotional disorders77. In a study of migraine-associated negative affect, nitroglycerin treatment was paired with a specific chamber which resulted in conditioned place aversion, and this aversion was prevented by SNC8074. This result suggests that δ agonists may also improve the negative emotional state associated with migraine.
δ agonists have been developed and tested for the treatment of pain, and anxiety and depression78 (Table 1). Two δ agonists, ADL5859 and ADL5747 were investigated for safety and efficacy in osteoarthritis of the knee, and ADL5747 was also tested in patients with postherpetic neuralgia. The results have not been published in a peer-reviewed journal, however, the posted results from ClinicalTrials.gov show that neither study met its primary endpoint, and the latter study was terminated early due to lack of efficacy. Although disappointing, these pain conditions are mechanistically distinct from migraine, and these trials may not reflect efficacy of δ agonists in headache. Two other δ agonists, AZD726879 and ADZ232780, have been investigated in phase I and phase IIa studies for safety and potential efficacy in major depressive disorder and anxiety. Again, neither study reached primary endpoints but secondary analysis indicated benefit in anxiety. It is important to note that in all of the clinical trials for δ agonists, all 4 compounds were generally well tolerated with limited to no adverse events . A more recent phase 1 clinical trial was performed to test TRV250 for acute migraine. This δ agonist was specifically developed to avoid seizure liability, while maintaining pain-relieving effects. Data from this study has only been disclosed in a press release from the company, and indicates that in this healthy volunteer study TRV250 showed safety, tolerability and a pharmacokinetic profile that would support subsequent trials for efficacy. The δ receptor appears to be a promising therapeutic target for migraine, and future studies are ongoing to determine its clinical utility.
Non-Mu Opioid Receptors – Kappa Opioid Receptor
The κ opioid receptor has also been implicated in a number of physiological processes, including the regulation of pain, reward, and stress. The κ receptor is distributed throughout the brain and high expression is observed in regions related to mood, motivation, and cognitive function such as the nucleus accumbens (NAc), cerebral cortex, hippocampus, and hypothalamus; as well as in pain processing regions such as the periaqueductal grey, spinal cord, and dorsal root ganglia51, 81–83. Activation of the κ receptor is pain relieving and agonists have been investigated for the treatment of pain84. However, κ agonists are also found to produce psychotomimesis85 and dysphoria84 in humans, which has limited their development for clinical use. Peripherally restricted κ agonists have been developed for pain management. ADL10-0101, was found to be effective for pancreatitis in a small study, but its effects in females were sensitive to menstrual cycle, and it also produced headache86. Several clinical trials have been performed to investigate the efficacy of CR845 in acute (hysterectomy and bunionectomy) and chronic (osteoarthritis) pain conditions with generally positive results (Table 1). However, to the best of our knowledge there are no published studies showing the effectiveness of κ agonists in headache models.
Inhibition of the κ receptor has emerged as a potential therapeutic strategy for headache disorders. Upregulation of the endogenous κ opioid peptide, dynorphin, has been identified as a marker of stress87; and for this reason κ antagonists are being investigated for the treatment of anxiety, depression, and drug abuse and relapse88. Stress has frequently been cited as the most common migraine trigger89, but despite its prevalence, the pathophysiology of how stress actually triggers migraine is unclear. Xie et al. used a preclinical model of stress-induced headache to examine the effect of κ antagonists on cephalic pain49. Rats were treated with chronic sumatriptan to model medication overuse headache, which in turn made them hypersensitive to a bright light stress cue. Stress produced an increase in plasma CGRP and cephalic and peripheral allodynia, which was blocked by systemic injection of two different κ antagonists49. Further, this hypersensitivity appears to be mediated through an upregulation of dynorphin in the amygdala, and intra-amygdala injection of κ antagonists were also anti-allodynic49. The novel kappa opioid receptor antagonist, CERC-501 (formerly known as LY2456302) is in clinical trials for mood and anxiety disorders and was shown to be safe and well-toleranated90 (Table 1). Given these early promising clinical results κ receptor antagonists may serve as promising therapies for headache.
Orexin
Orexins, also known as hypocretins, have been widely implicated in homeostatic regulation; and dysfunction of the orexinergic system in migraine has recently been explored91–93. The orexin family is comprised of two neuropeptides, orexin A (OXA) and orexin B (OXB), which are derived from the precursor peptide, prepro-orexin94. Orexin A is 33 amino acids in length, while orexin B is 28 residues, and mature versions of these peptides are entirely (OXA) or highly (OXB) conserved in mammals95. Although these peptides are synthesized exclusively in the dorsolateral hypothalamus, orexinergic neurons project throughout the CNS, including to other hypothalamic nuclei, thalamus, cortex, trigeminal nucleus caudalis, and spinal cord91. Both orexin peptides bind to orexin receptor 1 (ORX1) and orexin receptor 2 (ORX2), both of which are Gq GPCRs, and as such activation results in increased intracellular Ca2+94. OXA has equal affinity for both orexin receptors, while OXB has a ten-fold increased affinity for OXR294. The orexin receptors are broadly expressed in the brain, and usually correspond with orexin peptide expression91, 93.
The orexinergic system is involved in the dynamic regulation of a number of physiological processes; including feeding, sleep/wake cycle, stress, and motivation96. Many of these processes are disrupted during migraine, particularly in the premonitory phase. For example, changes in appetite are often observed during headache, and disruption to normal feeding patterns are considered a potential migraine trigger. Orexins may regulate this aspect of headache disorders, as they play an important role in appetite and food consumption96. Orexin peptide and receptor levels are upregulated following food deprivation or fasting, and activation of the arcuate nucleus by orexins promotes feeding behavior97, 98. Correspondingly, orexin levels are downregulated when blood glucose levels are high99, 100. Changes in orexins and other hypothalamic peptides that regulate energy and metabolism could contribute to the perturbation in appetite and feeding associated with headache disorders.
Orexins levels are tightly regulated with circadian rhythm, and is thought to facilitate the transition from sleep to wakefulness. Peak orexin levels are observed during wakefulness, and orexinergic neurons are most active during wake with frequent firing just before waking101, 102. This role of orexin is most starkly demonstrated in narcolepsy. Knockout of the orexin peptides or mutation of the ORX2 receptor results in murine103 and canine104 narcolepsy, and human narcoleptics show decreased circulating levels of orexin105. Consistent with the role of orexin in migraine, narcoleptic patients have a significantly higher prevalence for migraine, and 64% of female and 45% of males suffer from migraine compared to 30% and 8%, respectively, in the general population93, 106. More generally, sleep disturbances are known to increase susceptibility to migraine; and orexin may act at the interface between changes in circadian rhythm and headache disorders. A recent study found that following traumatic brain injury, mice showed decreased orexin positive neurons in the hypothalamus; and this may be involved in altered sleep patterns associated in TBI107 and subsequent PTH. Disturbances in the orexinergic system could serve as a link between headache disorders and sleep dysregulation.
The orexinergic system has also been implicated in pain processing. Knockout of orexin results in increased pain sensitivity in inflammatory pain models, and decreased stress induced hyperalgesia108. Orexins have also been shown to be modulatory in models of cephalic pain. Microinjection of orexin A or B into the posterior hypothalamus decreases or increases cephalic pain sensitivity, respectively109. Similarly, orexin A but not B can inhibit neurogenic dural vasodilation110 and trigeminal neuron firing111, and it appears that orexin A through activation of ORX1 signals distinctly from orexin B.
A role for the orexinergic system in headache pathophysiology has been supported by studies measuring orexin levels in various headache disorders. Significantly decreased levels of CSF orexin was observed in both episodic and chronic cluster headache patients relative to controls112. In contrast, in chronic migraine and medication overuse headache patients, orexin A levels were found to be increased113. Although seemingly contradictory, these studies support the notion that the orexin system is dysregulated in headache.
Dual orexin receptor antagonists (DORAs) have been developed for clinical use, and suvorexant (DORA-12) has been approved for the treatment of insomnia. In a preclinical model of temporomandibular joint inflammation, DORA-12 acutely attenuated facial allodynia114. Furthermore, this antagonist also blocked electrophysiological signaling in the trigeminocervical complex in response to dural stimulation115. Interestingly, DORA-12 also increased the threshold for CSD, a correlate of migraine aura115. These studies suggest that antagonism of the orexin system could be an effective migraine therapy. There has been one small clinical trial to test the effect of filorexant, another dual orexin receptor antagonist, as a migraine preventive (Table 1). In this randomized, double-blind, placebo controlled trial patients were treated with 10 mg nightly for three months. There was no statistically significant difference between filorexant treated and placebo controls116, which may be explained by the dosing regimen or half-life of the drug93. Although disappointing, there is still hope for targeting the orexinergic system in migraine. Preclinical studies indicate that ORX1 and ORX2 can have divergent behavioral effects109–111; which is supported by the finding that certain CNS regions only express one receptor (eg locus coeruleus and ORX1, and the rostroventromedial medulla and ORX2). Further studies are required to clearly define how changes in the orexinergic system correspond to headache susceptibility and symptomatology.
Nitric Oxide Pathway
Nitric oxide (NO) is an endogenous gaseous signaling molecule that is important for several biological processes including vascular tone, neurotransmission and immune defense. NO is ubiquitously expressed in almost every mammalian cell type117. NO production and signaling has been heavily implicated in migraine pathophysiology118, 119. The known human migraine trigger, nitroglycerin, is a NO donor and has been used extensively in human migraine provocation63 studies and as an animal model of migraine120, 121. Nitroglycerin is a potent vasodilator, and it produces a rapid headache in both migraineurs and healthy controls. However, migraine patients go on to develop a delayed migraine attack several hours after this vasodilatory effect122 again suggesting that vascular alterations and induction of migraine are not always coupled.
Endogenous NO is produced by the oxidation of L-arginine into NO and L-citrulline123. This reaction is catalyzed by three isoforms of nitric oxide synthase (NOS). Each of the NOS family members corresponds to the tissue type that they were initially discovered in and where they are largely expressed123. Neuronal NOS (nNOS) is expressed in neurons and can be found in both the central and peripheral nervous system117. Endothelial NOS (eNOS) was originally discovered in vascular endothelial cells, but is also expressed in platelets, cardiomyocytes, and the brain117. NO produced by eNOS appears to largely regulate vascular tone124. Both nNOS and eNOS are constitutively activated and rely on increase of intracellular calcium and subsequent binding to calmodulin117. The third NOS isoform, inducible NOS (iNOS), is expressed in macrophages, glia, and neurons. iNOS is unique because unlike the other two isoforms it is not constitutively active, but is induced by infection and pro-inflammatory cytokines and serves as part of the host immune defense system123. iNOS can rapidly produce up to 1,000X more NO than nNOS or eNOS117.
NO is highly membrane permeable and can activate signaling cascades both within the cell it was produced as well as in neighboring cells. The only known receptor for NO in the body is soluble guanylyl cyclase (sGC)123, which upon activation converts guanosine-5’-triphosphate (GTP) to cyclic guanosine monophosphate (cGMP). Signaling through this pathway includes activation of cGMP-specific ion channels, alterations in Ca2+ homeostasis, and activation of the transcription factor CREB125,126. Several lines of evidence indicate that the sGC pathway has an important function in migraine etiology and maintenance. cGMP is broken down to GMP by phosphodiesterase 5 (PDE5). The PDE5 inhibitor, sildenafil, can produce a headache in healthy control patients along with a migraine like attack in migraineurs121. In a preclinical study direct activation of the sGC pathway by the novel sGC stimulator, VL-102, produced cephalic allodynia which was blocked by known migraine treatments127. In addition, a sGC inhibitor, ODQ, completely abolished migraine-associated allodynia induced by nitroglycerin127. Interestingly, in a chronic migraine model, ODQ was also able to inhibit established allodynia in the absence of an exogenous NO donor, which suggests that upregulation of the sGC pathway may be a key mediator for the maintenance of the chronic migraine state. This study strengthens the connection between the NO-sGC pathway and headache, and open the possibility for inhibitors or negative modulators of sGC as novel migraine therapeutics.
Although the sGC pathway is seen as the primary NO signaling mechanism, NO can also produce significant oxidative stress through the formation of the reactive nitrogen species peroxynitrite117. The role of peroxynitrite in migraine is still unlcear128, but migraine is commonly observed in patients with mitochondrial diseases129, and antioxidants such as riboflavin and coenzyme Q10 can be effective migraine preventives130, 131. Future studies will determine if peroxynitrite scavengers could be promising migraine therapies132.
The blockade of endogenous NO production by NOS inhibitors (NOSi) has been extensively investigated for the treatment of migraine, and has been summarized in a recent review118. In a clinical study, the non-selective NOS inhibitor, L-NG -methylarginine hydrochloride (546C88) provided significant headache relief during spontaneous migraine attacks relative to placebo133. A different non-selective NOSi, NG L-arginine hydrochloride (L-NMMA) was more efficacious than placebo in chronic tension-type headaches134. These studies provide proof of concept for the targeting of NO for the treatment of migraine. However, non-selective NOSi produce significant cardiovascular effects and increased blood pressure which limits their therapeutic potential118. Similarly, eNOS is found predominantly in the vascular endothelium and is considered to be fundamental for healthy cardiovascular function135 and therefore is also not a highly desirable target for migraine.
iNOS has been explored for the treatment of headache disorders. In a rodent model of migraine, nitroglycerin results in an upregulation of iNOS mRNA and protein in dural meningeal macrophages 136, 137. Similarly, in humans blood levels of iNOS are increased during a migraine attack, and levels lower after the attack had subsided138. Despite the evidence, targeting iNOS for migraine has proven to be less effective than hoped. Two clinical trials investigated the effect of the iNOS inhibitor, GW274150, to prevent or treat migraine attacks, and neither reached primary endpoints (Table 1)139, 140. These negative results suggest that targeting iNOS might not be sufficient to inhibit migraine, and this treatment strategy has been largely abandoned118.
nNOS is expressed throughout peripheral and central regions that regulate headache pain, and therefore could serve as an ideal candidate for drug development141. Similar to iNOS, nitroglycerin administration in rodents evoked increased nNOS expression in two key pain processing regions, the trigeminal ganglia and the trigeminal nucleus caudaulis142, 143. A nNOS inhibitor decreased meningeal blood vessel diameter and inhibited neurogenic dural meningeal blood vessel dilation144. In a separate model the selective nNOS inhibitor NXN-413 inhibited the elevation in CGRP by KCl145. Similarly, in a dural inflammation model NXN-413 inhibited increased vasodilation145. In another preclinical model of MOH a different nNOS inhibitor, NXN-323, was able to reverse established periorbital and hind paw allodynia146. Clinically, NXN-188, a mixed nNOSi-triptan, has been investigated in two clinical trial studies for the treatment of migraine with and without aura. Preliminary studies suggest that this molecule may be effective in migraine treatment147, 148, but there has been limited movement on this strategy since 2010. nNOS alone, or in combination with a 5-HT1B/1D agonist, may still prove to be a novel and effective strategy for the treatment of migraine.
Conclusions
It is an exciting time in migraine research and patient care. The success of CGRP based therapies reveals the power of a targeted treatment strategy for migraine. It has also generated increased interest in basic and translational headache research. A few of the discussed targets were tested in clinical trials, but did not reach primary endpoints. An emphasis on understanding the dominant signaling pathways underlying different types of headache could result in more directed clinical trials with a greater chance of success. Considering the prevalence and heterogeneity of migraine the development of mechanistically diverse targets will lead to greater treatment options. There are a number of promising targets that have been identified as migraine regulators and future work will hopefully lead to the next breakthrough drug.
Acknowledgements
This work was supported by NIH grants DA040688 and NS109862. ZB is a member of the Graduate Program in Neuroscience at University of Illinois at Chicago.
Footnotes
Declaration of Competing Interests
The authors declare no potential conflicts of interest with respect to the research, authorship, and/or publication of this article.
Bibliography
- 1.Global, regional, and national incidence, prevalence, and years lived with disability for 301 acute and chronic diseases and injuries in 188 countries, 1990-2013: a systematic analysis for the Global Burden of Disease Study 2013. Lancet. 2015;386:743–800. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Global, regional, and national burden of neurological disorders during 1990-2015: a systematic analysis for the Global Burden of Disease Study 2015. Lancet Neurol. 2017;16:877–897. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Dodick DW. CGRP ligand and receptor monoclonal antibodies for migraine prevention: Evidence review and clinical implications. Cephalalgia. 2019;39:445–458. [DOI] [PubMed] [Google Scholar]
- 4.Charles A Migraine: a brain state. Curr Opin Neurol. 2013;26:235–239. [DOI] [PubMed] [Google Scholar]
- 5.Migraine Charles A.. N Engl J Med. 2017;377:553–561. [DOI] [PubMed] [Google Scholar]
- 6.Blumenfeld AM, Bloudek LM, Becker WJ, Buse DC, Varon SF, Maglinte GA, Wilcox TK, Kawata AK, Lipton RB. Patterns of use and reasons for discontinuation of prophylactic medications for episodic migraine and chronic migraine: results from the second international burden of migraine study (IBMS-II). Headache. 2013;53:644–655. [DOI] [PubMed] [Google Scholar]
- 7.Bigal ME, Serrano D, Reed M, Lipton RB. Chronic migraine in the population: burden, diagnosis, and satisfaction with treatment. Neurology. 2008;71:559–566. [DOI] [PubMed] [Google Scholar]
- 8.Tepper SJ. History and Review of anti-Calcitonin Gene-Related Peptide (CGRP) Therapies: From Translational Research to Treatment. Headache. 2018;58 Suppl 3:238–275. [DOI] [PubMed] [Google Scholar]
- 9.Vollesen ALH, Amin FM, Ashina M. Targeted Pituitary Adenylate Cyclase-Activating Peptide Therapies for Migraine. Neurotherapeutics. 2018;15:371–376. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Akerman S, Goadsby PJ. Neuronal PAC1 receptors mediate delayed activation and sensitization of trigeminocervical neurons: Relevance to migraine. Sci Transl Med. 2015;7:308ra157. [DOI] [PubMed] [Google Scholar]
- 11.Waschek JA, Baca SM, Akerman S. PACAP and migraine headache: immunomodulation of neural circuits in autonomic ganglia and brain parenchyma. J Headache Pain. 2018;19:23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Vaudry D, Gonzalez BJ, Basille M, Yon L, Fournier A, Vaudry H. Pituitary Adenylate Cyclase-Activating Polypeptide and Its Receptors: From Structure to Functions. Pharmacological Reviews. 2000;52:269–324,. [PubMed] [Google Scholar]
- 13.Miyata A, Jiang L, Dahl RD, Kitada C, Kubo K, Fujino M, Minamino N, Arimura A. Isolation of a neuropeptide corresponding to the N-terminal 27 residues of the pituitary adenylate cyclase activating polypeptide with 38 residues (PACAP38). Biochem Biophys Res Commun. 1990;170:643–648. [DOI] [PubMed] [Google Scholar]
- 14.Arimura A, Somogyvári-Vigh A, Miyata A, Mizuno K, Coy DH, Kitada C. Tissue Distribution of PACAP as Determined by RIA: Highly Abundant in the Rat Brain and Testes. Endocrinology. 1991;129:2787–2789. [DOI] [PubMed] [Google Scholar]
- 15.Uddman R, Tajti J, Moller S, Sundler F, Edvinsson L. Neuronal messengers and peptide receptors in the human sphenopalatine and otic ganglia. Brain research. 1999;826:193–199. [DOI] [PubMed] [Google Scholar]
- 16.Ghatei MA, Takahashi K, Suzuki Y, Gardiner J, Jones PM, Bloom SR. Distribution, molecular characterization of pituitary adenylate cyclase-activating polypeptide and its precursor encoding messenger RNA in human and rat tissues. The Journal of endocrinology. 1993;136:159–166. [DOI] [PubMed] [Google Scholar]
- 17.Harmar AJ, Arimura A, Gozes I, Journot L, Laburthe M, Pisegna JR, Rawlings SR, Robberecht P, Said SI, Sreedharan SP, Wank SA, Waschek JA. International Union of Pharmacology. XVIII. Nomenclature of receptors for vasoactive intestinal peptide and pituitary adenylate cyclase-activating polypeptide. Pharmacol Rev. 1998;50:265–270. [PMC free article] [PubMed] [Google Scholar]
- 18.Harmar AJ, Fahrenkrug J, Gozes I, Laburthe M, May V, Pisegna JR, Vaudry D, Vaudry H, Waschek JA, Said SI. Pharmacology and functions of receptors for vasoactive intestinal peptide and pituitary adenylate cyclase-activating polypeptide: IUPHAR review 1. Br J Pharmacol. 2012;166:4–17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Birk S, Sitarz JT, Petersen KA, Oturai PS, Kruuse C, Fahrenkrug J, Olesen J. The effect of intravenous PACAP38 on cerebral hemodynamics in healthy volunteers. Regulatory peptides. 2007;140:185–191. [DOI] [PubMed] [Google Scholar]
- 20.Schytz HW, Birk S, Wienecke T, Kruuse C, Olesen J, Ashina M. PACAP38 induces migraine-like attacks in patients with migraine without aura. Brain : a journal of neurology. 2009;132:16–25. [DOI] [PubMed] [Google Scholar]
- 21.Amin FM, Asghar MS, Guo S, Hougaard A, Hansen AE, Schytz HW, van der Geest RJ, de Koning PJ, Larsson HB, Olesen J, Ashina M. Headache and prolonged dilatation of the middle meningeal artery by PACAP38 in healthy volunteers. Cephalalgia : an international journal of headache. 2012;32:140–149. [DOI] [PubMed] [Google Scholar]
- 22.Amin FM, Hougaard A, Schytz HW, Asghar MS, Lundholm E, Parvaiz AI, de Koning PJ, Andersen MR, Larsson HB, Fahrenkrug J, Olesen J, Ashina M. Investigation of the pathophysiological mechanisms of migraine attacks induced by pituitary adenylate cyclase-activating polypeptide-38. Brain : a journal of neurology. 2014;137:779–794. [DOI] [PubMed] [Google Scholar]
- 23.Amin FM, Hougaard A, Magon S, Asghar MS, Ahmad NN, Rostrup E, Sprenger T, Ashina M. Change in brain network connectivity during PACAP38-induced migraine attacks: A resting-state functional MRI study. Neurology. 2016;86:180–187. [DOI] [PubMed] [Google Scholar]
- 24.Guo S, Vollesen AL, Hansen YB, Frandsen E, Andersen MR, Amin FM, Fahrenkrug J, Olesen J, Ashina M. Part II: Biochemical changes after pituitary adenylate cyclase-activating polypeptide-38 infusion in migraine patients. Cephalalgia. 2017;37:136–147. [DOI] [PubMed] [Google Scholar]
- 25.Tuka B, Helyes Z, Markovics A, Bagoly T, Szolcsanyi J, Szabo N, Toth E, Kincses ZT, Vecsei L, Tajti J. Alterations in PACAP-38-like immunoreactivity in the plasma during ictal and interictal periods of migraine patients. Cephalalgia. 2013;33:1085–1095. [DOI] [PubMed] [Google Scholar]
- 26.Zagami AS, Edvinsson L, Goadsby PJ. Pituitary adenylate cyclase activating polypeptide and migraine. Annals of clinical and translational neurology. 2014;1:1036–1040. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Tuka B, Szabo N, Toth E, Kincses ZT, Pardutz A, Szok D, Kortesi T, Bagoly T, Helyes Z, Edvinsson L, Vecsei L, Tajti J. Release of PACAP-38 in episodic cluster headache patients - an exploratory study. J Headache Pain. 2016;17:69. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Markovics A, Kormos V, Gaszner B, Lashgarara A, Szoke E, Sandor K, Szabadfi K, Tuka B, Tajti J, Szolcsanyi J, Pinter E, Hashimoto H, Kun J, Reglodi D, Helyes Z. Pituitary adenylate cyclase-activating polypeptide plays a key role in nitroglycerol-induced trigeminovascular activation in mice. NeurobiolDis. 2012;45:633–644. [DOI] [PubMed] [Google Scholar]
- 29.Baun M, Pedersen MH, Olesen J, Jansen-Olesen I. Dural mast cell degranulation is a putative mechanism for headache induced by PACAP-38. Cephalalgia. 2012;32:337–345. [DOI] [PubMed] [Google Scholar]
- 30.Bhatt DK, Gupta S, Olesen J, Jansen-Olesen I. PACAP-38 infusion causes sustained vasodilation of the middle meningeal artery in the rat: possible involvement of mast cells. Cephalalgia. 2014;34:877–886. [DOI] [PubMed] [Google Scholar]
- 31.Jansen-Olesen I, Hougaard Pedersen S. PACAP and its receptors in cranial arteries and mast cells. J Headache Pain. 2018;19:16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Donahue RJ, Venkataraman A, Carroll FI, Meloni EG, Carlezon WA, Jr. Pituitary Adenylate Cyclase-Activating Polypeptide Disrupts Motivation, Social Interaction, and Attention in Male Sprague Dawley Rats. Biol Psychiatry. 2016;80:955–964. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Lehmann ML, Mustafa T, Eiden AM, Herkenham M, Eiden LE. PACAP-deficient mice show attenuated corticosterone secretion and fail to develop depressive behavior during chronic social defeat stress. Psychoneuroendocrinology. 2013;38:702–715. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Hashimoto H, Shintani N, Tanaka K, Mori W, Hirose M, Matsuda T, Sakaue M, Miyazaki J, Niwa H, Tashiro F, Yamamoto K, Koga K, Tomimoto S, Kunugi A, Suetake S, Baba A. Altered psychomotor behaviors in mice lacking pituitary adenylate cyclase-activating polypeptide (PACAP). Proc Natl Acad Sci U S A. 2001;98:13355–13360. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Lind MJ, Marraccini ME, Sheerin CM, Bountress K, Bacanu SA, Amstadter AB, Nugent NR. Association of Posttraumatic Stress Disorder With rs2267735 in the ADCYAP1R1 Gene: A Meta-Analysis. J Trauma Stress. 2017;30:389–398. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Ressler KJ, Mercer KB, Bradley B, Jovanovic T, Mahan A, Kerley K, Norrholm SD, Kilaru V, Smith AK, Myers AJ, Ramirez M, Engel A, Hammack SE, Toufexis D, Braas KM, Binder EB, May V. Post-traumatic stress disorder is associated with PACAP and the PAC1 receptor. Nature. 2011;470:492–497. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Shi N, Boado RJ, Pardridge WM. Antisense imaging of gene expression in the brain in vivo. ProcNatlAcadSciUSA. 2000;97:14709–14714. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Hoffmann J, Martins-Oliveira M, Akerman S, Supronsinchai W, Xu C, Goadsby PJ. PAC-1 receptor antibody modulates nociceptive trigeminal activity in rat. Cephalalgia : an international journal of headache. 2016;36:141. [Google Scholar]
- 39.Moldovan Loomis C, Dutzar B, Ojala EW, Hendrix L, Karasek C, Scalley-Kim M, Mulligan J, Fan P, Billgren J, Rubin V, Boshaw H, Kwon G, Marzolf S, Stewart E, Jurchen D, Pederson SM, Perrino McCulloch L, Baker B, Cady RK, Latham JA, Allison D, Garcia-Martinez LF. Pharmacologic Characterization of ALD1910, a Potent Humanized Monoclonal Antibody against the Pituitary Adenylate Cyclase-Activating Peptide. J Pharmacol Exp Ther. 2019;369:26–36. [DOI] [PubMed] [Google Scholar]
- 40.Borsodi A, Caló G, Chavkin C, Christie MJ, Civelli O, Cox BM, Devi LA, Evans CJ, Henderson G, Höllt V, Kieffer B, Kitchen I, Kreek MJ, Liu-Chen LY, Meunier JC, Portoghese PS, Shippenberg TS, Simon EJ, Toll L, Traynor JR, Ueda H, Wong YH. Opioid receptors: μ receptor. IUPHAR/BPS Guide to PHARMACOLOGY; In; 2019. [Google Scholar]
- 41.Bigal ME, Lipton RB. Excessive opioid use and the development of chronic migraine. Pain. 2009;142:179–182. [DOI] [PubMed] [Google Scholar]
- 42.Bigal ME, Lipton RB. Excessive opioid use and the development of chronic migraine. Pain. 2009;142:179–182. [DOI] [PubMed] [Google Scholar]
- 43.Headache Classification Committee of the International Headache Society (IHS) The International Classification of Headache Disorders, 3rd edition. Cephalalgia. 2018;38:1–211. [DOI] [PubMed] [Google Scholar]
- 44.Colas R, Munoz P, Temprano R, Gomez C, Pascual J. Chronic daily headache with analgesic overuse: epidemiology and impact on quality of life. Neurology. 2004;62:1338–1342. [DOI] [PubMed] [Google Scholar]
- 45.Reid MC, Engles-Horton LL, Weber MB, Kerns RD, Rogers EL, O’Connor PG. Use of Opioid Medications for Chronic Noncancer Pain Syndromes in Primary Care. Journal of General Internal Medicine. 2002;17:173–179. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Diener HC, Holle D, Solbach K, Gaul C. Medication-overuse headache: risk factors, pathophysiology and management. Nat Rev Neurol. 2016;12:575–583. [DOI] [PubMed] [Google Scholar]
- 47.Le Merrer J, Becker JA, Befort K, Kieffer BL. Reward processing by the opioid system in the brain. Physiol Rev. 2009;89:1379–1412. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Charles A, Pradhan AA. Delta-opioid receptors as targets for migraine therapy. Curr Opin Neurol. 2016;29:314–319. [DOI] [PubMed] [Google Scholar]
- 49.Xie JY, De Felice M, Kopruszinski CM, Eyde N, LaVigne J, Remeniuk B, Hernandez P, Yue X, Goshima N, Ossipov M, King T, Streicher JM, Navratilova E, Dodick D, Rosen H, Roberts E, Porreca F. Kappa opioid receptor antagonists: A possible new class of therapeutics for migraine prevention. Cephalalgia. 2017;37:780–794. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Pradhan AA, Befort K, Nozaki C, Gaveriaux-Ruff C, Kieffer BL. The delta opioid receptor: an evolving target for the treatment of brain disorders. Trends Pharmacol Sci. 2011;32:581–590. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Mansour A, Khachaturian H, Lewis ME, Akil H, Watson SJ. Anatomy of CNS opioid receptors. Trends Neurosci. 1988;11:308–314. [DOI] [PubMed] [Google Scholar]
- 52.Mennicken F, Zhang J, Hoffert C, Ahmad S, Beaudet A, O’Donnell D. Phylogenetic changes in the expression of delta opioid receptors in spinal cord and dorsal root ganglia. JComp Neurol. 2003;465:349–360. [DOI] [PubMed] [Google Scholar]
- 53.Bardoni R, Tawfik VL, Wang D, Francois A, Solorzano C, Shuster SA, Choudhury P, Betelli C, Cassidy C, Smith K, de Nooij JC, Mennicken F, O’Donnell D, Kieffer BL, Woodbury CJ, Basbaum AI, MacDermott AB, Scherrer G. Delta opioid receptors presynaptically regulate cutaneous mechanosensory neuron input to the spinal cord dorsal horn. Neuron. 2014;81:1312–1327. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Lutz PE, Kieffer BL. Opioid receptors: distinct roles in mood disorders. Trends Neurosci. 2013;36:195–206. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Negus SS, Gatch MB, Mello NK, Zhang X, Rice K. Behavioral effects of the delta-selective opioid agonist SNC80 and related compounds in rhesus monkeys. JPharmacolExpTher. 1998;286:362–375. [PubMed] [Google Scholar]
- 56.Stevenson GW, Folk JE, Rice KC, Negus SS. Interactions between delta and mu opioid agonists in assays of schedule-controlled responding, thermal nociception, drug self-administration, and drug versus food choice in rhesus monkeys: studies with SNC80 [(+)-4-[(alphaR)-alpha-((2S,5R)-4-allyl-2,5-dimethyl-1-piperazinyl)-3-methoxybenz yl]-N,N-diethylbenzamide] and heroin. JPharmacolExpTher. 2005;314:221–231. [DOI] [PubMed] [Google Scholar]
- 57.Do Carmo GP, Folk JE, Rice KC, Chartoff E, Carlezon WA Jr., Negus SS. The selective non-peptidic delta opioid agonist SNC80 does not facilitate intracranial self-stimulation in rats. EurJPharmacol. 2009;604:58–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Brandt MR, Furness MS, Rice KC, Fischer BD, Negus SS. Studies of tolerance and dependence with the delta-opioid agonist SNC80 in rhesus monkeys responding under a schedule of food presentation. JPharmacolExpTher. 2001;299:629–637. [PubMed] [Google Scholar]
- 59.May CN, Dashwood MR, Whitehead CJ, Mathias CJ. Differential cardiovascular and respiratory responses to central administration of selective opioid agonists in conscious rabbits: correlation with receptor distribution. Br J Pharmacol. 1989;98:903–913. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Gallantine EL, Meert TF. A comparison of the antinociceptive and adverse effects of the mu-opioid agonist morphine and the delta-opioid agonist SNC80. Basic ClinPharmacolToxicol. 2005;97:39–51. [DOI] [PubMed] [Google Scholar]
- 61.Pradhan AA, Smith ML, Kieffer BL, Evans CJ. Ligand-directed signalling within the opioid receptor family. BrJPharmacol. 2012;167:960–969. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Vicente-Sanchez A, Segura L, Pradhan AA. The delta opioid receptor tool box. Neuroscience. 2016;338:145–159. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Ashina M, Hansen JM, BO AD, Olesen J. Human models of migraine - short-term pain for long-term gain. Nat Rev Neurol. 2017. [DOI] [PubMed] [Google Scholar]
- 64.Dripps IJ, Boyer BT, Neubig RR, Rice KC, Traynor JR, Jutkiewicz EM. Role of signalling molecules in behaviours mediated by the delta opioid receptor agonist SNC80. Br J Pharmacol. 2018;175:891–901. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Moye LS, Tipton AF, Dripps I, Sheets Z, Crombie A, Violin JD, Pradhan AA. Delta opioid receptor agonists are effective for multiple types of headache disorders. Neuropharmacology. 2018;148:77–86. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Moye LS, Novack ML, Tipton AF, Krishnan H, Pandey SC, Pradhan AA. The development of a mouse model of mTBI-induced post-traumatic migraine, and identification of the delta opioid receptor as a novel therapeutic target. Cephalalgia. 2018:333102418777507. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Cahill CM, Holdridge SV, Morinville A. Trafficking of delta-opioid receptors and other G-protein-coupled receptors: implications for pain and analgesia. Trends PharmacolSci. 2007;28:23–31. [DOI] [PubMed] [Google Scholar]
- 68.Gendron L, Cahill CM, von Zastrow M, Schiller PW, Pineyro G. Molecular Pharmacology of delta-Opioid Receptors. Pharmacol Rev. 2016;68:631–700. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Pradhan A, Smith M, McGuire B, Evans C, Walwyn W. Chronic inflammatory injury results in increased coupling of delta opioid receptors to voltage-gated Ca2+ channels. Mol Pain. 2013;9:8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Rice FL, Xie JY, Albrecht PJ, Acker E, Bourgeois J, Navratilova E, Dodick DW, Porreca F. Anatomy and immunochemical characterization of the non-arterial peptidergic diffuse dural innervation of the rat and Rhesus monkey: Implications for functional regulation and treatment in migraine. Cephalalgia. 2017;37:1350–1372. [DOI] [PubMed] [Google Scholar]
- 71.Charles AC, Baca SM. Cortical spreading depression and migraine. Nat Rev Neurol. 2013;9:637–644. [DOI] [PubMed] [Google Scholar]
- 72.Ayata C, Jin H, Kudo C, Dalkara T, Moskowitz MA. Suppression of cortical spreading depression in migraine prophylaxis. AnnNeurol. 2006;59:652–661. [DOI] [PubMed] [Google Scholar]
- 73.Bogdanov VB, Multon S, Chauvel V, Bogdanova OV, Prodanov D, Makarchuk MY, Schoenen J. Migraine preventive drugs differentially affect cortical spreading depression in rat. NeurobiolDis. 2011;41:430–435. [DOI] [PubMed] [Google Scholar]
- 74.Pradhan AA, Smith ML, Zyuzin J, Charles A. delta-Opioid receptor agonists inhibit migraine-related hyperalgesia, aversive state and cortical spreading depression in mice. Br J Pharmacol. 2014;171:2375–2384. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Filliol D, Ghozland S, Chluba J, Martin M, Matthes HW, Simonin F, Befort K, Gaveriaux-Ruff C, Dierich A, LeMeur M, Valverde O, Maldonado R, Kieffer BL. Mice deficient for delta- and mu-opioid receptors exhibit opposing alterations of emotional responses. NatGenet. 2000;25:195–200. [DOI] [PubMed] [Google Scholar]
- 76.Broom DC, Jutkiewicz EM, Folk JE, Traynor JR, Rice KC, Woods JH. Convulsant activity of a non-peptidic delta-opioid receptor agonist is not required for its antidepressant-like effects in Sprague-Dawley rats. Psychopharmacology (Berl). 2002;164:42–48. [DOI] [PubMed] [Google Scholar]
- 77.Silberstein SD, Dodick D, Freitag F, Pearlman SH, Hahn SR, Scher AI, Lipton RB. Pharmacological approaches to managing migraine and associated comorbidities--clinical considerations for monotherapy versus polytherapy. Headache. 2007;47:585–599. [DOI] [PubMed] [Google Scholar]
- 78.Spahn V, Stein C. Targeting delta opioid receptors for pain treatment: drugs in phase I and II clinical development. Expert Opin Investig Drugs. 2017;26:155–160. [DOI] [PubMed] [Google Scholar]
- 79.Astra-Zeneca-Pharmaceuticals: A phase IIa, multicenter, randomized, double-blind, double-dummy, active and placebo controlled, parallel group study to assess the efficacy and safety of azd7268 in patients with major depressive disorder, clinical study report synopsis. https://clinicaltrialsgov/ct2/show/NCT01020799?term=AZD7268&rank=3. 2012.
- 80.Richards EM, Mathews DC, Luckenbaugh DA, Ionescu DF, Machado-Vieira R, Niciu MJ, Duncan WC, Nolan NM, Franco-Chaves JA, Hudzik T, Maciag C, Li S, Cross A, Smith MA, Zarate CA, Jr. A randomized, placebo-controlled pilot trial of the delta opioid receptor agonist AZD2327 in anxious depression. Psychopharmacology (Berl). 2016;233:1119–1130. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.DePaoli AM, Hurley KM, Yasada K, Reisine T, Bell G. Distribution of kappa opioid receptor mRNA in adult mouse brain: an in situ hybridization histochemistry study. Molecular and cellular neurosciences. 1994;5:327–335. [DOI] [PubMed] [Google Scholar]
- 82.Jamensky NT, Gianoulakis C. Content of dynorphins and kappa-opioid receptors in distinct brain regions of C57BL/6 and DBA/2 mice. Alcoholism, clinical and experimental research. 1997;21:1455–1464. [PubMed] [Google Scholar]
- 83.Peng J, Sarkar S, Chang SL. Opioid receptor expression in human brain and peripheral tissues using absolute quantitative real-time RT-PCR. Drug Alcohol Depend. 2012;124:223–228. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Pande AC, Pyke RE, Greiner M, Wideman GL, Benjamin R, Pierce MW. Analgesic efficacy of enadoline versus placebo or morphine in postsurgical pain. Clinical neuropharmacology. 1996;19:451–456. [DOI] [PubMed] [Google Scholar]
- 85.Pfeiffer A, Brantl V, Herz A, Emrich HM. Psychotomimesis mediated by kappa opiate receptors. Science. 1986;233:774–776. [DOI] [PubMed] [Google Scholar]
- 86.Eisenach JC, Carpenter R, Curry R. Analgesia from a peripherally active kappa-opioid receptor agonist in patients with chronic pancreatitis. Pain. 2003;101:89–95. [DOI] [PubMed] [Google Scholar]
- 87.Bruchas MR, Land BB, Chavkin C. The dynorphin/kappa opioid system as a modulator of stress-induced and pro-addictive behaviors. Brain research. 2010;1314:44–55. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Carlezon WA Jr., Krystal AD. Kappa-Opioid Antagonists for Psychiatric Disorders: From Bench to Clinical Trials. Depress Anxiety. 2016;33:895–906. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Maleki N, Becerra L, Borsook D. Migraine: maladaptive brain responses to stress. Headache. 2012;52 Suppl 2:102–106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Lowe SL, Wong CJ, Witcher J, Gonzales CR, Dickinson GL, Bell RL, Rorick-Kehn L, Weller M, Stoltz RR, Royalty J, Tauscher-Wisniewski S. Safety, tolerability, and pharmacokinetic evaluation of single- and multiple-ascending doses of a novel kappa opioid receptor antagonist LY2456302 and drug interaction with ethanol in healthy subjects. Journal of clinical pharmacology. 2014;54:968–978. [DOI] [PubMed] [Google Scholar]
- 91.Holland PR. Biology of Neuropeptides: Orexinergic Involvement in Primary Headache Disorders. Headache. 2017;57 Suppl 2:76–88. [DOI] [PubMed] [Google Scholar]
- 92.Noseda R, Borsook D, Burstein R. Neuropeptides and Neurotransmitters That Modulate Thalamo-Cortical Pathways Relevant to Migraine Headache. Headache. 2017;57 Suppl 2:97–111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Strother LC, Srikiatkhachorn A, Supronsinchai W. Targeted Orexin and Hypothalamic Neuropeptides for Migraine. Neurotherapeutics. 2018;15:377–390. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Gotter AL, Webber AL, Coleman PJ, Renger JJ, Winrow CJ. International Union of Basic and Clinical Pharmacology. LXXXVI. Orexin receptor function, nomenclature and pharmacology. Pharmacol Rev. 2012;64:389–420. [DOI] [PubMed] [Google Scholar]
- 95.Wong KK, Ng SY, Lee LT, Ng HK, Chow BK. Orexins and their receptors from fish to mammals: a comparative approach. Gen Comp Endocrinol. 2011;171:124–130. [DOI] [PubMed] [Google Scholar]
- 96.Mahler SV, Moorman DE, Smith RJ, James MH, Aston-Jones G. Motivational activation: a unifying hypothesis of orexin/hypocretin function. Nat Neurosci. 2014;17:1298–1303. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Sakurai T, Amemiya A, Ishii M, Matsuzaki I, Chemelli RM, Tanaka H, Williams SC, Richardson JA, Kozlowski GP, Wilson S, Arch JR, Buckingham RE, Haynes AC, Carr SA, Annan RS, McNulty DE, Liu WS, Terrett JA, Elshourbagy NA, Bergsma DJ, Yanagisawa M. Orexins and orexin receptors: a family of hypothalamic neuropeptides and G protein-coupled receptors that regulate feeding behavior. Cell. 1998;92:573–585. [DOI] [PubMed] [Google Scholar]
- 98.de Lecea L, Kilduff TS, Peyron C, Gao X, Foye PE, Danielson PE, Fukuhara C, Battenberg EL, Gautvik VT, Bartlett FS 2nd, Frankel WN, van den Pol AN, Bloom FE, Gautvik KM, Sutcliffe JG. The hypocretins: hypothalamus-specific peptides with neuroexcitatory activity. Proc Natl Acad Sci U S A. 1998;95:322–327. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Burdakov D, Gerasimenko O, Verkhratsky A. Physiological changes in glucose differentially modulate the excitability of hypothalamic melanin-concentrating hormone and orexin neurons in situ. J Neurosci. 2005;25:2429–2433. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Yamanaka A, Beuckmann CT, Willie JT, Hara J, Tsujino N, Mieda M, Tominaga M, Yagami K, Sugiyama F, Goto K, Yanagisawa M, Sakurai T. Hypothalamic orexin neurons regulate arousal according to energy balance in mice. Neuron. 2003;38:701–713. [DOI] [PubMed] [Google Scholar]
- 101.Zeitzer JM, Buckmaster CL, Parker KJ, Hauck CM, Lyons DM, Mignot E. Circadian and homeostatic regulation of hypocretin in a primate model: implications for the consolidation of wakefulness. J Neurosci. 2003;23:3555–3560. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Lee MG, Hassani OK, Jones BE. Discharge of identified orexin/hypocretin neurons across the sleep-waking cycle. J Neurosci. 2005;25:6716–6720. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Chemelli RM, Willie JT, Sinton CM, Elmquist JK, Scammell T, Lee C, Richardson JA, Williams SC, Xiong Y, Kisanuki Y, Fitch TE, Nakazato M, Hammer RE, Saper CB, Yanagisawa M. Narcolepsy in orexin knockout mice: molecular genetics of sleep regulation. Cell. 1999;98:437–451. [DOI] [PubMed] [Google Scholar]
- 104.Lin L, Faraco J, Li R, Kadotani H, Rogers W, Lin X, Qiu X, de Jong PJ, Nishino S, Mignot E. The sleep disorder canine narcolepsy is caused by a mutation in the hypocretin (orexin) receptor 2 gene. Cell. 1999;98:365–376. [DOI] [PubMed] [Google Scholar]
- 105.Peyron C, Faraco J, Rogers W, Ripley B, Overeem S, Charnay Y, Nevsimalova S, Aldrich M, Reynolds D, Albin R, Li R, Hungs M, Pedrazzoli M, Padigaru M, Kucherlapati M, Fan J, Maki R, Lammers GJ, Bouras C, Kucherlapati R, Nishino S, Mignot E. A mutation in a case of early onset narcolepsy and a generalized absence of hypocretin peptides in human narcoleptic brains. Nat Med. 2000;6:991–997. [DOI] [PubMed] [Google Scholar]
- 106.Dahmen N, Kasten M, Wieczorek S, Gencik M, Epplen JT, Ullrich B. Increased frequency of migraine in narcoleptic patients: a confirmatory study. Cephalalgia. 2003;23:14–19. [DOI] [PubMed] [Google Scholar]
- 107.Thomasy HE, Opp MR. Hypocretin Mediates Sleep and Wake Disturbances in a Mouse Model of Traumatic Brain Injury. J Neurotrauma. 2019;36:802–814. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Watanabe S, Kuwaki T, Yanagisawa M, Fukuda Y, Shimoyama M. Persistent pain and stress activate pain-inhibitory orexin pathways. Neuroreport. 2005;16:5–8. [DOI] [PubMed] [Google Scholar]
- 109.Bartsch T, Levy MJ, Knight YE, Goadsby PJ. Differential modulation of nociceptive dural input to [hypocretin] orexin A and B receptor activation in the posterior hypothalamic area. Pain. 2004;109:367–378. [DOI] [PubMed] [Google Scholar]
- 110.Holland PR, Akerman S, Goadsby PJ. Orexin 1 receptor activation attenuates neurogenic dural vasodilation in an animal model of trigeminovascular nociception. J Pharmacol Exp Ther. 2005;315:1380–1385. [DOI] [PubMed] [Google Scholar]
- 111.Holland PR, Akerman S, Goadsby PJ. Modulation of nociceptive dural input to the trigeminal nucleus caudalis via activation of the orexin 1 receptor in the rat. Eur J Neurosci. 2006;24:2825–2833. [DOI] [PubMed] [Google Scholar]
- 112.Barloese M, Jennum P, Lund N, Knudsen S, Gammeltoft S, Jensen R. Reduced CSF hypocretin-1 levels are associated with cluster headache. Cephalalgia. 2015;35:869–876. [DOI] [PubMed] [Google Scholar]
- 113.Sarchielli P, Rainero I, Coppola F, Rossi C, Mancini M, Pinessi L, Calabresi P. Involvement of corticotrophin-releasing factor and orexin-A in chronic migraine and medication-overuse headache: findings from cerebrospinal fluid. Cephalalgia. 2008;28:714–722. [DOI] [PubMed] [Google Scholar]
- 114.Cady RJ, Denson JE, Sullivan LQ, Durham PL. Dual orexin receptor antagonist 12 inhibits expression of proteins in neurons and glia implicated in peripheral and central sensitization. Neuroscience. 2014;269:79–92. [DOI] [PubMed] [Google Scholar]
- 115.Hoffmann J, Supronsinchai W, Akerman S, Andreou AP, Winrow CJ, Renger J, Hargreaves R, Goadsby PJ. Evidence for orexinergic mechanisms in migraine. Neurobiol Dis. 2015;74:137–143. [DOI] [PubMed] [Google Scholar]
- 116.Chabi A, Zhang Y, Jackson S, Cady R, Lines C, Herring WJ, Connor KM, Michelson D. Randomized controlled trial of the orexin receptor antagonist filorexant for migraine prophylaxis. Cephalalgia. 2015;35:379–388. [DOI] [PubMed] [Google Scholar]
- 117.Moncada S, Higgs EA. The discovery of nitric oxide and its role in vascular biology. Br J Pharmacol. 2006;147 Suppl 1:S193–201. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Pradhan AA, Bertels Z, Akerman S. Targeted Nitric Oxide Synthase Inhibitors for Migraine. Neurotherapeutics. 2018;15:391–401. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Olesen J The role of nitric oxide (NO) in migraine, tension-type headache and cluster headache. PharmacolTher. 2008;120:157–171. [DOI] [PubMed] [Google Scholar]
- 120.Bates EA, Nikai T, Brennan KC, Fu YH, Charles AC, Basbaum AI, Ptacek LJ, Ahn AH. Sumatriptan alleviates nitroglycerin-induced mechanical and thermal allodynia in mice. Cephalalgia. 2010;30:170–178. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Pradhan AA, Smith ML, McGuire B, Tarash I, Evans CJ, Charles A. Characterization of a novel model of chronic migraine. Pain. 2014;155:269–274. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Iversen HK, Olesen J, Tfelt-Hansen P. Intravenous nitroglycerin as an experimental model of vascular headache. Basic characteristics. Pain. 1989;38:17–24. [DOI] [PubMed] [Google Scholar]
- 123.Bryan NS, Bian K, Murad F. Discovery of the nitric oxide signaling pathway and targets for drug development. Front Biosci (Landmark Ed). 2009;14:1–18. [DOI] [PubMed] [Google Scholar]
- 124.Michel T, Feron O. Nitric oxide synthases: which, where, how, and why? J Clin Invest. 1997;100:2146–2152. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.He C, Chen F, Li B, Hu Z. Neurophysiology of HCN channels: from cellular functions to multiple regulations. Prog Neurobiol. 2014;112:1–23. [DOI] [PubMed] [Google Scholar]
- 126.Francis SH, Busch JL, Corbin JD, Sibley D. cGMP-dependent protein kinases and cGMP phosphodiesterases in nitric oxide and cGMP action. Pharmacol Rev. 2010;62:525–563. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Ben Aissa M, Tipton AF, Bertels Z, Gandhi R, Moye LS, Novack M, Bennett BM, Wang Y, Litosh V, Lee SH, Gaisina IN, Thatcher GR, Pradhan AA. Soluble guanylyl cyclase is a critical regulator of migraine-associated pain. Cephalalgia : an international journal of headache. 2018;38:1471–1484. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Taffi R, Vignini A, Lanciotti C, Luconi R, Nanetti L, Mazzanti L, Provinciali L, Silvestrini M, Bartolini M. Platelet membrane fluidity and peroxynitrite levels in migraine patients during headache-free periods. Cephalalgia. 2005;25:353–358. [DOI] [PubMed] [Google Scholar]
- 129.Finsterer J, Zarrouk-Mahjoub S. Headache in mitochondrial disorders. Clin Neurol Neurosurg. 2018;166:44–49. [DOI] [PubMed] [Google Scholar]
- 130.Schoenen J, Jacquy J, Lenaerts M. Effectiveness of high-dose riboflavin in migraine prophylaxis. A randomized controlled trial. Neurology. 1998;50:466–470. [DOI] [PubMed] [Google Scholar]
- 131.Sandor PS, Di Clemente L, Coppola G, Saenger U, Fumal A, Magis D, Seidel L, Agosti RM, Schoenen J. Efficacy of coenzyme Q10 in migraine prophylaxis: a randomized controlled trial. Neurology. 2005;64:713–715. [DOI] [PubMed] [Google Scholar]
- 132.Janes K, Neumann WL, Salvemini D. Anti-superoxide and anti-peroxynitrite strategies in pain suppression. Biochim Biophys Acta. 2012;1822:815–821. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Lassen LH, Ashina M, Christiansen I, Ulrich V, Grover R, Donaldson J, Olesen J. Nitric oxide synthase inhibition: a new principle in the treatment of migraine attacks. Cephalalgia. 1998;18:27–32. [DOI] [PubMed] [Google Scholar]
- 134.Ashina M, Lassen LH, Bendtsen L, Jensen R, Olesen J. Effect of inhibition of nitric oxide synthase on chronic tension-type headache: a randomised crossover trial. Lancet. 1999;353:287–289. [DOI] [PubMed] [Google Scholar]
- 135.Moncada S, Palmer RM, Higgs EA. Nitric oxide: physiology, pathophysiology, and pharmacology. Pharmacol Rev. 1991;43:109–142. [PubMed] [Google Scholar]
- 136.Reuter U, Bolay H, Jansen-Olesen I, Chiarugi A, Sanchez del Rio M, Letourneau R, Theoharides TC, Waeber C, Moskowitz MA. Delayed inflammation in rat meninges: implications for migraine pathophysiology. Brain : a journal of neurology. 2001;124:2490–2502. [DOI] [PubMed] [Google Scholar]
- 137.Reuter U, Chiarugi A, Bolay H, Moskowitz MA. Nuclear factor-kappaB as a molecular target for migraine therapy. Annals of neurology. 2002;51:507–516. [DOI] [PubMed] [Google Scholar]
- 138.Sarchielli P, Floridi A, Mancini ML, Rossi C, Coppola F, Baldi A, Pini LA, Calabresi P. NF-kappaB activity and iNOS expression in monocytes from internal jugular blood of migraine without aura patients during attacks. Cephalalgia : an international journal of headache. 2006;26:1071–1079. [DOI] [PubMed] [Google Scholar]
- 139.Palmer JE, Guillard FL, Laurijssens BE, Wentz AL, Dixon RM, Williams PM. A randomised, single-blind, placebo-controlled, adaptive clinical trial of GW274150, a selective iNOS inhibitor, in the treatment of acute migraine. Cephalalgia : an international journal of headache. 2009;29:124. [Google Scholar]
- 140.Hoivik HO, Laurijssens BE, Harnisch LO, Twomey CK, Dixon RM, Kirkham AJ, Williams PM, Wentz AL, Lunnon MW. Lack of efficacy of the selective iNOS inhibitor GW274150 in prophylaxis of migraine headache. Cephalalgia. 2010;30:1458–1467. [DOI] [PubMed] [Google Scholar]
- 141.Ramachandran R, Ploug KB, Hay-Schmidt A, Olesen J, Jansen-Olesen I, Gupta S. Nitric oxide synthase (NOS) in the trigeminal vascular system and other brain structures related to pain in rats. Neuroscience letters. 2010;484:192–196. [DOI] [PubMed] [Google Scholar]
- 142.Dieterle A, Fischer MJ, Link AS, Neuhuber WL, Messlinger K. Increase in CGRP- and nNOS-immunoreactive neurons in the rat trigeminal ganglion after infusion of an NO donor. Cephalalgia : an international journal of headache. 2011;31:31–42. [DOI] [PubMed] [Google Scholar]
- 143.Pardutz A, Krizbai I, Multon S, Vecsei L, Schoenen J. Systemic nitroglycerin increases nNOS levels in rat trigeminal nucleus caudalis. Neuroreport. 2000;11:3071–3075. [DOI] [PubMed] [Google Scholar]
- 144.Akerman S, Williamson DJ, Kaube H, Goadsby PJ. Nitric oxide synthase inhibitors can antagonize neurogenic and calcitonin gene-related peptide induced dilation of dural meningeal vessels. British journal of pharmacology. 2002;137:62–68. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Bhatt DK, Gupta S, Jansen-Olesen I, Andrews JS, Olesen J. NXN-188, a selective nNOS inhibitor and a 5-HT1B/1D receptor agonist, inhibits CGRP release in preclinical migraine models. Cephalalgia : an international journal of headache. 2013;33:87–100. [DOI] [PubMed] [Google Scholar]
- 146.De Felice M, Ossipov MH, Wang R, Dussor G, Lai J, Meng ID, Chichorro J, Andrews JS, Rakhit S, Maddaford S, Dodick D, Porreca F. Triptan-induced enhancement of neuronal nitric oxide synthase in trigeminal ganglion dural afferents underlies increased responsiveness to potential migraine triggers. Brain. 2010;133:2475–2488. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Medve RA, Lategan TW. A phase 2 multicenter, randomized, double-blind, parallel-group, placebo-controlled study of NXN-188 dihydrochloride in acute migraine without aura. Journal of Headache and Pain. 2010;11:S38–S38. [Google Scholar]
- 148.Medve RA, Andrews JS. Effects of fixed dose combination of nNOS inhibition and 5HT agonism on progrression of migraine with and without aura. Cephalalgia. 2009;29:126. [Google Scholar]
