- Review
- Open access
- Published:
Hallmarks of primary headache: part 1 – migraine
The Journal of Headache and Pain volume 25, Article number: 189 (2024)
Abstract
Background and aim
Migraine is a common disabling conditions which, globally, affects 15.2% of the population. It is the second cause of health loss in terms of years lived with disability, the first among women. Despite being so common, it is poorly recognised and too often undertreated. Specialty centres and neurologists with specific expertise on headache disorders have the knowledge to provide specific care: however, those who do not regularly treat patients with migraine will benefit from a synopsis on the most relevant and updated information about this condition. This paper presents a comprehensive view on the hallmarks of migraine, from genetics and diagnostic markers, up to treatments and societal impact, and reports the elements that identify migraine specific features.
Main results
The most relevant hallmark of migraine is that it has common and individual features together. Besides the known clinical manifestations, migraine presentation is heterogeneous with regard to frequency of attacks, presence of aura, response to therapy, associated comorbidities or other symptoms, which likely reflect migraine heterogeneous genetic and molecular basis. The amount of therapies for acute and for prophylactic treatment is really wide, and one of the difficulties is with finding the best treatment for the single patient. In addition to this, patients carry out different daily life activities, and might show lifestyle habits which are not entirely adequate to manage migraine day by day. Education will be more and more important as a strategy of brain health promotion, because this will enable reducing the amount of subjects needing specialty care, thus leaving it to those who require it in reason of refractory condition or presence of comorbidities.
Conclusions
Recognizing the hallmarks of migraine and the features of single patients enables prescribing specific pharmacological and non-pharmacological treatments. Medical research on headaches today particularly suffers from the syndrome of single-disease approach, but it is important to have a cross-sectional and joint vision with other close specialties, in order to treat our patients with a comprehensive approach that a heterogeneous condition like migraine requires.
Introduction
Migraine is a common neurological disorder with higher prevalence rates in the productive age, between the second and fourth decade, particularly among women. According to the 2021 estimates of the Global Burden of Disease study (GBD) [1], migraine prevalence is 15.2% globally (18.9% among women and 11.4% among men), a figure which is similar to that of a recent review [2], and is among the leading causes of health loss as measured by GBD’s years lived with disability (YLDs). Globally, it is ranked second after major depression for both sexes combined, as it accounts for 4.73% of all-cause YLDs (6.92% among people aged 15–49); among women, it is ranked first, as it accounts for 5.57% of all-cause YLDs (7.51% among women aged 15–49). The core symptom of migraine is a moderate or severe headache, which is often accompanied by a large amount of reversible symptoms, such as photophobia, phonophobia, cutaneous allodynia, nausea vertigo, and dizziness [3]. In around 30% of the cases, a set of reversible neurological symptoms lasting 5–60 min can occur before the onset, during, or in the absence of pain (migraine aura), the most common being visual ones [3]. In addition to this, migraine is associated to a wide variety of comorbidities, such as anxiety, depression and hypertension (pooled proportions comprised between 23 and 25%) [4], which makes migraine impact even higher.
Migraine impacts on a variety of daily life domains, to the point that almost all kind of activities are likely to be impacted [5], and the degree of such an impact is likely underestimated, due to the partial recognition of several mediators, in particular the effect of ageing process and of the use of medications, which can be considered as proxy indicators for migraine severity [6]. The objectification of migraine-related disability is of importance to monitor treatment effects and make management decisions, and to account for migraine impact on inter-ictal phases [6]. Such a situation, at the global level, is made event more complex by the fact that around 80% of the persons with migraine live in low and middle income countries, with limited access to care [7], and a poor recognition of the real burden of migraine that requires more data, and innovative approaches, in order to be recognized [6, 8].
Raising awareness on migraine is needed, as poor recognition of such condition has been reported both among patients and among healthcare professionals [9, 10]. This is critical as, given the large prevalence of migraine and the lack of specialized clinics in many low and medium income countries, most of migraine care has to be carried out at primary and secondary levels [7, 11,12,13], a situation that might also involve high-income countries in the near future, where better access to treatment for people with migraine is needed [14]. Specialty centres and neurologists with specific training on headache disorders have knowledge and skills to provide specific care, from diagnosis to treatment [15,16,17]. However, healthcare professionals and researchers who do not deal with migraine for clinical work-up and on a regular basis might benefit from a synopsis on the most relevant and updated information about migraine, from genetics and diagnostic markers, up to treatments and societal impact.
This paper is aimed to present the hallmarks of migraine in all these fields. It is organized into a set of sub-sections, and each of them is aimed to report the elements that identify migraine specific features, summarizing the most comprehensive and precise evidence possible for each topic.
Genetic basis
Migraine results from the interplay between genetic and environmental factors [18, 19]. Monogenic forms of migraine are caused by a pathogenic mutation in a single gene, and include Familial Hemiplegic Migraine (FHM) and migraine with aura associated with hereditary disorders such as CADASIL (Cerebral Autosomal Dominant Arteriopathy with Subcortical Infarcts and Leukoencephalopathy) [20,21,22,23,24]. Other forms of migraine are considered polygenic, where combined genetic risk factors (and not causal mutations) may contribute to the disease aetiology. Genome-Wide Association Studies (GWAS) have identified more than 180 susceptibility variants for migraine [25]. Each variant slightly increases the risk of migraine, and the combination of variants creates a "pro-migraine" condition [26]. Finally, recent studies have highlighted the importance of shared genetic factors between migraine and its major comorbidities, including depression and hypertension [27, 28].
FHM is an autosomal dominant disease, with four genes identified to date. FHM-1 is attributable to mutations of CACNA1A, encoding a subunit of a voltage-dependent calcium channel expressed in cerebral and cerebellar presynaptic terminals [20]. Mutations in ATP1A2, encoding a subunit of a Na + /K + transmembrane pump, are involved in FHM-2 [21], while mutations in SCN1A, encoding a subunit of a voltage-dependent neuronal sodium channel, lead to FHM-3 [22]. PRRT2, known as the major gene for paroxysmal kinesigenic dyskinesias, is considered responsible for type 4 FHM [23]. These genes all encode neuronal or astrocytic proteins expressed at synapses. Their mutations result in cortical hyperexcitability, which facilitates the onset of cortical spreading depression, the mechanism behind migraine aura [29]. However, these four genes account for only a minority (about 20%) of cases suspected of having FHM and referred for genetic assessment [30].
Hemiplegic Migraine (HM) can be familial or sporadic. In FHM, patients have at least one first- or second-degree relative with similar attacks. In sporadic forms, no relatives are affected. Both forms manifest with auras that include motor deficits associated with visual, sensory and/or speech/language symptoms [31]. HM is rare, with a prevalence estimated at 0.01% [32]. It typically manifests with an early onset at a mean age of 12–17 years. During typical attacks, muscle weakness varies from mild weakness of a limb to flaccid hemiplegia [33]. The frequency of attacks varies from more than one per week to a few throughout life, with an average of 3–4 per year. Their severity often decreases in adulthood. HM is characterized by great clinical variability in terms of aura symptoms (location, intensity, duration) and headache characteristics in a patient over their lifetime and among affected relatives within the same family. While patients with HM do not experience migraine attacks without aura more often than the general population, the prevalence of typical migraines with aura and migraines with brainstem aura is higher in patients with HM [34]. Depending on the involved genes, HM may be associated with inter-ictal manifestations such as epilepsy, cerebellar syndrome, or neurodevelopmental disorders. Permanent cerebellar ataxia is present in about 60% of cases of FHM-1 but is very rare in FHM-2 or FHM-3 [31]. Epilepsy is possible in all types of HM and affects 20–30% of FHM-2 cases [35].
Other forms of monogenic migraine with aura have been described in the context of genetic syndromes. Migraine with aura is frequently observed in patients with CADASIL, a hereditary small-vessel disease responsible for recurrent lacunar infarcts caused by mutations in the NOTCH3 gene [24]. Other hereditary small vessel diseases caused by mutations in COL4A1, COL4A2, or TREX1 also include migraine, often with aura [36, 37].
Mutations in the genes KCNK18, CSNK1D and ALPK1 have been reported in some families with migraine with aura, but further studies are needed to confirm the link between these mutations and the development of migraine with aura. KCNK18 encodes the TRESK channel, a potassium channel involved in membrane excitability. A frameshift mutation was found in a family presenting with visual auras in an autosomal dominant pattern [38]. CSNK1D, one of the "clock genes" involved in advanced sleep phase syndrome, has been reported in two large families presenting with this sleep disorder associated with migraine [39]. An autosomal dominant missense mutation in the ALPK1 gene has been identified in five families affected by ROSAH syndrome (Retinal dystrophy, Optic nerve oedema, Splenomegaly, Anhidrosis, and migraine Headache) [40].
Apart from those monogenic forms, more common migraine also has a heritability estimated between 35 to 60% [18, 19], but is attributed to a polygenic mechanism involving the interaction of multiple genetic variants, each conferring a low risk of developing the disease (Fig. 1). GWAS analyse millions of polymorphisms called Single Nucleotide Polymorphisms (SNPs) in large cohorts of patients and controls. Over the past 10 years, more than 180 SNPs have been identified as significantly associated with migraine [25, 41, 42]. The identified variants are located in non-coding regions or in affected genes whose functions are not all known. However, there appears to be higher expression of the genes involved in pro-migraine network in vascular and neuronal tissues, confirming that migraine is a polygenic neurovascular disorder. The latest GWAS study by Hautakangas in 2022 showed that three variants were relatively specific to migraine with aura, two were specific to migraine without aura, and nine were associated with both types of migraine [25]. Each of these SNPs explains only a small fraction of the genetic risk, and their sum does not account for the entire heritability of migraine.
The Polygenic Risk Score (PRS) assesses the overall genetic risk of migraine as the sum of all pro-migraine SNPs in an individual [26]. Studies show that the PRS is higher in familial cases, in migraine with aura compared to migraine without aura, and in forms with an earlier onset [43].
Finally, genetics has highlighted the importance of shared genetic factors between migraine and its major comorbidities. Several analyses of GWAS data have revealed a shared genetic susceptibility between migraine and psychiatric disorders [27], ischemic stroke [44], coronary artery disease [45], hypertension [28], sleep disorders [46], and also endometriosis [47].
Concluding remarks
In summary, there have been substantial advancements in the genetics of migraine in the past years. The investigation of monogenic migraines has underscored the strong links between migraine and neurovascular disorders. Genome-wide association studies have identified more than 180 variants mainly expressed in vascular and neuronal tissues, which are predisposing for migraine. In the future, advances in genetics might help to better quantify the genetic susceptibility to migraine and specific associated comorbidities for each patient, guide the choice of treatment, and inform prevention strategies.
Molecular pathways
The Trigeminovascular System (TVS) plays a pivotal role in migraine headache pathophysiology. Activation of this system results in the release of vasoactive neuromodulators, such as the Calcitonin Gene-Related Peptide (CGRP), amylin, the Pituitary Adenylate Cyclase Activating Polypeptide (PACAP), and Nitric Oxide (NO), amongst others. This section will discuss the pharmacology of these neuromodulators and the downstream signaling involved after the activation of the TVS.
CGRP
CGRP is recognized as a key mediator of migraine. Elevated levels of CGRP have been detected in the plasma, saliva, tears, and cerebrospinal fluid of migraine patients [48, 49]. Most importantly, intravenous administration of CGRP has been shown to induce migraine-like headaches in 57% of migraine patients [50].
In the TVS, CGRP has been shown to be expressed in C-fibers, whereas its receptor has been described to be present in Aδ-fibers and Schwann cells [51, 52]. The canonical CGRP receptor is a heterodimer comprised of the Calcitonin-Like Receptor (CLR), and the Receptor Activity-Modifying Protein 1 (RAMP1). The CLR is a G-Protein Coupled Receptor (GPCR), coupled to a Gs protein, therefore, binding of CGRP to its receptor activates the adenylate cyclase, leading to the accumulation of cyclic Adenosine Monophosphate (cAMP) [53].
Despite the clear role of CGRP in migraine, its specific site of action is still unclear. As shown in Fig. 2, the main site of action of CGRP is certainly the TVS [54]; however, the release of CGRP has several roles, including dilation of meningeal vessels, promotion of local neurogenic inflammation and dural mast cells degranulation, which, in turn, releases pro-inflammatory and pro-nociceptive molecules [55]. Additionally, mechanical allodynia and hyperalgesia could be mediated by shear stress and enhanced pulse waves in vessels, caused by mechanosensitive channels present in the trigeminal ganglion (TG) and surrounding satellite glial cells [56].
Role of CGRP and Amylin in the trigeminovascular system. Notes. Representation of the key elements of the trigeminovascular system, comprising meningeal blood vessels, local mast cells and trigeminal nerve fibres during a migraine headache attack. Binding of CGRP and amylin to their receptors results in dilation, and shear stress in the meningeal vessels. CGRP additionally activates mast cells, triggering the release of a plethora of pro-nociceptive compounds such as serotonin, histamine, leukotrienes, prostaglandins, ATP, and NO, further exciting the nociceptive fibres and promoting more CGRP release. ATP, adenosine triphosphate; CGRP, calcitonin gene-related peptide; NO, nitric oxide
Amylin
The neuroendocrine hormone amylin has been shown to be involved in pain signalling (Fig. 2). In preclinical models of migraine, amylin administration has been shown to increase neuronal activation of the TVS and orofacial allodynia [57, 58]. Moreover, infusion of pramlintide, an amylin analogue, induced migraine-like attacks in 41% of migraine patients [57]. The actions of amylin in the TVS are thought to be mediated via activation of the Amylin 1 (AMY1) receptor, with studies describing the expression of this receptor in C-fiber neurons in the TG [59]. CGRP can also bind to this receptor, with an affinity similar to amylin [53]; therefore, CGRP may exert its biological effects through various receptors and signalling pathways, while amylin exclusively acts on the AMY1 receptor.
As part of the calcitonin family of receptors, the AMY1 receptor is also a heterodimer comprised by the Calcitonin Receptor (CTR) and RAMP1. However, the pharmacology of this receptor is complex, since several CTR splice variants (i.e. isoforms) have been described. Of relevance for migraine, activation of the AMY1 receptor can result in either accumulation of cAMP, or in activation of Protein Kinase C (PKC) that leads to inhibition of large conductance Calcium-Activated Potassium (BKCa) channels [53, 60]. Determining the differential expression of these isoforms could shed light on which variants are the best candidates to target, and which may lead to unwanted side effects.
PACAP
PACAP belongs to a wider family of peptides that also includes the Vasoactive Intestinal Peptide (VIP) [61]. Two isoforms of PACAP have been reported, a 38 amino acid peptide (PACAP-38), and a cleaved 27 amino acid peptide (PACAP-27), with PACAP-38 being most prevalent form [62]. Infusion of these peptides has been shown to provoke migraine-like attacks in 58–72% of migraine patients [63, 64].
Studies have described the expression of PACAP and its receptors in the TVS [65]. PACAP acts via three GPCRs, the PAC1, VPAC1 and VPAC2 receptors [61]. While PACAP and VIP bind to VPAC1/2 receptors with similar affinity, PACAP exhibits a 100-fold higher activity than VIP at the PAC1 receptor. All three receptors lead to activation of the AC; nevertheless, alternative splicing of the PAC1 receptor gene results in different receptor variants [65]. For each variant, the PAC1 receptor can activate AC and PKC, with activation of AC being the predominant pathway [66]. Understanding the expression profile of these splice variants would enable developing novel drugs with higher efficacy, reducing off-target effects, leading to more personalized medicine.
Nitric oxide
The molecular basis of migraine is intricately linked to NO, a key blood flow regulator. As shown in Fig. 3, NO diffuses into cells and activates the soluble guanylate cyclase, increasing the levels of cyclic Guanosine Monophosphate (cGMP), leading to vasodilation, and further causing the release of neuropeptides [67]. Accordingly, glyceryl trinitrate, a precursor for NO, triggers migraine-like headaches in migraine patients [68]. Moreover, inhibiting NO synthases has been shown to effectively treat migraine attacks [69]. In contrast, substances such as reserpine, m-chlorophenylpiperazine, fenfluramine, and prostacyclin can trigger migraine attacks via activation of NO synthases [67].
Molecular pathways underlying the pathophysiology of migraine headache. Notes. Activation of the trigeminovascular system results in the release of CGRP, amylin and PACAP from c-fibers, and their release can be modulated via de activation of presynaptic receptors. Binding of these peptides to their receptors results in changes in vascular tone and nociceptive transmission. These responses can be regulated by modulating the different components of their signalling cascades, which has been studied using provocation models, in which potential trigger molecules are used to induce migraine attacks in humans. ATP, adenosine triphosphate; cAMP, cyclic adenosine monophosphate; cGMP, cyclic guanosine monophosphate; CGRP, calcitonin gene-related peptide; CGRPR, CGRP receptor; GTP, guanosine triphosphate; NO, nitric oxide; PACAP, Pituitary adenylate cyclase activating polypeptide; PDE3, phosphodiesterase 3; PDE5, phosphodiesterase 5; VIP, vasoactive intestinal peptide
Second messengers
The main neuromodulators described to be involved in the pathophysiology of migraine headache, bind to GPCRs that lead to the accumulation of cAMP or cGMP (Fig. 3). A strict regulation of the levels of these nucleotides is crucial for maintaining cellular homeostasis; accordingly, Phosphodiesterases (PDEs) hydrolyze cAMP and cGMP. Eleven families of phosphodiesterases have been described, each one with tissue- and nucleotide-specific profiles. Of relevance for migraine, PDE3 and PDE5 have been shown to be expressed in endothelial and vascular smooth muscle cells of cerebral arteries [70, 71], with preclinical studies also suggesting expression in the different components of the TVS [72]. Due to their role in vascular function, PDE inhibitors have been developed for cardiovascular disorders. Of interest, cilostazol and sildenafil were developed for PDE3 and PDE5, respectively [67]. Therefore, inhibition of these PDEs causes accumulation of their respective nucleotides. In accordance with this, cilostazol has been shown to induce migraine-like headaches in 73–86% of migraine patients [73, 74]. Similarly, infusion of sildenafil provokes migraine like headaches in 63–89% of migraine patients [75, 76].
Potassium channels
Accumulation of cAMP and cGMP leads to activation of protein kinase A and protein kinase G, respectively. These kinases modulate the activity of the ATP-sensitive potassium channels and the BKCa channels [77].
The ATP-sensitive potassium channels are composed of two subunits, an inward rectifier potassium (Kir) channel, and a Sulfonylurea Receptor (SUR) subunit [78]. Different isoforms exist of each subunit, all with specific properties and tissue distribution. In the TVS, Kir6.1, Kir6.2, SUR1 and SUR2B have been described. Interestingly, administration of NN414, a neuronal-specific Kir6.2/SUR1 channel opener did not provoke migraine-like headaches [79]. In contrast, levcromakalim, a selective Kir6.1/SUR2B channel opener expressed in cerebral vasculature [80], induced migraine-like headaches in 82–100% of migraine patients, with 59% of patients developing aura [81, 82], suggesting migraine-specific isoforms.
The BKCa channels are also formed by two subunits, α and β. Four different β subunits have been reported, with the β1 subunit described to be expressed throughout the TVS [60]. In accordance, infusion of the BKCa channel opener MaxiPost triggered migraine-like attacks in 95% of migraine patients [83]. Together with levcromakalim, MaxiPost is one of the most effective experimental migraine triggers, reinforcing the role of these channels in migraine pathophysiology.
Presynaptic modulation
Modulation of the TVS can also be achieved via the activation of presynaptic receptors present in the trigeminal sensory fibers. For example, the Serotonin (5-HT) 1D/1F receptors, are GPCRs coupled to an inhibitory G protein, therefore, upon activation, these receptors will inhibit the release of neuromodulators from the synaptic terminal, such as CGRP [84, 85]. In contrast, activation of the channels belonging to the Transient Receptor Potential (TRP) family, namely, the ankyrin 1 (TRPA1), vanilloid 1 and 4 (TRPV1 and TRPV4,), and melastatin 8 (TRPM8), results in vesicle release (e.g. CGRP) [86]. Similarly, studies have shown expression of the AMY1 receptor in C-fibers, suggesting than CGRP (and amylin) could activate this receptor in an autocrine manner, thus, facilitating trigeminovascular sensitization [53].
Concluding remarks
In summary, studies have shown that activation of the TVS results in the release of vasoactive neuromodulators like CGRP, amylin, PACAP and NO, which play a crucial role in migraine headache, as well as migraine chronification. Binding of these molecules to their receptors and their subsequent signalling cascades, results in changes in vascular tone and nociceptive transmission. Understanding these pathways will allow to not only develop novel targets, but to also optimise current therapeutic interventions for migraine.
Central Nervous System (CNS) and Peripheral Nervous System (PNS) implications
Migraine is a complex neurological disorder that causes ictal and interictal symptoms. Its manifestations include not only headache, but also hypersensitivity to certain stimuli, cranial autonomic symptoms, transient neurological symptoms, cognitive manifestations, homeostatic, and/or affective symptoms, which can present before, during, or after the headache [87]. The ictal and peri-ictal phase has been frequently divided into four possible sub-phases: premonitory symptoms, aura, the attack itself and the postdromal phase [88]. Current knowledge suggests that this classification is more academic than clinical, since many symptoms and phases overlap during the attacks, and some may not always be clinically evident [87, 88]. Regarding interictal manifestations, some patients may exhibit symptoms such as allodynia, sensory hypersensitivity, mood disorders or cognitive symptoms, among others [89]. Research has associated many of these features with CNS and PNS abnormalities, which will be summarized.
Regarding migraine episodes, premonitory symptoms may precede the onset of pain up to 48 h [90]. The most commonly reported symptoms include fatigue, altered mood, and hunger [91]. These symptoms have been associated with the activation of certain brain areas, with the hypothalamus, the dorsal pons, the limbic system, and the ventral tegmentum being the most frequently reported in imaging studies [92]. Other premonitory symptoms are typical, or even diagnostic, migraine symptoms such as nausea, vomiting, photophobia, phonophobia or osmophobia [90, 91, 93]. This overlap, along with the anatomic and functional interconnection of these regions with other areas more associated with the ictal phase, such as the brainstem and the thalamus, contribute to the network pathophysiology of migraine [94].
The second phase is aura; however, it is not consistently reported by all patients. Around 20–30% of patients report experiencing aura preceding or coinciding with migraine attacks [95]. These symptoms can vary but often include visual disturbances, such as seeing flashing lights, zigzag patterns, or temporary blind spots in the visual field. Some individuals may experience sensory symptoms like tingling or numbness in the hands, face, or mouth. In rare cases, people may have difficulty with speech or experience muscle weakness. Aura usually lasts from 5 to 60 min and can serve as a warning sign for the impending headache phase. The pathophysiology of aura has been linked with Cortical Spreading Depression (CSD) [96]. The combination of positive and negative visual, sensory, motor, and speech symptoms of aura seems to be explained by the wave of depolarization that occurs during CSD [95, 97]. It propagates across the cortex, usually in a postero-anterior direction, initiating a series of events at the cortical surface, leading neuronal and glial depolarization, the release of inflammatory mediators and neurotransmiters, and degranulation of the dural mast cells. This process results in the constriction and dilation of surface vessels housing trigeminal afferents, along with the direct depolarization of nociceptive afferents via the release of K + and other mediators into the extracellular space [98]. The net result is an increase in the spontaneous firing rate of both TG and TNC neurons.
During the ictal phase, both CNS and PNS structures are relevant. These include trigeminovascular system activation, and trigeminal nerve activation and autonomic nervous system involvement. Craniofacial nociceptive afferents originate from the TG and the dorsal root ganglia of cervical roots C1–C3. Their central projections terminate in the trigeminocervical complex, which includes the Trigeminal Subnucleus Caudalis (TNC) and the dorsal horn of the first cervical segments, representing the initial CNS relay in craniofacial nociceptive circuitry [99, 100]. TNC neurons extend projections to the Ventroposteromedial and posterior nuclei of the thalamus. Ventroposteromedial neurons primarily innervate the somatosensory cortex, while posterior neurons project more broadly to various sensory cortices, the insula, and association cortex regions. Additionally, TNC neurons establish connections with affective and motivational circuits via the nucleus tractus solitarius and parabrachial nucleus, which have diffuse projections to the hypothalamus, thalamic nuclei, amygdala, insular cortex, and frontal cortex [100]. Moreover, TNC neurons directly project to output structures involved in pain modulation and autonomic regulation, including the hypothalamus, periaqueductal gray, superior salivatory nucleus, and rostral ventromedial medulla [101]. These connections play essential roles in coordinating pain perception, emotional responses, and autonomic functions associated with craniofacial nociception. In summary, craniofacial afferents synapsing within the TNC project, either directly or indirectly, to regions associated with the sensory/discriminatory, salience/alerting, and affective/motivational dimensions of pain.
Migraine postdromes may include sensory, gastrointestinal, neuropsychiatric and general symptoms [98]. Interestingly, the most frequently reported postdromal symptoms are fatigue, concentration difficulties and mood changes [102, 103]. In addition, in the few studies that have specifically examined postdromes, no unique features have been observed [97]. This could suggest that the clinical presentation of postdromes may reflect the persistent activation of some of the brain structures that are implicated in the premonitory and/or ictal phases.
Migraine patients also exhibit structural and functional abnormalities between attacks, referred to as the interictal phase of migraine. The two main functional features are lack of habituation and sensitization [104]. The first refers to the inability of the migraine brain to decrease the amount of attention directed towards non-relevant stimuli, such as disturbing ambient noise [104]. The second is related to the decreased threshold that patients exhibit regarding the exaggerated perception of some sensory inputs, such as lights, sounds, smells, touch, or cranial movements [105]. Allodynia is an example of sensitization, where the response of nociceptive neurons is heightened [106]. Sensitization has been subdivided into peripheral and central sensitization, depending on the body regions where the patient perceives it and the neurological structures involved. Peripheral sensitization is limited to the cranial area and has been linked to the heightened response of the trigeminal ganglion afferents [107]. In contrast, central sensitization may be observed when patients exhibit symptoms beyond the head and has been associated with second, third or fourth-order neurons in the trigeminal nuclei, thalamus, or somatosensory cortex, respectively. Sensitization is not exclusive of migraine and can be observed in other headache disorders or pain syndromes [108]. In the case of migraine, its occurrence is more frequent in patients with higher frequency of attacks, being one of the changes linked to migraine chronification, by amplifying pain signalling and contributing to the persistence and intensity of migraine attacks [109].
Another aspect related with the interictal phase of migraine is the triggering of episodes. Some patients associate their episodes with weather changes, sleep deprivation or excess, stress, hormonal changes, alcohol intake or certain foods [110]. However, there is a notable variability both within attacks and among patients. The threshold for triggering attacks varies over time, and has also been associated to the frequency of episodes [111].
Concluding remarks
In summary, migraine is a disorder that involves both the central and peripheral nervous system, with the activation and inter-connection of multiple regions during the episodes and along with the chronification of the disorder.
Neuroimaging in migraine
Despite inconsistent findings in migraine neuroimaging studies and the fact that there are no reliable and robust neuroimaging biomarkers due to both not completely understood pathogenesis and clinical and neuroimaging heterogeneity [112], emerging advanced neuroimaging techniques are helping to elucidate common neuroanatomical substrates and functional abnormalities in migraine patients. In particular, we presented the neuroimaging hallmarks associated with Episodic Migraine (EM) with a special focus on migraine with aura, and Chronic Migraine (CM), highlighting key findings from recent research.
Burke et al. employed novel brain network mapping techniques to link neuroimaging findings to a common neuroanatomical substrate [113]. Their study demonstrated that regions of grey matter volume loss in migraine patients localize to a brain network involving the visual cortex, insula, and hypothalamus. These regions are strongly connected to the visual cortex V3/V3A, previously implicated in cortical spreading depression mechanisms [114, 115]. The specificity of these findings compared to chronic pain and neurological control groups suggests that these connectivity relationships may be present across all migraine subgroups, not just those with visual aura. Furthermore, Puledda et al. concluded that altered visual cortex excitability may be a neuroimaging hallmark of migraine [116]. In addition, the hypothalamus has been closely implicated in migraine as a hallmark of migraine attack initiation, along with the brainstem [117, 118]. These regions are thought to contribute to the autonomic and pain-processing abnormalities observed in migraine patients.
Functional Magnetic Resonance Imaging (fMRI) studies have consistently shown atypical brain responses to sensory stimuli, the absence of the normal habituating response between attacks, and atypical functional connectivity of sensory processing regions in migraine patients [119]. In particular, specific fMRI studies investigating thermal pain-induced brain activations revealed differential activation in migraine patients compared to healthy controls in several brain regions, including the temporal pole, parahippocampal gyrus, anterior cingulate cortex, lentiform nuclei, fusiform gyrus, subthalamic nucleus, hippocampus, middle cingulate cortex, somatosensory cortex, dorsolateral prefrontal cortex, secondary somatosensory cortex, precentral gyrus, superior temporal gyrus, and brainstem [120, 121]. Furthermore, fMRI studies suggest an imbalance of pain facilitation and inhibition as a neuroimaging hallmark of migraine [119, 122]. In other words, migraineurs exhibit stronger activation in pain-facilitating regions and hypoactivity in pain-inhibiting regions, indicating a disrupted pain modulation network.
Resting-state functional connectivity MRI studies have shown that migraine is associated with atypical connectivity in several brain regions and networks. Tessitore et al., Schwedt et al., and Chong et al. demonstrated that migraine patients exhibit altered connectivity in the somatosensory cortex, anterior and posterior insula, middle and anterior cingulate cortex, hippocampus, amygdala, parahippocampal gyrus, periaqueductal grey, nucleus cuneiformis, and hypothalamus [119, 123, 124]. Additionally, there are abnormalities in resting-state networks, including the default mode network, salience network, frontoparietal network, executive network, and sensorimotor network [119, 123, 124]. Moreover, the salience network, somatosensory network and default mode network showed altered connectivity during the attack versus outside of the attack [125]. In addition, Szabo et al. reported lower fractional anisotropy in the frontal lobe, indicating altered white matter integrity [126]. Moreover, MR Spectroscopy (MRS), a non-invasive method of investigating the biochemical composition of the brain, has been used to highlight various biological changes within the brain in patients with migraine. Phosphorous (31P)-MRS studies have implied abnormal energy metabolism and potential mitochondrial dysfunction may occur in patients with migraine. Imaging changes in migraine patients who do not experience aura are subtler, with studies varying widely in the literature [127].
Migraine with aura is likely mediated by widespread brain dysfunction in areas involving, but not limited to, visual cortex, somatosensory and insular cortex, and thalamus [128]. Gaist et al. observed a thicker visual cortex in patients with migraine with visual aura [129]. These structural changes may contribute to the visual disturbances and heightened cortical excitability seen in migraine with aura. Moreover, MRS studies have highlighted abnormal biochemical changes in the brains of migraine patients. Bridge et al. noted reduced GABA (γ-Aminobutyric Acid) levels in patients with migraine aura, suggesting reduced inhibition in the occipital cortex consistent with occipital hyperexcitability [130]. Additionally, there is a positive correlation between glutamate levels and BOLD (blood oxygenation level dependent) activation in the visual cortex during visual stimulation, indicating enhanced glutamate activation and abnormal excitation-inhibition coupling. Furthermore, Tedeschi et al. revealed that patients with migraine with aura have altered resting-state visual network connectivity compared to those with migraine without aura and healthy controls [131]. Datta et al. found a greater response to visual stimulation within the primary visual cortex in patients with migraine with aura, further differentiating between migraine subtypes [132]. In addition, Faragò et al. examined functional and structural brain differences between migraine patients with and without aura and concluded that these two subtypes of migraine should be handled separately in future studies [133]. Other fMRI imaging studies have also provided evidence of ictal and interictal alterations in functional connectivity within the visual and extrastriate cortex, somatosensory cortex, and executive cortical areas [112, 134, 135]. Perfusion abnormalities have been also observed in migraine patients, particularly those with aura. Lauritzen et al. first demonstrated objective alterations in regional Cerebral Blood Flow (CBF) during migraine aura using intra-arterial Xenon techniques [136]. Furthermore, Fu et al. reported higher CBF in regions such as the superior frontal gyrus and cerebellum, and lower CBF in the middle frontal gyrus and thalamus in patients with migraine with aura [137]. Increased vascular permeability, as a hallmark of inflammation, was mostly found in hemiplegic migraine, and was atypical in migraine with and without aura [138]. In general, it seems that cerebral hypoperfusion in one or more vascular territories occurs mainly unilaterally early during aura in the ictal phase and persists throughout the aura phase [128].
Silvestro et al. suggested that higher extrastriate brain changes may lead to the propagation of aura from simple visual symptoms to more complex phenotypes [139]. Pure visual aura compared to visual aura with other sensory or speech symptoms as well, may involve different functional reorganization of brain networks and additional mitochondrial dysfunction mediating more aura symptoms [128]. Several studies have underscored the notable distinctions between individuals experiencing pure visual aura and those who in addition experience somatosensory and dysphasic aura [139,140,141,142,143,144]. These findings hold significant potential for enhancing the accuracy of diagnosis, classification, and identification of biomarkers specific to distinct migraine with aura subtypes.
Patients with CM show increased resting-state functional connectivity of pain-processing areas, including the anterior cingulate cortex and the dorsal raphe nucleus, while resting-state functional connectivity between pain-processing areas and the hypothalamus is decreased [145]. In addition, Coppola et al. found decreased grey matter volume in patients with CM in regions such as the right cerebellum, left pallidum, amygdala, and orbitofrontal, temporal, and occipital cortex [146]. Patients with medication overuse headache show further reductions in grey matter volume, particularly in the bilateral orbitofrontal cortex and left middle occipital gyrus [147]. In addition, Li et al. demonstrated decreased neurovascular coupling in the orbitofrontal cortex [148].
Concluding remarks
In summary, atypical brain responses to sensory stimuli and altered functional connectivity in sensory processing regions and default mode network are consistent findings in migraine patients. Further neuroimaging research should focus on distinguishing between migraine subtypes to discover specific biomarkers and accurate predictive models, allowing therapeutic strategies tailored to each subtype.
Neurophysiological aspects of migraine
The complex neurophysiological mechanisms underlying migraine pathology have yet to be fully elucidated. The hallmark neurophysiological features of migraine (Fig. 4), identified through behavioural, electrophysiological, or neuroimaging investigations, are outlined in the following section; nonetheless, these features warrant further validation in the future.
Regarding the neural circuitry responsible for and encoding the subjective experience of pain, the migraine brain is characterized by alterations in perception and processing of noxious inputs; this dysfunction manifests in patients through several phenomena. (a) Temporal summation. Individuals with migraine often exhibit enhanced temporal summation, where quickly occurring repetitive noxious stimuli evoke progressively increasing levels of pain [149]. This indicates sensitization in pain processing at both the spinal cord and the supraspinal levels leading to exacerbation of headache intensity during attacks. (b) Conditioned pain modulation (CPM) dysfunction. Also known in animals as diffuse noxious inhibitory control, CPM is a surrogate measure of descending inhibitory system whereby a noxious stimulus applied to one part of the body inhibits pain perception in another part. Dysfunction of the CPM system has been observed in individuals with migraine [150]; it might reduce pain inhibition and enhances pain sensitivity. Impairment in pain modulation may contribute to the persistence and severity of migraine attacks. (c) Sensory sensitivity disturbances. Individuals with migraine often report a combination of altered multimodal sensorial perceptions during attacks—including changes in visual, auditory, olfactory, and tactile sensations, all part of the multidimensional neural activity related to pain [151]. Notably, multimodal sensitivity dysfunctions are not limited to the migraine attack itself; it can also manifest during the interictal period, indicating persistent alterations in multisensory pain-related information processing.
Imbalance in the autonomic nervous system is a neural process that can be indirectly evaluated through Heart Rate Variability (HRV) analysis. In individuals with migraine, HRV can be used to detect dysregulation of autonomic function. Migraine attacks are often associated with activation of the sympathetic nervous system leading to increased heart rate and blood pressure or parasympathetic dysfunction characterized by reduced vagal tone and impaired baroreflex sensitivity [152, 153]. One study showed that HRV parameters are associated with the effects of preventive medication [154]. However, further research is required to fully understand the complex interplay between HRV, autonomic regulation, and migraine.
Altered peripheral processes in migraine involve neuromuscular abnormalities in the neck and facial muscles. Altered electromyographic readings have been reported to be associated with migraine, reflecting the complex neuromuscular interactions involved in this condition. The abnormalities observed in migraine include various manifestations. (a) Increased muscle tension individuals with migraine often exhibit increased muscle stiffness in the neck and facial muscles [155]. (b) Muscle hyperactivity: elevated activity in muscles such as the trapezius, sternocleidomastoid, and temporalis indicates hyperactivity or spasms, contributing to or resulting from migraine pain [156]. (c) Abnormal muscle response: individuals with migraine may exhibit abnormal muscle responses to stress or stimuli that reflect altered neuromuscular control [157, 158]. Altered electromyographic activity in the neck and facial muscles is a major finding in migraine research. Further investigations are needed to verify whether muscle activity involvement it is the consequence of migraine recurrence or part of its pathophysiology.
Regarding the central processes, CSD is a fundamental event in the pathophysiology of migraine with aura. CSD involves a wave of neuronal depolarization followed by prolonged neuronal suppression that propagates across the cerebral cortex at a rate of 2–5 mm/min [29]. This phenomenon transiently disrupts normal neuronal function and is accompanied by changes in cerebral blood flow and metabolic activity. It is believed to underlie the aura phase of migraine and may, at least in the animal model, be induced or sustained by the activation of trigeminal and parasympathetic afferents through brainstem connections [159]. CSD likely contributes to the pathophysiology of migraine by promoting neuronal dysexcitability, disrupting ion homeostasis, and inducing neurovascular changes that predispose individuals to migraine attacks [160]. To what extent CSD is an electro-cortical phenomenon specific to migraine or rather is a general trigger for various diseases such as epilepsy, stroke, transient ischemic attack, remains to be determined.
Habituation deficit and central sensitization, are pivotal features of migraine cerebral responsivity. They are two processes that blend variably according to the different phases of the migraine cycle and its frequency. The increased central sensitization to sensory and nociceptive inputs leads to aberrant brain network function, even during resting-state conditions. Accumulating neurophysiological evidence supports this mechanism. (a) Habituation deficit. Habituation is a fundamental neurophysiological process where the response to a repeated stimulus decreases over time. In the context of migraine, a habituation deficit refers to a reduced ability to develop decreased responsiveness to repeated stimuli in the visual, somatosensory, or auditory cortex [161, 162]. Assessing habituation patterns can aid in diagnosing migraine. The presence of a habituation deficit may serve as a neurophysiological marker for distinguishing individuals with migraine between attacks from those with other headache disorders [163]. (b) Altered synaptic plasticity and metaplasticity. They refer to the activity-dependent modification of the strength or efficacy of communication between synapses, and their abnormalities are key characteristics of the migraine brain. Patients with migraine exhibit paradoxical and short-lived synaptic learning processes and altered cortico-subcortical metaplasticity in response to visual and sensorimotor learning techniques. [164, 165]. This state of altered basic neuronal mechanisms of learning and memory, probably genetically determined, can be observed across neural circuits and brain regions and plays a pivotal role in the pathophysiology of migraine. (c) Aberrant network function. Dysfunctional operation of hemodynamic and electro-cortical resting-state networks has been implicated in various neurological conditions, including migraine. Pain-related and salience networks may contribute to the sensory abnormalities in migraine [166,167,168]. The inability to cognitively and emotionally switch between the internalizing default-mode network and the externalizing executive and visual attentional networks by an aberrant salience network characterizes individuals with migraine [169,170,171].
Dysfunction of the sensorial thalamus and brainstem, which are involved in sensory/pain processing and modulation, may contribute to the initiation and propagation of migraine attacks. Key findings regarding such dysfunctions include the following. (a) Trigeminal spinal and caudal nuclei dysfunction. The trigeminovascular system, which is the major pain-signalling pathway of the visceral organ brain, originates in the brainstem, innervates the meninges and blood vessels of the brain and plays a crucial role in the migraine attack ignition. Dysfunction of the trigeminal sensory pathways—for example, abnormal processing of the brainstem nociceptive signals, as verified using blink and noxious-flexion reflexes, and pain-related cortical evoked potentials—may contribute to the pathophysiology of migraine [117, 118]. (b) Altered processing or modulation of somatosensory pathways. The brainstem and the thalamus contain nuclei involved in the modulation of spinal and cortical sensory processes, neuro-vascular coupling, and filtering of irrelevant peripheral inputs. Dysfunction of these sensory-modulatory circuits can lead to alterations in information processing and contribute to the development of migraine accompanied symptomology, such as photo-phono phobia and allodynia, and of the attack’s recurrence [172]. A recent study confirmed the dynamics of brainstem activation throughout the migraine cycle [173].
Concluding remarks
In summary, understanding the neurophysiology of migraine is crucial for developing effective diagnostic tools and treatment approaches targeting the mechanisms underlying this condition. Despite considerable advances having been made, much remains to be discovered regarding migraine; ongoing research continues to shed light on this complex condition.
Cardiovascular, cerebrovascular and psychiatric comorbidities of migraine
Cardiovascular diseases, psychiatric disorders, and migraine are among the ten neurological conditions with the highest age-standardized disability-adjusted life years over the past decades [1, 174]. In migraineurs, prevalence of cardiovascular diseases and psychiatric disorders revealed to be way greater than general population [4, 175]. A bidirectional association between migraine and these comorbidities has been hypothesized and documented, suggesting that they likely share a common biology despite some unclear details [176]. To obtain a complete clinical assessment of migraine subjects, comorbidities should be considered, and their assessment could orient treatment strategies and contribute to evaluate impact on subjects’ life [177].
Cerebrovascular and cardiovascular disorders comorbidity with migraine
In general, the risk factors for cerebrovascular events are age- and sex-dependent with traditional risk factors dominating in the age group of 44 years and older [178]. Hypertension is the most important risk factor in the age group above 44 years and migraine the most non-traditional risk factor in the age group below 35 years. The exact frequency of migrainous infarcts is unclear, especially since the differentiation between a migraine with aura associated infarct and a secondary aura triggered by a cerebral ischemia might be impossible in individual cases [179].
Population-based cross-sectional studies showed an increased risk of deep white matter lesions in patients with migraine depending on the frequency of migraine attacks, and also more often lesions in the cerebellar region and posterior circulation. Especially migraine with aura with more than 1 attack per month showed an increased risk [180,181,182]. There are some reports which showed a link between white matter lesions and the occurrence of right-left shunts [183]. Especially, permanent large right-left shunts are connected to migraine with aura [184]. Otherwise, it seems that the frequency of migraine does not have an effect on the frequency of white matter lesions [185].
Risk of haemorrhagic stroke may be increased in migraine [186], but not all studies showed such a correlation and no association with special forms of haemorrhagic strokes is known. Which factor increases the risk of white matter lesions is not completely elucidated; in a small number, there may be genetic reasons. In CADASIL, a hereditary small vessel disease, up to 54.5% of the patients have a history of migraine and 84% of them have a history of migraine with aura [187]. Otherwise right-left shunts could be one risk factor for ischemic strokes. The main risk factor for cerebral strokes is arterial hypertension and indeed there is evidence that women with migraine have a 21% increased risk of developing arterial hypertension during a follow-up period of more than 12 years (defined as values above 140 to 90 mmHg [188]).
As mentioned above, increased blood pressure is one main risk factor for stroke and the Genetic Epidemiology of Migraine study presented some evidence that migraine patients have a 27% increased risk of arterial hypertension [189]. Results from a summary of the literature conducted in 2020 regarding Relative Risk (95% CI) of ischemic stroke in migraine versus not migraine subjects are reported in Table 1 [190]: this study was the first study which showed an increased risk of death from myocardial infarction in migraine patients, especially for migraine with aura. A recent Danish population-based matched cohort study compared more than 50 000 patients with migraine with 510 000 matched patients without migraine and found that migraine patients had an increased risk for myocardial infarcts, venous thromboembolism, and arterial fibrillation but not for peripheral artery disease or heart failure [186]. Otherwise, there is no sign of increased coronary artery calcification as a surrogate marker of coronary arteriosclerosis [191].
A review including 2 meta-analyses, 11 controlled trials, and further studies saw evidence that regular intake of non-steroidal anti-inflammatory drugs over a longer period even with a frequency of only 5–14 days a month is correlated with an increased statistical risk of hypertension (RR = 1.21, 95% CI = 1.12–1.31) [192]. This was also true for paracetamol users. Triptans are migraine-specific drugs with a slight vasoconstriction due to the binding to the 5-HT 1B receptor and therefore contraindicated in patients with ischemic heart disease or ischemic stroke. A study from Denmark, again using the nationwide Danish patient register, found 429 612 patients; of the patients, who initiated the triptan therapy for the first time, 11 had a myocardial infarction, and 18 an ischemic stroke. All the patients had a high-risk cardiovascular profile, and the mean age was 60 years old [193].
Concerning prophylactic migraine medication, only a low number of studies are published. There is an animal study, which showed in a stroke model for mice a significant increased infarct volume in mice who were pre-treated with olcegepant or rimegepant. Both substances are small CGRP antagonists and thereby block the compensatory vessel relaxation mediated due to CGRP [194]. In a analyses of data from the veterans’ health administration there was no evidence that CGRP-pathway blocker induces arterial hypertension in patients with normal blood pressure. Otherwise, there was some evidence that these CGRP-pathway blockers can increase the blood pressure in patients with a history of arterial hypertension [195].
Psychiatric disorders comorbidity with migraine
Psychiatric disorders include a wide spectrum of clinical and behavioural manifestations [196] and knowledge on their epidemiology and aetiology importantly evolved in the last decades, leading to clarify that genetics and epigenetics play a key role in their onset and evolution across lifespan [197]. These disorders are also associated with other several factors, such as events occurred in early childhood (e.g., traumas and abuse), personality traits (e.g., neuroticism and introversion), environmental stressors and lifestyle, social conditions, as well as other disorders or conditions, including sleep disorders, substances addiction, and cognitive patterns [196, 198]. Most of these factors also have a key role in influencing migraine pattern [199, 200], which, in turn, also recognize a genetic predisposition [176]. The biological predisposition of migraine subjects to show psychiatric disorders is further evident from the analysis of personality traits and distinctive cognitive styles of such individuals [176]. Distinctive personality traits of migraine sufferers include apprehensiveness, avoidance, persistence, introversion, and neuroticism [201]. Migraineurs also differ from general population regarding emotional distress, locus of control, coping strategies, illness perceptions, and pain catastrophizing [197].
Anxiety disorders, bipolar spectrum disorders, depression, abuse (physical or emotional), and post-traumatic stress disorder showed high association with migraine, while also other psychiatric conditions seem to be associated with migraine, such as suicidal behaviour, psychosis, and panic disorder, other than sleep disorders [4, 199, 200, 202]. A stronger relationship was documented in subjects with CM, medication overuse, and migraine aura [199, 201, 203]. Several theories have been proposed to explain the relationship between psychiatric disorder and migraine. Theories are also referred to the predictive power of psychiatric disorders towards negative migraine evolution and complications, such as CM and medication overuse, although it was not demonstrated that improving the psychiatric disorder also improves migraine [199, 200]. In general, the association between psychiatric disorders and migraine is probably related to shared genetic basis, and to the influence of other biological and environmental factors able to impact both conditions. These factors encompass serotoninergic and dopaminergic pathways, involvement of the autonomic nervous system and the hypothalamus-pituitary axis, inflammation, hormones, central sensitivity/sensitisation of the sensory and emotional neural networks, and chronic stress, which, in turn, determines allostatic dysfunction and central sensitivity [176, 199, 200]. Theories have been also assessed through neuroimaging and considering subjects’ response to treatments indicated for both disorders. Neuroimaging showed that areas with a mood- and pain-modulating actions have a similar functional, structural, and connectivity alterations in subjects with migraine and psychiatric disorders often co-existent with migraine. These regions include anterior cingulate cortex, anterior insula, prefrontal cortex, hippocampus, amygdala, and periaqueductal gray [199, 200, 204].
Regarding psychiatric disorders more often associated with migraine and investigated in the literature, a summary of these theories, involved circuits and mechanisms, neuroimaging evidence, and shared treatment response is reported in Table 2 [199, 200, 204,205,206,207]. In terms of preventive therapy, the presence of psychiatric comorbidity needs careful consideration. Generally, a unified treatment approach for both conditions is advisable, if possible, and if the psychiatric disorder is mild [199, 200]. These considerations need to be performed by considering also disorder-specific treatment guidelines.
Concluding remarks
In summary, cardiovascular, cerebrovascular and psychiatric conditions constitute the most relevant comorbidities of migraine with relevance to the frequency of their co-occurrence. Shared mechanisms of action are hypothesized and common therapeutic pathways exist, which enable providing a valid treatment which might impact on the both migraine and of these comorbidities, thus helping to reduce the impact of these burdensome conditions.
The migraine cycle: prodromes, ictal phase, and postdromes
Migraine prodromal phase is the symptomatic non-painful period that precede up to 48 h the headache in migraine attack, according to ICHD-3 (International Classification of Headache Disorders, 3rd version) [33], and they are reported in a range from 30 to 80% of patients [208]. Most frequent prodromal symptoms reported in literature are mood changes, photophobia, fatigue, neck pain/stiffness, yawning, dizziness, difficulties in concentration, craving and nausea [111]. Some patients can even predict migraine attacks when experiencing prodromal symptoms, with a pretty accurate rate [209, 210]. Migraine triggers are elements capable of initiating a migraine attack, i.e. stress, odors, foods and alcohol, sleep deprivation, or fatigue are among the reported triggers in studies [111, 211]. Among them we can recognize endogenous triggers as stress, periods, sleep deprivation and exogenous triggers as foods or alcohol and visual stimulation [212].
Some doubts about the strict distinction between prodromes and trigger. Indeed, commonly reported trigger factors are not independent precipitators of migraine pain, but they could be misinterpreted as enhanced attention to some stimulation facilitated by premonitory symptoms [213]; moreover, triggers that overlap with corresponding special premonitory symptoms may be just a form of premonitory symptoms [214].
Ictal phase of migraine is composed by pain, associated symptoms and aura phenomenon. The typical pain of the ictal phase is widely accepted as result of TVS activation, that leads nociceptive information from the meninges to deep nucleus and to the cortex; the activation of the TVS take place peripherally where nociceptive terminals from the dura mater release vasoactive neuropeptides (i.e. calcitonin gene-related peptide CGRP, PACAP-38) [88]. The features of migraine pain are a clinical hallmark, typically described as moderate or severe, unilateral, and throbbing or pulsatile, aggravated by physical activity [33]. Aside from pain, patients usually report some other symptoms, such as nausea, vomiting, photophobia or phonophobia, reported as the most bothersome symptoms [33, 88, 215,216,217,218]. Up to 30% of patients with migraine presents aura phenomenon, defined as a fully reversible recurrent episodes of visual, sensory or other CNS symptoms that arise gradually, last less than 60 min, and are usually followed by typical migraine attack. Aura is thought to be generated by CSD a neurophysiological depression phenomenon discovered by Leao in 1944 followed by a prolonged phase of oligemia [33, 88, 95, 216, 219].
Postdrome phase is characterized from many symptoms as fatigue, neck stiffness, difficult in concentrating, increased appetite, and dull head pain, and they are reported by almost the 80% of the patients; this phase is poorly understood, still can also contributes in a significant way to the global disability of the migraine attack, indeed recently they are focused as new therapeutic outcome for acute treatment [93, 220,221,222,223,224].
Concluding remarks
In summary, the whole course of migraine is composed of distinct phases, all of which are associated to specific neurobiological changes, which are well defined and move beyond the features of migraine headache attack. Understanding these phases is of importance for clinicians and patients in order to timely treat migraine.
Pharmacological targets for acute treatment and their side effects: serotonin and CGRP
Serotonin
Considering the involvement of serotonergic neurotransmission in the pathophysiology of migraine, serotonin receptors emerged as pivotal targets for acute migraine treatment [225]. In total, there are seven distinct classes of 5-HT receptors [225]. Current acute migraine therapies primarily focus on modulating the 5-HT1 receptor family, which encompasses receptors distributed across both the vascular and neuronal components of the TVS [226]. Activation of 5-HT1 receptors induces a reduction in cAMP levels, with 5-HT1B receptors eliciting vasoconstriction and 5-HT1D/1F inhibiting neurotransmitter and neuropeptide release [84, 226]. Two main classes of acute migraine treatments that act upon 5-HT receptors are available: triptans, with highest affinity to the 5-HT1B/1D receptor, and ditans, with highest affinity to the 5-HT1F receptor (Table 3) [226].
The first triptan that was developed was sumatriptan [227], which entered the market during the early 1990s, followed by six subsequent triptans with different formulations and pharmacokinetic profiles. The agonistic triptan activity to the 5-HT1B receptor on blood vessels induces vasoconstriction, particularly accentuated in the cranial vasculature [228]. Initially, this was perceived as the primary mechanism for alleviating pain, aligning with the vascular theory of migraine pathophysiology [229, 230]. However, the simultaneous activation of the 5 HT1D receptor on trigeminal nerve fibers also inhibits the release of CGRP and protein extravasation [226]. Furthermore, agonism at the 5-HT1B, 5 HT1D, and 5-HT1F receptors within central nervous system structures can modulate nociception [226]. While sumatriptan, due to its low lipophilicity, predominantly acts on vascular structures and the peripheral nervous system without crossing the Blood–Brain Barrier (BBB), some second-generation triptans, being more lipophilic, may exert effects on the central nervous system as well [226]. Meta-analyses evaluating the efficacy of triptans in acute migraine treatment revealed rates of pain freedom at 2 h ranging from 18–50% [231,232,233]. Adverse events are typically mild, leading to discontinuation of therapy in less than 10% of patients [234]. These adverse events often encompass the so-called "triptan symptoms" or "triptan sensations", such as tingling or sensation of warmth. While the incidence of cardiovascular events following triptan administration is exceedingly low [235], the use of triptans is indeed associated with a higher risk of ischemic stroke and myocardial infarction with odds ratios of 3.2 and 3.3, respectively [193].
Therefore, due to their vasoconstrictive effects, triptans pose contraindications in individuals with cardiovascular risk factors [236]. Consequently, research has pivoted towards exploring alternative medications targeting different receptors, notably directing attention towards the 5-HT1F receptor [237]. The presence of 5-HT1F receptors within the brain and on trigeminal neurons, while absent in vascular smooth muscle cells, indicates that ditans exert their effects only through neuronal mechanisms such as inhibition of trigeminal nociception and neuropeptide release rather than through vascular mechanisms [84, 228]. Among clinically developed ditans, lasmiditan has gained approval in 50 mg and 100 mg tablet formulations for the acute management of migraine attacks [238, 239]. In Phase-III randomized Clinical Trials (RCTs), administration of lasmiditan 100 mg resulted in pain freedom within 2 h in approximately one-third of participants, significantly surpassing placebo rates [237]. The most reported adverse events linked to lasmiditan affect the central nervous system, with dizziness, somnolence, and fatigue among the most prevalent. Cardiovascular events occurred at similar rates with lasmiditan and placebo across studies, even in participants with cardiovascular risk factors, confirming the absence of clinically significant vasoconstrictive properties of this drug.
CGRP
Due to the key role of CGRP in trigeminal nociceptive transmission and migraine pathophysiology [240,241,242], drugs antagonizing the CGRPergic system were developed initially for the acute treatment of migraine [243]. Gepants, selective small-molecule CGRP receptor antagonists, were synthetized and proved to be effective in RCTs [244]. However, the first generation of gepants (e.g., telcagepant and olcegepant) did not reach the market due to hepatotoxic and pharmacokinetic limitations [243]. In the last years, a new generation of gepants has been developed with no demonstrable changes in serum transaminases [245]; and all demonstrated efficacy in RCTs [246], leading to their approval for clinical use. As shown in Table 3, three gepants are currently available for the acute treatment of migraine, two administered orally (rimegepant and ubrogepant) and one intranasally (zavegepant) [247].
The new gepants bind with high affinity to the canonical CGRP receptor [248], however, cross-reactivity has been reported with AMY1 receptors [248], one of the three amylin receptors expressed in trigeminal sensory neurons [249]. Nevertheless, the clinical implications of antagonizing both CGRP and AMY1 receptors remains unexplored. Based on the small molecular weight of the gepants, these drugs could cross the BBB [243], antagonizing both peripheral and central CGRP receptors. However, different studies indicate that gepants have a very limited ability to cross the BBB [245], and do not require to block central CGRP receptors to exert their antimigraine action (reviewed in [247]). Therefore, it remains to be determined whether these drugs can enter central brain areas lacking a BBB (circumventricular organs) to interact with CGRP receptors, and the clinical relevance, if any [247].
Meta-analyses of gepants in acute migraine treatment revealed rates of pain freedom at 2 h ranging from 23–58% [250, 251]. Gepants have shown a more favourable safety and tolerability profile than triptans, and the lack of vascular adverse events make them a promising alternative to patients with cardio- and/or cerebro-vascular disease or risk factors [252]. Adverse events are typically mild and rarely lead to discontinuation, with nausea, dizziness, somnolence and dry mouth most commonly reported [251]. Dysgeusia and nasal discomfort were also reported after zavegepant [252]. Importantly, there was no evidence that these drugs led to medication over-use headache [252].
Concluding remarks
In summary, advancements in understanding migraine pathophysiology and the discovery of new targets have significantly enhanced acute treatment options. Serotonergic drugs, such as triptans and ditans, target specific 5-HT receptors involved in migraine pathophysiology to provide effective pain relief. Triptans act primarily on 5-HT1B and 5-HT1D receptors, while ditans specifically target the 5-HT1F receptor and do not engage in vascular mechanisms. Additionally, the advent of gepants targeting the CGRP receptor marks a new era in acute migraine management. These innovations underscore the potential for medications that precisely address the underlying mechanisms of migraine, promising improved outcomes for patients.
Pharmacological targets for prophylaxis and their side effects: CGRP and PACAP
CGRP
CGRP is a 37 amino acid vasodilatory peptide that is widely expressed in the central and peripheral nervous system, especially in the trigeminal and dorsal root ganglia [253]. There are two CGRP receptors, calcitonin receptor-like receptor / receptor activity-modifying protein 1 complex (CLR/RAMP1 or CGRP receptor) and CGRP-responsive calcitonin receptor/RAMP1 complex (CTR/RAMP1 or AMY1 receptor). CGRP is a key player in the trigeminal system and plays a pivotal role in migraine pathophysiology. Human trigeminal ganglion contains the highest concentration of CGRP expressing neurons [254]. Following the release of CGRP from the trigeminovascular system, CGRP binds to the CGRP receptor and is involved in vasodilation, inflammation and nociceptive transmission [255]. Increased CGRP levels were reported in serum and saliva during induced or spontaneous migraine attacks [241, 255, 256]. Intravenous injection of CGRP led to experimentally induced migraine attacks in migraine with and without aura patients [50, 257].
Monoclonal Antibodies (mAbs) targeting CGRP or CGRP receptor are effective for migraine prophylaxis in both EM and CM patients [258]. Erenumab is the first anti-CGRP mAb that targets the CGRP receptor while galcanezumab, fremanezumab and eptinezumab target the CGRP ligand. The antibodies have a half-life of weeks and because of their peptide nature they cannot be used orally. Erenumab, galcanezumab and fremanezumab are administered subcutaneously once per month while eptinezumab is administered via intravenous route once every trimester. Gepants are small molecule CGRP receptor antagonists that have been developed for acute and preventive treatment of migraine. Rimegepant, atogepant and ubrogepant are second generation gepants whereas zavegepant is the only third generation gepants. Rimegepant was documented to have efficacy in both acute [259] and preventive treatment in EM patients [260] while atogepant is used for migraine prophylaxis. Atogepant is effective in the preventive treatment of both EM [261] and CM patients [262]. Zavegepant intranasal spray was effective in acute migraine in a phase III, randomized, double blind, placebo controlled trial [263].
In a systematic review and network meta-analysis of phase 3 RCTs that evaluated the efficacy of CGRP mAbs and gepants, fremanezumab, galcanezumab, erenumab and eptinezumab showed significant reductions in Monthly Migraine Days (MMD) compared to placebo [264]. Only the reduction for eptinezumab 300 mg was not statistically significant [14]. All CGRP mAbs showed significantly ≥ 50% responder rate compared to placebo [264]. In the same network meta-analysis, atogepant and rimegepant reduced MMD compared to placebo with atogepant 60 and 120 mg having the highest effect and rimegepant 75 mg every other day showing the lowest effect [264]. Atogepant at all doses showed ≥ 50% responder rate compared to placebo, however, due to the small number of patients, the results were not statistically significant [264]. Recently, a double-blind, double-dummy RCT in EM patients assessed whether galcanezumab 120 mg/day was superior to rimegepant 75 mg (q.o.d) in migraine prevention and galcanezumab was not superior to rimegepant regarding ≥ 50% reduction in MMD [265]. A meta-analysis found that good response to triptans, unilateral pain with autonomic symptoms and presence of vomiting during migraine were associated with good response to CGRP mAbs [266]. Conversely, absence of accompanying symptoms, obesity, interictal allodynia, psychiatric comorbidities, daily headaches and high number of unsuccessful previous migraine preventive treatments were predictors of a poor response to CGRP mAbs [266]. In clinical practice, up to one third of patients are defined as non-responders to CGRP mAbs [267]. Half of the patients not responding to CGRP mAbs within the first 3 months of treatment benefited from a longer treatment and were defined as late-responders [268]. Therefore, evaluation of treatment outcomes may be extended to 6 months [269].
PACAP
PACAP, a peptide discovered in the hypothalamus of sheep, belongs to the glucagon/secretin superfamily alongside peptides like vasoactive intestinal peptide (VIP), secretin, and others [270, 271]. It exists in two main forms: PACAP-38 and its truncated version, PACAP-27, with PACAP-38 being predominant [272]. Several neurological structures related to migraine express PACAP-38, including trigeminal ganglion, sphenopalatine ganglion, and trigeminal nucleus caudalis [273,274,275,276]. PACAP-38 activates three receptors – PAC1, VPAC1, and VPAC2 – leading to increased intracellular cAMP concentration [277]. PACAP receptors are found in various locations, including intracranial arteries, dura mater, trigeminal ganglion, and immune cells [278,279,280]. Both PACAP-27 and PACAP-38 induce migraine when administered intravenously, with responses to sumatriptan, a specific migraine treatment [63, 64, 281]. Plasma levels of PACAP-38 are elevated during migraine attacks and reduced between attacks [282, 283]. Mechanisms of PACAP-induced migraine include modulation of nociceptive transmission and mast cell degranulation, although infusion of PACAP38 does not increase blood markers for mast cell degranulation [284, 285]. Initially, it was thought that PAC1 receptor activation triggers migraine, leading to the development of AMG 301, a monoclonal antibody targeting the PAC1 receptor. However, a phase II trial showed no therapeutic benefit [286]. Two anti-PACAP monoclonal antibodies, Lu AG09222 and LY3451838, are in development. Lu AG09222 showed promise in preventing PACAP38-induced physiological responses and headache in a proof-of-mechanism study conducted in healthy volunteers [287]. A phase 2a trial of Lu AG09222 in migraine prevention demonstrated significant reduction in monthly migraine days compared to placebo (NCT05133323). In a phase II trial, LY3451838 was not more effective than placebo in preventing migraine, but the study only enrolled 19 individuals per treatment arm (NCT04498910).
Concluding remarks
In summary, the development of targeted treatments for migraine prophylaxis is advancing by focusing on specific mechanisms underlying the disorder. The identification of key pharmacological targets, such as CGRP and PACAP, has led to the creation of new medications that directly address these mechanisms. Monoclonal antibodies and gepants have demonstrated efficacy in reducing migraine frequency and severity by specifically targeting CGRP or its receptor and provide better treatment options for migraine compared to previous prophylactic treatments such as antidepressants and antiepileptics. Similarly, ongoing research into PACAP-related therapies aims to provide more targeted options for patients. These advancements highlight the potential for more effective and personalized migraine management, offering hope for better outcomes for individuals suffering from this debilitating condition.
Non-pharmacological targets: neuromodulation
The term “neuromodulation” is referred to any intervention able to safely interfere with the physiological nerve transmission within central or peripheral nervous system by inhibiting or potentiating it. Since the discovery of neurostimulation as a useful tool for pain relief, devices and approaches gradually evolved from invasive to non-invasive in order to guarantee effectiveness with lower risk of complications [288]. Non-invasive brain stimulation techniques can induce plastic changes that outlast the period of the stimulation not only in the cortex but also in brainstem and diencephalic structures. Although approved guidelines are often lacking due to heterogeneous stimulation targets and settings, different protocols have been used with encouraging results [289].
Non-invasive neuromodulation treatments for migraine are all carried out with electrical or magnetic stimulation, and applied to stop acute attacks or to prevent migraine recurrence [290]. The most known techniques include Transcranial Magnetic Stimulation (TMS), Vagus Nerve Stimulation (VNS), Supraorbital Nerve Stimulation (SNS), Occipital Nerve Stimulation (ONS) and transcranial Direct Current Stimulation (tDCS) or transcranial Alternate Current Stimulation (tACS).
In 2008 FDA approved the use of TMS for the treatment of depression and obsessive–compulsive disorders. This technique involves the generation of magnetic field delivered to the superficial layers of cerebral cortex through a coil placed against the scalp, with single pulse TMS or repetitive TMS stimulation protocols [291]. In migraineurs brain hyper-excitability or deficient intracortical inhibitory tone have often been reported and TMS therapeutic effects have been related to the increase of brain threshold and the inhibition of thalamic nociceptive neurons [292, 293]. Repetitive TMS can better influence brain plasticity beyond the stimulation period and is therefore used in prophylaxis whereas single pulse TMS has been tested in the acute phase of migraine [294]. Although limited due to methodological flaws and heterogeneity of studies, a recent overview confirmed that TMS may improve migraine severity and frequency [295]. Clinical trials also confirmed the feasibility and the efficacy of single pulse TMS for migraine associated with aura, supporting the FDA approval for its use either in acute and preventive setting [296].
Electric current based devices may be placed over the neck or several parts of the head, resulting in non-invasive stimulation of different available targets. In VNS, the Gammacore device is placed against both sides of the neck in order to electrically stimulate the vagus nerve, with the rationale of the treatment lying in the reduction of trigeminal nucleus caudalis glutamate concentration, decreased inflammation, oxidative stress and sympathetic activity and potentiation of opioids activity. Common protocols are based on stimulation between 30 to 60 min after a migraine attack [297,298,299].
The PRESTO study proposed VNS as a treatment for acute migraine, showing significant effectiveness if compared to the sham condition. The efficacy of the Gammacore was also demonstrated by an open-label study in patients with menstrual migraine whereas a meta-analysis published in 2023 reported the effectiveness of VNS in EM and encouraging results in the management of CM (in terms of monthly reduced headache days, pain-free rates, ≥ 50% responder rate, headache intensity and monthly acute medication reduction days) [300,301,302]. The VNS approach with Gammacore has been approved in US and all European countries for acute and preventive treatment of migraine in adults and adolescents.
SNS has been approved by FDA in 2014 for the treatment of EM and is based upon the transcutaneous stimulation of supraorbital and supratrochlear branches of the ophthalmic nerve (V1) through a bipolar self-adhesive electrode targets. Different studies showed promising results of supraorbital stimulation in the treatment of EM and CM, likely through the inhibition of trigeminal sensory pain routes. Protocols commonly are based on a 20-min daily session for prevention or impromptu for acute migraine attack [303,304,305].
Contrasting cortical hyperexcitability has been postulated as a mechanism underlying the effectiveness of ONS, in which two electrodes are placed over bilateral occipital nerves and may deliver currents in a common frequency range from 2 to 100 Hz [306]. ONS may reduce the monthly headache days and improve pain relief in patients with EM and CM but not significantly different when compared with pharmacological treatments [307].
tDCS may affect several cortical areas due to double stimulation: one electrode may deliver anodal facilitating and the other one a cathodal inhibiting current (typically 1–2 mA of amplitude). Therefore, the device may simultaneously or selectively affect the activity of different cortex areas depending on whether one of the electrodes is placed on an active or inactive site, respectively [308, 309]. Two recent systematic reviews and meta-analysis of randomized controlled trials reported that tDCS with primary motor and visual cortex activation (anodal) or visual cortex inhibition (cathodal) could reduce the number of migraine days per month [310, 311]. Other stimulation sites include the primary sensory cortex and dorsolateral prefrontal cortex. People undergoing stimulation occasionally reported burning sensations, dizziness, drowsiness, fatigue, headache, itching, nausea, pain, skin redness, and tingling with no statistical difference between active or sham.
In tACS there is no fixed anode/cathode position because current owns a sinusoidal waveform and the polarity alternates between the electrodes during stimulation. Therefore, this modulation seems to mimic in a more physiological way the endogenous brain oscillations if compared to tDCS [312]. In a double-blind, sham-controlled, randomized study, tACS over the visual cortex confirmed the potential to terminate migraine attacks although the high drop-out rate due to poor compliance seems to limit the treatment reliability [313].
Concluding remarks
In summary, contrasting evidence on the effectiveness of neuromodulation in migraine. Further randomized placebo-controlled studies with homogeneous protocols are needed in order to develop guidelines and clarify the long-term effectiveness of neuromodulation and its role in migraine management.
Non-pharmacological targets: cognitive behavioral therapy, relaxation and mindfulness
Pharmacological treatments for migraine are essential, however they often provide incomplete relief or cause side effects. Therefore, a multidisciplinary approach is crucial. Research indicates that behavioural treatments can reduce headache frequency and severity, enhance medication effectiveness, empower patient self-management, and decrease reliance on medication with potential side effects [314,315,316].
Cognitive Behavioural Therapy (CBT) has gained increasing attention as a complementary approach to headache management, offering potential benefits beyond traditional pharmacotherapy. Numerous clinical trials have demonstrated the efficacy of CBT in reducing the frequency, severity, and duration of primary headaches, with effects that are sustained over time, also compared with pharmacotherapy alone [317, 318]. This result was also confirmed in a sample of children and adolescents diagnosed with migraine, where the addition of CBT to medication led to a greater reduction in headache frequency and a reduction in migraine-related disability [319]. CBT typically involves several components, including education about headache triggers and mechanisms, relaxation techniques, cognitive restructuring, and behavioural interventions. Moreover, CBT has been associated with improvements in mood, quality of life, and medication adherence among headache sufferers. The mechanisms underlying the effectiveness of CBT in treating primary headaches are complex and multifactorial, encompassing cognitive interventions, behavioural techniques, and improvements in self-management skills. Cognitive interventions aim to identify and modify maladaptive beliefs and attitudes about pain. For example, many patients with chronic headaches develop anticipatory fear of pain, believing that their headaches are uncontrollable and devastating. CBT helps to restructure these thoughts, replacing them with more adaptive and realistic beliefs. This change reduces anticipatory anxiety and improves the patient’s coping capacity. Specifically, three main features and benefits of CBT applied to migraine include:
-
Enhanced Self-Efficacy: CBT helps individuals develop a stronger belief in their ability to manage and cope with challenges, including pain and associated symptoms. By fostering self-efficacy, CBT empowers patients to take proactive steps in their treatment and daily life, which can lead to greater overall well-being [320,321,322].
-
Promotion of Internal Locus of Control: CBT encourages individuals to recognize and harness their internal resources and capabilities in dealing with health issues. This shift from external factors to internal strengths empowers patients to feel more in control of their health outcomes and fosters a sense of empowerment and resilience [323, 324].
-
Reduction of Pain Catastrophizing: CBT addresses maladaptive thought patterns that exaggerate the perceived threat of pain. By restructuring these catastrophic thoughts, CBT helps individuals develop more balanced and realistic perspectives on pain, which can alleviate emotional distress and improve coping strategies [325, 326].
Behavioural techniques help to reduce muscle tension, a common trigger for headaches, and by reducing muscle tension, the frequency and intensity of headaches also decrease. Among these, biofeedback is a technique that teaches patients to control physiological functions through visual or auditory feedback. Patients can learn to reduce muscle tension or regulate breathing and heart rate, so preventing or mitigating headaches. Powers et al. demonstrated that behavioural treatments are effective for managing migraines in children and adolescents. Youths aged 10 to 17 with CM received amitriptyline and either 10 sessions of headache education or a tailored CBT program with biofeedback. After 12 months, 86% of those receiving CBT and medication had a 50% or greater reduction in headache days, and 88% showed significant reductions in disability, scoring below 20 on the Paediatric Migraine Disability Assessment [319]. Therefore, CBT combines cognitive and behavioural strategies to manage primary headaches, improving pain management, reducing muscle tension, and enhancing self-efficacy. It can be delivered individually, in groups, in person, or via telehealth, tailored to individual needs [327]. Integrating CBT with other treatments can enhance its effectiveness, though barriers like therapist availability and costs need addressing [328], but it might find solutions through digital health platforms [329].
Additionally, relaxation and meditation techniques, including progressive muscle relaxation, autogenic training and mindfulness, are effective methods for managing stress and pain by reducing stress responses and muscle tension [330, 331]. Progressive Muscle Relaxation (PMR) involves alternating between tensing and relaxing different muscle groups to achieve "extreme relaxation.” Patients focus on the contrasting sensations of tension and relaxation in each muscle group. Once the basic technique is mastered, patients can learn advanced skills such as recalling relaxation, using relaxation on command, and maintaining relaxation in muscles that are not actively used. Initially practiced in a calm environment, these skills can be applied in everyday situations. PMR has been successfully used to treat anxiety and phobias, and online applications now offer guided exercises specifically for migraine treatment. Studies have shown a reduction of up to 41% in migraine attacks per month and up to 43% in the number of migraine days per month following PMR training [330, 331]. Autogenic training (AT) is a relaxation technique that involves focusing on bodily perceptions (such as the weight or warmth of legs and arms, heartbeat, or breathing) and using self-suggestion. Typically performed while sitting or lying down, patients concentrate on specific body parts or vital functions and repeat phrases like “my right arm is heavy” or “my forehead is cool.” The goal is to induce a deep state of relaxation, triggering a “relaxation response” characterized by slower breathing, a reduced heart rate, changes in brain wave activity, increased body temperature, and reduced muscle tension. This response helps to decrease anxiety and stress [330]. Regular practice of autogenic training has been found effective in improving the quality of life for individuals with chronic pain, including those with migraines, by enhancing stress management and reducing headache frequency. Dobos et al. found positive correlations (Spearman’s rho: 0.541, p = 0.085) between the fMRI deactivation in the migraine-associated dorsal pons and the number of migraine attacks per month after a 16-week AT course [332].
Mindfulness is a form of meditation that focuses on being present with an intense awareness of sensations, without judgment. Practicing mindfulness involves various techniques, such as breathing methods, guided imagery, and other practices to relax the body and mind and help reduce stress [333]. Over the past 20 years, mindfulness-based interventions have gained popularity for treating various pain conditions, including migraines. From the mindfulness construct, different therapies have been developed under the umbrella term of Mindfulness-Based Interventions [334]. One of the most widely employed and researched is the Mindfulness-Based Stress Reduction (MBSR) protocol. Developed in 1979, MBSR helps individuals change their response to stressful thoughts and events, reducing emotional reactivity and improving cognitive appraisal [335, 336]. The standard MBSR program involves an 8-week course with weekly group sessions, totalling 26 h [337]. Another popular mindfulness-based intervention is the Acceptance and Commitment Therapy (ACT) [338]. This therapy uses metaphors, paradoxes, and mindfulness skills, along with various experiential practices. The aim of ACT is to improve patients’ quality of life by achieving emotional acceptance of physical pain and self-acceptance. ACT focuses on encouraging individuals to observe their experiences with openness and develop psychological flexibility [339, 340]. Recent Italian studies on mindfulness-based treatments combined with pharmacological treatment show greater improvements in several outcomes compared to pharmacological treatment alone, such as headache frequency, medication intake, headache impact, loss of productive time, disease cost, and better outcomes in disability and quality of life at 12 months from baseline. These results were found in a large sample of patients with CM and medication overuse headache (N = 177) [341]. In these patients, mindfulness promoted an increase in salience network connectivity and in cingulate cortical thickness, which are deemed to improve body-awareness of painful sensation and the cognitive processing of nociceptive information [342].
Concluding remarks
In summary, the effectiveness of PMR, autogenic training, and mindfulness has been demonstrated in many studies, and a recent one showed a superior efficacy of a combined treatment in which mindfulness was added to treatment as usual in patient with CM associated to Medication Overuse Headache (MOH). However, the majority of studies are uncontrolled ones and it is therefore necessary to increase our understanding of behavioural treatments efficacy by applying rigorous and homogeneous methodological criteria, and by including studies on CM during developmental age [343].
Non-pharmacological targets: diet and nutraceuticals
Overuse of medication for pain relief can lead to medication-overuse headaches and other adverse health effects. Therefore, strategies to reduce overuse and promote appropriate medication use are crucial. In this line, non-pharmacological and alternative therapies can be a suitable replacement for some patients, especially in children and adolescents [344,345,346,347,348,349]. In addition, recognizing and managing comorbid diseases and risk factors associated with headache disorders including migraine seems highly beneficial. Therefore, implementing a multi-modal management model that combines pharmacological and non-pharmacological treatments are encouraged. Here we present that dietary considerations and nutraceuticals, comprising vitamins, minerals, and herbal remedies can be considered as alternative approaches for managing migraine in adults, paediatrics, and pregnancy. While dietary factors and nutraceuticals offer potential benefits for migraine management, including migraine prophylaxis, it is essential to recognize the variability in study outcomes and potential adverse effects. Patients considering dietary alteration and the use of nutraceuticals should consult with healthcare professionals to assess their suitability and ensure appropriate monitoring.
Diet
Diet plays a significant role in managing migraine, and understanding the potential triggers can help patients make informed dietary choices. Some key points have been extracted from recent reviews [344,345,346, 350,351,352] to consider for dietary lifestyle modification:
-
Preventing hunger and fasting: Regular meals and avoiding long periods without eating can help stabilize blood sugar levels and prevent migraine attacks.
-
Sticking to frequent meals: Eating smaller, more frequent meals throughout the day can help maintain steady energy levels and prevent hunger-related migraine triggers.
-
Avoiding specific food items: Certain foods are common triggers for migraines, including alcohol, chocolate, caffeine-containing products (such as coffee, tea, cola), processed foods, seafood, fish, ice cream, foods containing nitrates (e.g., bacon, hot dog, ham, salami), foods containing tyramine (e.g., aged cheese, cheddar cheese, beans, smoked fish), citrus fruits, avocados, bananas, onions, and foods containing monosodium glutamate. However, it is important to note that triggers can vary from person to person, and keeping a food diary can help identify individual triggers. Patients can avoid or reintroduce potential trigger foods based on their own experiences [353].
-
Proper fluid intake and hydration: Staying hydrated is important for overall health and may help prevent dehydration-related migraines.
Ketogenic Diet (KD) is a recently suggested treatment for migraine patients. KD is characterized by a severe depletion of carbohydrates’ intake with a relative increase of fat assumption, leading metabolism to obtain energy from lipids through fatty acids oxidation and the formation of ketone bodies. In adults, KD can be effective, especially in patients requiring a hypocaloric diet [354, 355]. Although case reports have suggested a potential efficacy of KD even in childhood, compliance problems may raise due to the strict dietary regimen that is required [353].
Nutraceuticals
Various guidelines from organizations like the American Academy of Neurology/American Headache Society, Canadian Headache Society, and European Federation of Neurological Societies have addressed nutraceuticals with differing levels of detail, sometimes resulting in conflicting recommendations. A review [356] has summarized existing guidelines regarding the use of specific nutraceuticals, including riboflavin, coenzyme Q10, magnesium, butterbur, feverfew, and omega-3 polyunsaturated fatty acids.
-
Riboflavin, a member of the vitamin B (B2) family, riboflavin acts as a cofactor for flavoprotein enzyme function in the electron transport chain of the Kreb’s cycle, in addition to contributing to membrane stabilization and the maintenance of energy-related cellular functions. Schonen et al. [357] conducted a RCT showing significantly reduced attack frequency in adult patients treated with riboflavin compared to placebo, with a higher responder rate (56% vs. 19%).
-
Coenzyme Q10 (CoQ10) serves as an essential cofactor in the electron transport chain, safeguarding against mitochondrial collapse by maintaining proper energy output. While Sandor et al. [358] showed a significantly higher reduction in migraine attacks with CoQ10 than placebo, other RCTs did not reach the same positive results [359].
-
Magnesium is thought to play a pivotal role in establishing a threshold for migraine attacks through various pathophysiological mechanisms, including neuroinflammatory blockade and calcium channel blocking effects, among others. A review [360] of oral magnesium supplementation in migraine prevention suggests that low magnesium levels are associated with migraines, but due to limited evidence, increasing dietary magnesium intake may be advisable.
-
Butterbur, scientifically known as Petasites hybridus, has proved useful in migraine prevention [361]. However, there have been reports of hepatotoxicity [362] associated with certain butterbur formulations, leading to regulatory actions in different countries.
-
Feverfew, scientifically known as Tanacetum parthenium, has been investigated in several studies which showed contrasting results. A Cochrane Review [363] concluded that the evidence for feverfew’s efficacy in preventing migraines is mixed.
-
Omega-3 polyunsaturated fatty acids (OPFAs). While an early RCT involving 196 migraine patients found no significant difference in the mean number of attacks between those taking OPFAs and those on placebo over a 16-week period [364], there are two more recent RCTs suggesting OPFA efficacy in migraine prophylaxis [365, 366].
-
Palmitoylethanolamide (PEA) and Gingkolide B. PEA is an endogenous fatty acid amide widely distributed in different tissues, including nervous tissues. The anti-inflammatory effects of PEA seem to be mainly related to its ability to modulate mast cell activation and degranulation. In a pilot study, involving adult patients with migraine with aura, administration of PEA resulted in a significant reduction in the frequency and intensity of migraine attacks [367]. An open-label study in paediatric patients showed that after 3 months of treatment headache frequency was reduced by > 50% in 63.9% of patients [368]. Gingkolide B is an herbal constituent extract from Ginkgo biloba tree leaves. In a prospective trial [369] involving young patients with migraine without aura, a combination therapy of gingkolide B, CoQ10, vitamin B2, and magnesium significantly reduced the number of monthly migraine attacks after three months of treatment.
Nutraceuticals in women during pregnancy
Due to the limited evidence supporting the use of medications during pregnancy, alternative treatment options are often considered. There are common misconceptions that all nutraceuticals are safe during pregnancy, which is not always true for those commonly used in non-pregnant migraine patients [370]. Magnesium has multiple indications during pregnancy, such as relieving muscle cramps, constipation, and pre-eclampsia. According to a Cochrane review [371], there was no significant difference in perinatal or postnatal mortality, small-for-gestational-age infants, or pre-eclampsia. Although the optimal dose of riboflavin for migraine prevention is 400 mg daily, during pregnancy, riboflavin is classified as category A within the recommended daily allowance of 1.4 mg per day and category C if intake exceeds this dosage [372]. CoQ10 is considered safe during pregnancy and has been shown to lower the risk of pre-eclampsia in women who are at risk for the condition [373]. Feverfew can induce uterine contractions and should not be recommended during pregnancy [374]. Due to its potential to cause congenital malformations, also butterbur should not be recommended during pregnancy [375].
Concluding remarks
In summary, nutraceuticals can be considered for migraine prevention in patients who prioritize efficacy, speed of onset, and absence of side effects when choosing a preventive treatment. Since there are no head-to-head trials comparing nutraceuticals to pharmacological agents for migraine prevention, patients should be counselled about the unknown relative efficacy of nutraceuticals compared to conventional medications. However, since most nutraceutical trials show low rates of adverse effects, patients concerned about side effects may consider consulting their physicians and trying nutraceuticals before pharmacological agents. The very low prevalence of serious adverse events makes nutraceuticals particularly appropriate for migraine prophylaxis in children and adolescents. However, when prescribing nutraceuticals, the clinician should be aware that RCTs supporting their efficacy in the paediatric populations are currently lacking [376].
Non-pharmacological targets: exercise and physical therapy
Still an important proportion of patients do not achieve sufficient improvement with preventive medication: indeed, more than 50% of migraine patients search also for non-pharmacological options, such as physiotherapy [377]. Due to the role of the trigeminocervical complex, problems such as neck pain and musculoskeletal impairments may be comorbid and have an influence on migraine characteristics [378, 379]. Therefore, a proper clinical examination and an assessment of pain characteristics are needed to understand the possible role of physiotherapy in migraine patients.
Physiotherapy includes exercises, manual therapy (MT), and educational interventions. A recent systematic review conducted by Onan et al., found that occipital transcutaneous electrical stimulation, acupressure, osteopathic manual therapy, soft tissue mobilization and occipital transcutaneous electrical stimulation complemented with home exercises, facial proprioceptive neuromuscular facilitation and connective tissue massage, aerobic exercise, hydrotherapy approaches significantly improves headache intensity, frequency, duration, and quality-of-life. In the meta-analysis results, MT combined with medication treatment significantly improved headache intensity, and MT or aerobic exercise combined with medication treatment significantly improved the number of headache days per month [378].
Exercise
Exercise is widely used in the management of migraine patients, however different exercise modalities, and different dosages and parameters exist. According to the results of a systematic review and meta-analysis that included 21 studies mostly carried out on episodic migraine, on aerobic exercise versus strengthening exercises, strengthening exercises were found to have the highest effectiveness in improving migraine symptoms, followed by high-intensity aerobic exercise [380]. However, it is important to consider that only three of the included studies specifically focused on strengthening exercises, with a smaller participant pool compared to studies on aerobic exercise [381,382,383], and the last and largest of these studies targeting vestibular migraine [383] rather than general headache symptoms, which limits the possibility to generalize the effectiveness of strengthening exercise in migraine. A recently published guideline indicated evidence-based parameters for different exercise modalities [384] (Table 4).
Another important issue that should be emphasized is that, according to ICHD-3 criteria, migraine can be triggered by physical activity (PA) [33]. It is stated that migraine may be triggered by PA and exercise due to dysfunction of the hypocretin neuropeptide produced by the hypothalamus [385], by lactate metabolism [386], and CGRP released during exercise [387]. Indeed, exercising during a migraine attack (ictal phase), may be a worsening factor, thus requiring modification of the exercise in a gradual and individualized exposure [384]. On the other hand, regular exercise can help patients for the management of migraine symptoms, and provides functional and psychological benefits [388]. The mechanisms behind the improvement, are increased in beta-endorphin levels in plasma after regular exercise, and the rearrangement of brain-derived neurotrophic factor levels [389] and endocannabinoid levels [390, 391]. Amin et al. stated that the trigger threshold of migraine may change in migraine patients who exercise regularly. Thus exercise may provide a prophylactic effect, if performed in the right phase of the migraine cycle [390]. Its efficacy is confirmed by the fact that several societies include exercise as part of the therapeutic option for migraine management, such as the French Headache Society [392], the Danish Headache Society [393], and the American Headache Society [394]. A recent Delphi study, highlighted how the exercise prescriptions should be individualized for every migraine patient, considering the just mentioned different exercise modalities, based on the patients’ preferences, psychological aspects, level of PA, and possible adverse effects [395]. Indeed, the effect of exercise (acting as a migraine trigger or having a therapeutic effect) depends on migraine frequency (chronic or episodic), the type, the dosage, the duration, and the intensity of the exercise itself, but most importantly on the phase in which the exercise is performed. However, no specific exercise parameters emerge from these recommendations, with many studies referring to episodic migraine patients, representing a strong limitation when trying to apply in clinical practice exercises for migraine patients, and the role of exercise is still open to interpretation.
Manual therapy
MT is a commonly used non-pharmacological treatment, including soft tissue and articulatory techniques [396]. In a systematic review and meta-analysis of 6 studies on spinal manipulative therapy, with high heterogeneity, spinal manipulative therapy was shown to significantly reduce migraine days and headache intensity [397]. Results from a systematic review suggest that MT may be as effective as propranolol and topiramate for migraine [398]. Recent research confirmed that MT may be useful to decrease the frequency and intensity of migraine attacks [398, 399]. The mechanisms beyond MT interventions are based on the neurophysiological processes at both spinal and supraspinal level happening as consequences of the applied mechanical force, producing nociception modulation [400]. It has been reported that a combination of soft tissue and articulatory techniques yields larger improvements on headache intensity than the two approaches alone [401]. The main issues regarding MT include the heterogeneity of techniques, the number of sessions, and the duration of each session.
Neck pain (NP) and musculoskeletal impairment (MI) in the neck region are highly prevalent in migraine patients [402], and thus they should be properly assessed with a physical examination, to design a tailored treatment plan. The presence of NP in migraine patients is associated with worse headache characteristics, more severe MI, higher psychological burden, and enhanced signs and symptoms of sensitization [403]. A systematic review concluded that 4 out of 20 different assessment procedures (range of cervical motion, flexion-rotation test, pressure pain thresholds, and forward head posture) enable distinguishing migraine patients from controls. Manual joint testing and myofascial trigger points tests are usually positive in migraine patients, but they were not included in the meta-analyses because of heterogeneity of procedures [404]. If NP and MI are migraine comorbidities, i.e. the consequence of repetitive migraine episodes, or a sign of sensitization present only during the ictal phases ia matter of debate, but a recent study found that MI are present in all phases of the migraine cycle, independent on the presence of NP [405]. To further confirm the need for a proper physiotherapy assessment of migraine patients, a recent study found that more than 56% of migraine patients reported benefits from MT, and more than 90% expressed interest in MT to improve their migraine symptoms [406].
Pain neuroscience education
Pain Neuroscience Education (PNE) is a cognitive-educational approach, aiming at improving patients’ knowledge about pain mechanisms, improve coping strategies, reduce fear, disability, catastrophizing, and avoidance [407]. This approach is helpful with strong-moderate evidence for intermediate-term effectiveness in migraine patients [408]. The combination of PNE with physiotherapy is more effective than physiotherapy alone, thus supporting the integration of different approaches, targeting together pain sensitivity and MI [409]. Alteration of pain sensitivity levels has been largely found in migraine patients [410], and increased pain sensitivity, together with MI, could be used as biomarkers to identify different subgroups of migraine patients with different needs. Recently, various clusters of migraine patients have been identified, according to pain sensitivity and MI [411]: in the ictal phase 81% of migraine patients exhibited increased pain sensitivity and MI, while in the interictal phase 45% only increased pain sensitivity, and 37% increased pain sensitivity and MI. This could be a first step in designing a more tailored treatment approach [412].
Concluding remarks
In summary, evidence exists that exercise, MT and PNE show effect on the improvement of migraine course, and are generally appreciated by patients. In the future high quality studies addressing aforementioned points, such as dosage, modalities, and frequency, should be designed, especially in exercise and MT. This will enable improving the knowledge behind physiotherapy in migraine patients, and possible have a role in international guidelines.
Pharmacovigilance
Pharmacovigilance is defined by the World Health Organization (WHO) as “the science and activities relating to the detection, assessment, understanding and prevention of adverse effects or any other medicine/vaccine-related problem” [413]. Information on the safety of different medications is typically published alongside clinical trials, real-world studies, and case reports, in addition to pharmacovigilance studies about databases such as the WHO Safety Database (VigiBase®) [414] and the United States Food and Drug Administration Adverse Event Reporting System (FAERS) [415]. This chapter summarizes current evidence on the pharmacovigilance of acute and preventive migraine treatments.
Regarding acute medication, pharmacovigilance of triptans, lasmiditan, ubrogepant, and rimegepant have been investigated using VigiBase®, FAERS, and other reporting systems. According to the FAERS, triptans are associated with adverse events (AEs) such as ischemic cerebrovascular events, aneurysms, artery dissections, and pregnancy-related vascular events [416]. In terms of drug dependence, triptans have been found to be similar to ergot derivatives (10.9% and 9.3% of all reports of drug dependence for triptans and ergot derivatives, respectively), according to a French pharmacovigilance database [417]. Interestingly, these proportions were not significantly different from those of benzodiazepines. Additionally, whether triptans with higher lipophilicity (i.e., naratriptan and zolmitriptan) exacerbate depressive illness compared to those with lower lipophilicity (i.e., sumatriptan) has been questioned. A UK-based study using the West Midlands General Practice Research Database, comprising more than 20,000 patients with migraine, revealed that the rates of newly diagnosed depression after treatment were similar between patients prescribed triptans with higher lipophilicity and those with lower lipophilicity (3.9% versus 4.2%) [418]. Recently, the AEs of triptans in relation to breastfeeding have been evaluated based on four (inter)national pharmacovigilance databases. The 26 reports that were identified included breast and/or nipple pain, painful lactation, reflex, and decreased milk production [419].
For new classes of acute medication for migraine, a VigiBase® study comprising 826 and 47,433 reports on lasmiditan and triptans, respectively, found that the most commonly reported AEs for lasmiditan were dizziness (26.8%), feeling abnormal (13.0%), and somnolence (11.6%) [420]. Sedation, serotonin syndrome, euphoric mood, and autoscopy showed the strongest disproportionality signals for lasmiditan. While signals for cardiovascular adverse events were confirmed in triptans, lasmiditan did not show any such signals. Moreover, a recent analysis of 820 reports and 1,661 AEs associated with lasmiditan in the FAERS database focused on differences in AE signals according to demographic factors [421]. The analysis revealed that females were more likely to develop paraesthesia, whereas males tended to experience dizziness. The most common AEs were more likely to occur in older people and at higher doses.
Regarding gepants, 10 and 25 disproportionality signals were identified through the FAERS for ubrogepant and rimegepant, respectively, most of which were related to psychiatric, neurological, gastrointestinal, skin, vascular, and infectious AEs [422]. Another study on ubrogepant-related AEs based on the FAERS reported nausea, somnolence, oral paraesthesia, dizziness, hemiparesis, mental impairment, dysstasia, tinnitus, chest pain, cold sweats, and neck pain [423].
For preventive treatment, a recent systematic literature review on CM found that in real world studies that were published between January 2010 and February 2020, AEs have been reported mostly focusing on OnabotulinumtoxinA [424] and revealed that 0–25.1% of patients reported at least one treatment-related AE. Several studies have investigated AEs associated with CGRP-targeted medications. One of the most frequently reported AEs associated with CGRP mAbs is constipation, which has become more evident in the post-marketing period [425], especially with erenumab [426, 427]. Several vascular complications have also been identified with the use of CGRP-targeted medications, although conclusive evidence is yet to be reported [428]. A real-world database study showed a significant disproportionality signal of Raynaud’s phenomenon with CGRP-targeted medications with an information component of 3.3 (95% confidence interval: 3.0–3.5), compared to triptans and beta-blockers [414]. In particular, erenumab has been associated with worsening blood pressure control irrespective of pre-treatment for hypertension [429]. In addition, myocardial infarction has been reported as a potential AE of erenumab [430]. These vascular AEs of CGRP-targeted medications could result from the potentially impaired vasodilation associated to CGRP. Furthermore, several post-marketing studies of CGRP-targeted medications have revealed that alopecia is a potential side effect of CGRP inhibitors [412]. In this context, it has been hypothesized that CGRP antagonism may impair the promotion of hair growth, the protective effect of hair follicles, and the blood flow supply to hair follicles. Given that many female patients with migraine are of childbearing age, the potential risks of using CGRP mAbs during the perinatal period have been investigated in recent years. Previous pharmacovigilance studies have found no major adverse effects on maternal health, birth defects, or spontaneous abortion; however, long-term data are still needed [431, 432].
Concluding remarks
In summary, several pharmacovigilance studies have investigated AEs associated with migraine treatment, focusing on triptans, OnabotulinumtoxinA, lasmiditan, gepants, and CGRP mAbs. When interpreting the above evidence, caution must be exercised, particularly regarding the following limitations. First, pharmacovigilance reporting systems often do not contain detailed information on patients’ backgrounds and would also pose the risk of underestimation. Second, AEs have been shown to be generally underreported due to a lack of knowledge among medical care providers and patients, with only 5–10% of all adverse drug reactions officially submitted to such systems [433]. Third, AEs of new classes of medication tend to be reported more frequently during the first years of marketing, peaking at the end of the second year after regulatory approval, a phenomenon called “Weber effect” [434]. Nevertheless, it is important to continue gathering information on AEs associated with migraine treatment and to provide patients with updated evidence when necessary, and to enable finding the most appropriate and safe therapy for each patient.
Migraine prognosis
The natural history of migraine can go through a phase of chronification, with a certain degree of variability of progression from EM to CM and it is often accompanied by acute drugs overuse [435]. CM is defined from ICHD-3 as the presence of headaches on ≥ 15 days/month for ≥ 3 months [436]. It has been estimated that Western countries suffer from headache on at least 15 days per month [437], and that CM prevalence has been estimated recently to be 4.6% of the global population [2]. CM prevalence is three times more common in women than men (18.9% vs. 9.8%) and presents two peaks between ages of 18–29 and 40–49 years-old [438]. The term CM has replaced the previously accepted definition of ‘transformed migraine’ which meant a type of headache with increase of the frequency and intensity, but this definition was not unequivocal on the meaning of chronicity of the disease, and includes also headaches with changes of clinical features which could underlie more severe disorders [439].
CM tends to be resistant to analgesics and can lead to the phenomenon of acute overuse of drugs, but should be differentiated from the MOH, which may complicate every headache type and can be caused by all medications used for treating headache [440]. The most effective way to prevent MOH is to identify patients at risk and to educate them about the use of acute medication. The risk is higher in patients with frequent headaches, use of opioids and comorbid anxiety and depression [441]. The burden of CM and MOH can be very high as can have a significant, negative impact on physical, social, and emotional functioning [442, 443].
A recent analysis of the Medication Overuse Treatment Strategy trial has demonstrated how an improvement of preventive medications for migraine can be associated with reduction of the disease burden, measured by a significant reduction of migraine-related physical impairment, anxiety, and symptoms of depression [444]. The best treatment options have been debated for long time, as they were found to be only temporarily effective in the majority of cases and did not guarantee long-term benefits. Topiramate and local injection of OnabotulinumtoxinA have shown efficacy as re-prophylaxis agents after CM detoxification, with a better tolerability profile of OnabotulinumtoxinA [435]. However, a high relapse rate has been observed especially in patients with overuse of opioids [445]. The new “era” of managing CM and MOH has been started from the introduction of monoclonal antibodies against CGRP or the CGRP-receptors. These new drugs have demonstrated to be effective in reducing monthly migraine days in CM and in MOH [446, 447].
Risk factors of migraine chronification and the measures that are effective for reducing the risk progression from EM to CM have been a challenge for several years. EM is defined by the occurrence of episodes of migraine for less of 15 days per month [448]. The recent updates of the GBD study confirms migraine as one of the leading causes of disability, particularly among women under 50 years [1, 2, 449]. Migraine prevalence is approximately 15% among the global population, and it is always more frequently observed in women than in men, in both chronic and episodic forms.
The rate of transformation from EM to CM has been estimated to be 2.5% approximately while only a limited proportion with CM revert back to EM [202]. Factors that increase the risk of progression are genetic predisposition, the occurrence of multiple comorbidities, chronic pain disorders, including fibromyalgia, back and neck pain, and unhealthy lifestyle. Also a high personal and societal burden, e.g. with a variety of psychological and personality traits, stressful life events, and major life changes has been considered as predominant risk factors [448]. Interestingly, also a lower education status, obesity, cutaneous allodynia, female sex and age increase the CM risk [450]. A systematic review and meta-analysis of 13 longitudinal cohort studies and 4 case-controlled studies has found that the most important modifiable risk factors for CM, which may provide important targets for intervention are depression, and medication overuse/high-frequency use, both in adults and adolescent/children [451]. This data confirms the importance of avoiding the drug abuse also in the earliest phases of the disease.
The recent COVID-19 pandemics has highlighted other potential risk factors of severe and persistent headache or chronification of migraine also after the acute infection. A prior history of headache, especially migraine, is an important risk factor of persistence of pain also in the chronic course (defined long COVID headache). Another predictive factor of persistence of headache after the acute infection from SARS-CoV-2 is the resistance to common analgesics during the acute disease [452]. Also headache intensity (a more severe headache during the acute SARS-CoV-2 infection) seems to predict the persistence of pain in the long-term course. Other symptoms can accompany headache such as dizziness, cognitive dysfunction, insomnia/sleep disorders and fatigue [453, 454]. Older patients with multiple comorbidities and frailty might experience a more severe disease as compared to young subjects [455,456,457].
A small but still undetermined percentage of people with migraine experience disabling attacks that various therapeutic strategies are unable to control. Resistant and refractory migraine are new clinical definitions resulting primarily from the discovery of migraine-specific preventive treatments but also from a better understanding of the pathogenesis of this disease [458]. Characterized by ≥ 8 monthly days of debilitating headaches and inadequate response, intolerance or contraindication to ≥ 3 or all classes of preventive medications, respectively [459]. Resistant migraine is mainly due to drug failure and refractory migraine has complex and still unknown mechanisms that undermine the effectiveness of preventive treatment. Factors leading to or promoting resistance and refractoriness to preventative migraine drugs are as follows: psychiatric comorbidities and sleep disorders, poor diet, limited exercise, bad sleep habits, delayed or lack of access to medical care [451, 460, 461].
Concluding remarks
In summary, migraine prognosis is largely confounded by the presence of comorbidities, avoidable risk factors and, in particular, the possibility to get an accurate diagnosis and a specific treatment, with prophylaxis playing a specific role for reverting a chronic into an episodic course. Most likely, those who suffer from migraine will be subject to recurrent episodes of migraine headache during period of life in which migraine prevalence reaches its higher peaks. Considering the very good response rates or new specific therapies, i.e. the monoclonal antibodies against CGRP or CGRP-receptors, as well as those which will enter in the market soon, it is possible to presume that in the next forthcoming years the prognosis of migraine will be more favourable.
Psychosocial impact of migraine
The psychosocial impacts attributed to migraine include a broad variety of personal difficulties that contribute to the global disability faced by patients. Data concerning measures of global disability have been clearly reported and demonstrate that migraine is the 2nd leading cause of disability across the global population [462]. Additionally, migraine remains the leading cause of disability amongst women under the age of 35 years [462]. When considering the extreme psychosocial impact faced by individuals with migraine, the value of both defining and addressing common psychosocial difficulties (PSDs) and their role in the clinical course of an individual’s diagnosis cannot be understated.
Measures that aim to estimate global disability experienced by a patient with migraine have been used in clinical practice for over 20 years. Currently, the most used scales of migraine disability are the Migraine Disability Assessment Test, the 6-items Headache Impact Test, and the Migraine Specific Quality of Life questionnaire [463,464,465]. While these scales remain frequently used due to their simple methodology, authors have voiced concerns regarding the accuracy, validity, and value of findings derived from such a general approach to a measure of disease related disability [466, 467]. Additionally, non-disease specific scales of disability such as the Patient Health Questionnaire-9, Visual Analog Scale, Work Productivity and Activity Impairment (WPAI), and WHO Disability Assessment Schedule 2.0 are frequently assessed in patients with migraine [468, 469].
While the burden of global disability attributed to migraine has been well reported, data defining specific psychosocial impacts and individual PSDs experienced by patients remain incompletely described. When attempting to interpret the PSDs experienced by a patient with migraine, it is crucial to assess the patient through a biopsychosocial model, as defined by the International Classification of Functioning, Disability and Health [470]. The classification defines PSDs as impairment of mental function, limitation of activity, restriction of participation in events that involve social interactions, such as in work, family life and leisure activities, as well as daily activities such as those connected to daily routing, homework or mobility [9]. Therefore, measuring disability with respect to all contributing PSDs is necessary to adequately define the true disability attributed to migraine.
A leading PSD reported by patients with migraine is related to impaired emotional function [471]. Individuals with migraine report significantly higher levels of comorbid mood disturbances such as depressed mood and anxiety when compared to the general population [472,473,474]. Strong evidence has demonstrated a correlation between headache frequency, global disability attributed to migraine, and worsening emotional function [471, 474,475,476]. In contrast, a recent study investigating the therapeutic role of CGRP mAbs in the preventative treatment of migraine concluded that CGRP mAbs therapy, regardless of patient response status, was associated with improved depressive symptoms [477]. While less thoroughly investigated, emotional disturbances including stress and anger are often reported by patients with migraine [471, 478, 479].
Another category of PSDs that are frequently reported by patients with migraine are factors associated with energy and drive functions including vitality, fatigue, and motivation [471]. Pathologic fatigue is reported by approximately 60% of patients with migraine and is highly associated with headache intensity and global disability [480]. Furthermore, it is proposed that impaired vitality and fatigue attributed to migraine are bidirectionally associated with mood disturbances and emotional functioning [471, 473, 481]. Further evidence of this relationship has been demonstrated in multiple studies reporting that patients treated with adequate acute and preventative migraine therapy report significantly lower burdens of fatigue and generally report improved vitality and motivation [475, 480, 481]. Furthermore, sleep disturbances are common in patients with migraine which is likely correlated with global migraine burden and mood symptoms [471, 482,483,484]. Assuring optimal migraine therapy is likely to lessen the burden of sleep disturbances and energy and drive functions amongst patients with migraine.
A leading economic PSD reported by patients with migraine are factors associated with work and school activities [485, 486]. Economic studies have consistently demonstrated that patients with migraine report a significantly higher burden in work related function secondary to increased workplace absenteeism and presenteeism, which is highly correlated with global disability [487,488,489]. While factors related to employment have been more thoroughly studied, data investigating school-related PSDs reported by students remain poorly outlined. It is generally acknowledged that school-age children with migraine are at an increased risk of school-related and learning difficulties secondary to absenteeism and presenteeism [490]. However, high-quality data providing definitive findings related to the PSDs faced by school-age children remain scarcely available.
PSDs associated with social functioning are often reported by patients with migraine, including feelings of isolation and loneliness [471, 491]. Available data supports a strong relationship between pain intensity, headache frequency, and social impairment [492, 493]. While poorly investigated, a therapeutic reduction in migraine pain intensity and a reduction in monthly headache burden is correlated with improved social function [471]. Somewhat related to social function, a final PSD attributed to migraine is the sequelae associated with substance abuse. While data defining the burden attributed to medication overuse headache is diffuse, limited data has investigated the multiple overlapping features between patients with CM associated to MOH and patients with substance use disorder [494]. While the relationship between habitual medication consumption as a precursor to substance abuse remains poorly elucidated, factors including increased headache pain intensity, increased consumption of acute medication, and impaired emotional function are all correlated with an increased risk of substance abuse [6, 440, 494]. Future studies are of utmost necessity to isolate contributing factors that lead to substance abuse amongst patients with migraine.
Concluding remarks
In summary, migraine has a considerable psychosocial impact, which can be detected at the level of individuals and of society. Individuals experience unpredictable effects of their conditions, which hampers their ability to interact with others, attend work or school activities, which can moreover be worsened by associated mood symptoms. The impact at societal level is mostly due to reduced workforce participation and productivity, which makes migraine one of the leading causes of health expenditure.
Occupational health consideration in migraine
Work-related stress is reported to predict new onset migraine in workers with no prior migraine history [495]. Stress has an impact on the incidence, chronification, and perceived burden of migraine [478]. However, occupational stress may be particularly difficult to avoid for those who are employed, and the extent of migraine-associated disability in the workplace may be influenced by psychosocial factors, such as job satisfaction which mediates the association between perceived disability and work productivity in workers with migraine [496].
Occupational factors may influence migraine prevalence. One example is the type of work performed. For instance, compared to the general population, the migraine risk appears to be increased in health care professionals, with notable differences among physicians of different specialties [497]. A recent study also found that among bank employees, those working in data analysis and information technology had higher migraine rates [498]. More work is needed to firmly establish which job factors may account for differences in migraine prevalence, and the extent to which they may be modifiable. There is additionally accumulating evidence in the literature regarding the association of shift work with migraine, and shift work may also act as a barrier to optimal migraine management [499, 500].
Migraine has been associated with multiple chemical sensitivity (also known as idiopathic environmental intolerance) [501], a controversial condition that nevertheless can be extremely disabling, and may greatly interfere with work ability. Olfactory-induced migraine triggered by low-level chemical exposures should be considered in the differential diagnosis for multiple chemical sensitivity syndrome. Distinguishing olfactory-induced migraine from multiple chemical sensitivity syndrome matters greatly in clinical practice, as the former can often be effectively treated or even prevented with pharmacotherapy, whereas such options do not exist for the latter.
Migraine may impact workplace productivity, as it peaks during peoples’ most productive work years. A recent burden of disease analysis from the UK showed that being affected by migraine is associated with absence from work, unemployment, being disabled, and early retirement [502]. Another study found that 89% of migraine-related productivity loss is due to presenteeism, rather than absenteeism, and that migraine represents approximately 16% of total presenteeism in the United States [503]. Patients self-reported being only 46% effective while on the job with migraine symptoms [504]. Factors reportedly contributing to migraine-related productivity loss include migraine symptoms, and also the unpredictability of attacks, comorbidities, the emotional impact of migraine, the impact of underdiagnosis and under-management, and stigma associated with the condition [503]. Traditionally, the impact is measured in terms of lost workdays or days worked with reduced productivity, but in recent years questionnaire-based approaches have been implemented as well, e.g. the WPAI [505] and the HEADWORK questionnaire, which specifically addresses addressing the impact of migraine on work-related difficulties in terms of difficulties, and the factors contributing to these difficulties [506].
Workplace programs for headache education and evaluation are associated with significant productivity gains and cost savings for the employer [507]. Pharmacological therapy, notably rizatriptan, has also been studied along with workplace programming [508, 509] and appears to demonstrate benefits. The effect of workplace programming may be modified by the national context, in particular the integration of employment with health care insurance and/or provision of health services.
Migraine is widely acknowledged as a difficult condition to manage in the workplace, even by occupational physicians [510]. In addition to pain, associated cognitive symptoms, photo- and phonophobia are most highly associated with migraine-related disability [511] and must be considered when assessing fitness to work, particularly in safety-sensitive and decision-critical [512] positions. The approach to management of migraine in workers needs to take a number of factors into consideration, including the characteristics of the migraine and its triggers, the migraine treatments used and their adverse effects, the worker’s other health issues, as well as the workplace and job characteristics. The accident risk may be increased in newly diagnosed patients with migraine, as road safety data from older drivers suggest [513]. Whether a prodrome exists prior to the onset of disabling symptoms and how long this prodrome lasts may help inform fitness to work decisions. As well, attendance expectations may need to be adjusted and backup personnel available to take over safety-sensitive or decision-critical roles. Other examples of workplace accommodations for migraine that have been described in the literature include flexible scheduling, the ability to work from home when possible, a scent-free work environment, optimization of desk ergonomics, and provision of tinted glasses to reduce light sensitivity [503].
Concluding remarks
In summary, migraine impacts on work productivity, in terms of reduction in ability to carry out work duties, and some work-related factors may negatively impact on migraine course which, in turn, might further on reduce productivity. Migraine is acknowledged to be a difficult to treat condition in the context of workplace: however, some initiatives to improve workplaces’ inclusiveness, such as the “Migraine-Friendly Workplace” initiative led by the European Migraine and Headache Alliance exist (see: https://www.emhalliance.org/migraine-friendly-workplaces/).
Economic impact of migraine
The economic impact of migraine, covering both episodic migraine (EM) and chronic migraine (CM), is a significant concern. Migraine imposes direct costs on healthcare systems, including expenses for medication treatments, primary care visits, diagnostic tests, specialist consultations, emergency visits, and hospitalizations. These costs vary significantly across countries due to differences in healthcare systems and migraine management approaches. Additionally, migraine leads to substantial indirect costs, such as productivity losses, absenteeism, and reduced quality of life for individuals, families, employers, and societies. The economic impact extends to intangible costs, including pain, anxiety, and emotional distress, along with limitations in family and social activities. However, these intangible costs are difficult to quantify and are rarely accounted for. Understanding the economic impact of migraine is essential for health policymakers, providers, and stakeholders to develop effective and cost-efficient strategies for prevention, management, and resource allocation.
A narrative review by Leonardi on the burden of migraine from 1990 to 2018 identified 49 publications focusing on the burden or impact of migraine, including both EM and CM [5]. Among the identified main themes, impact on work or school activities, and disease costs were included. The impact on work-related activities and daily life was a significant focus due to tangible consequences like costs, and 22 studies addressing this aspect. Findings indicated that migraine significantly affected productivity, leading to considerable losses due to reduced productivity at work (‘presenteeism’) or absence from work (‘absenteeism’). On average, patients with migraine lost between 3.2 and 89.2 work-equivalent days per year, with presenteeism contributing significantly to productivity losses. Overall, indirect costs, including work loss and reduced productivity, outweigh direct medical expenses. For example, in Europe, 93% of the annual per-person cost of €1,177 for migraine is attributed to indirect costs [514].
Family life was also impacted by migraine, as evidenced by five studies in the review [5]. Results indicated that family burden increased with headache frequency, with caregivers facing challenges in caring for affected members, particularly children. Two studies highlighted the burden of caregiving for individuals with migraine, noting its impact on family life and workforce participation. Eleven studies from 2001 to 2017 provided data on the costs associated with both EM and CM [5]. Discrepancies in total costs were observed, likely due to variations in cost structures, such as focusing solely on direct costs versus including both direct and indirect costs, as well as differences in survey years. For example, a US-wide study reported the annual total cost of EM at $2,649 (€2,444) and CM at $8,243 (€7,606) [515]. In this study, direct medical costs accounted for 60–64% of migraine-related expenses. Conversely, a Europe-wide study found the average annual direct cost of EM to be €746 and CM to be €2,427 [515]. Generally, CM incurred higher costs compared to EM, with direct medical costs comprising a significant portion of total expenses. Studies consistently indicate that the costs associated with CM are three to four times higher than those of EM.
A subsequent review by Eltrafi et al. (2023) of thirteen studies on the economic impact of CM revealed significant variations in cost estimates, underscoring the substantial burden on patients and healthcare systems [516]. While direct costs like hospitalization and medication expenses were examined, indirect costs related to productivity losses were often underexplored. None of the studies quantified intangible costs, such as emotional and social impacts. Direct healthcare costs per patient ranged from £1,754 (€2,043) to £8,219 (€9,574) annually, with indirect costs ranging from £2,579 (€3,004) to £48,810 (€56,856) annually, influenced by methodological differences. The review highlights the need for comprehensive assessments of both direct and indirect costs to understand CM’s economic impact fully.
While most economic evidence focuses on CM or high-frequency EM, leaving out many migraine sufferers with fewer monthly headache episodes, a recent study in Spain aimed to address this gap [488]. The study evaluated the societal and economic impact of migraine using monthly headache days (MHD) as a measure, comparing it to individuals without migraine. The yearly cost per migraine patient was calculated at €8,894, with €894 (10.1%) as direct costs and €8,000 (89.9%) as indirect costs related to absenteeism and presenteeism. The findings advocate for stratifying patients based on MHD rather than distinguishing between EM and CM. Individuals with 1–3 MHD incurred an additional annual cost of €2,724 per person compared to those without migraine, a 1.5-fold increase. For patients with ≥ 15 MHD, the cost doubled compared to individuals without migraine.
In linking migraine-related impairment (symptom burden) and reduced productivity, frequency (number of MHD) emerges as the primary factor, surpassing headache intensity and episode duration [517]. This finding has significant implications for current headache care practices, health policy decisions, and healthcare resource investments. Despite being underused, preventive medications have considerable potential for delivering economic benefits, possibly leading to cost savings. Economic benefits, particularly in males, are substantial, with preventing one migraine day per month resulting in a recovery of approximately 28% of lost productivity from paid work every three months, surpassing the previously estimated 20% threshold for structured headache services to be cost-saving.
Headache disorders, including migraines, are highly prevalent in India, significantly impacting productivity and quality of life. In a door-to-door survey of 2,329 adults in Bangalore, 63.9% reported experiencing headaches in the past year. Migraine prevalence was 25.2%, with severe intensity and significant disability, reducing the functional capacity of the adult population by 0.46%. Despite the high burden, fewer than a quarter of sufferers sought medical help. Structured headache services in primary care are recommended as an efficient, effective, and equitable solution, highlighting the need for increased political awareness and will to address this issue [518].
A recent Australian study quantified the health and productivity burden of migraines in Australia using quality-adjusted life years (QALYs), productivity-adjusted life years (PALYs), and associated costs. A Markov model simulated outcomes for Australians aged 20–64 over 10 years, comparing current migraine prevalence to a hypothetical scenario without migraines. Results showed 8.5% of this population suffers from migraines, leading to a loss of 2.58 million QALYs and AU$1.67 billion in health-care costs. Additionally, 384,740 PALYs were lost, costing AU$68.13 billion in GDP. These findings highlight the significant economic impact of migraines and the potential return on investment in effective interventions [519].
Concluding remarks
In summary, migraine has a significant economic impact on societies, measured both with mathematical models and with primary data, and the majority of the cost of migraine is associated to indirect costs, i.e. reduced productivity and loss of workdays. The advocacy by “Lifting The Burden”, as part of its Global Campaign against Headache, for structured headache services [520] as an equitable and efficient healthcare solution for managing headaches is supported by evidence of cost-effectiveness derived from theoretical economic analytical modelling [12, 521]. Effective care, by reducing symptom burden, has the potential to restore lost productivity, suggesting that investment in care could yield cost-saving benefits.
Underserved populations
Migraine affects individuals across all socio-economic strata and can be particularly severe in those who are less privileged. This group of patients is often underdiagnosed, lacks access to specialized care, and rarely receives targeted treatments. Underserved populations have specific needs that should be addressed to improve migraine management and care. Underserved populations are not defined by a single criterion. Broadly speaking, they include individuals who face barriers to accessing healthcare due to socioeconomic status, geographical location, ethnicity, gender, or other factors. These barriers result in significant inequalities, providing these individuals with considerably fewer opportunities compared to others. The impact of these barriers on migraine care is herein discussed.
Socioeconomic status is a significant determinant of health outcomes, and this is also true for migraine sufferers. While the disease affects individuals from economic backgrounds, research has shown that socioeconomic status influences headache prevalence and burden, particularly when implicated in access to healthcare [522]. In the US, migraine is more prevalent in low-income households [523], and these patients visit emergency departments for headache treatment more frequently, which may imply suboptimal care [524]. Similarly, studies from Russia and Georgia have shown that poverty is associated with experiencing 15 or more headache days per month, also suggesting an unmet treatment need [525, 526]. In practice, advancements in migraine therapeutics that have improved migraine management for those who can afford them are often inaccessible to most patients worldwide. Notably, an evaluation in Latin America revealed that there are very few specialized headache training opportunities in the region, and most physicians with specific training primarily treat the wealthiest minorities in their countries [527].
Geographical Barriers exist as well. Access to healthcare services is often limited in rural and remote areas, where healthcare infrastructure is sparse. This is true not only for lower-income economies [528, 529] but also for countries like the United States, where only a few patients live in the same city where they receive treatment [530], and most patients have to travel more than one hundred miles on average to see a specialist [531].
Ethnic and racial minorities face unique challenges in migraine management. In the United States, American Indian and Alaskan Native populations, which are often neglected by society, have the highest migraine prevalence [523]. Blacks and Hispanics are significantly less likely to be diagnosed with migraine compared to whites. Specifically, African American patients are 25% less likely to receive a migraine diagnosis, and Hispanic patients are 50% less likely to receive a migraine diagnosis compared to White patients [523]. Furthermore, Black individuals are less likely to be prescribed acute migraine therapy compared to White individuals [523] and similarly, Hispanics are less likely to receive adequate prophylactic treatment in ambulatory settings [532, 533].
Gender differences have to be acknowledged too. Despite migraine is more prevalent and often more severe in women [534], women’s pain is frequently under-recognized and under-treated, a phenomenon known as "gender bias" in pain management. This issue may be exacerbated in some countries where migraine is sometimes misunderstood as a conversion disorder, with its biological underpinnings being neglectfully overlooked [535, 536]. Additionally, while prevalence studies in gender minority groups are limited, available evidence suggests that migraine prevalence in these populations might be relatively high [537].
Finally, some consideration on research-related disparities have to be acknowledged. In both absolute terms and relative to its burden, migraine research is drastically underfunded [538, 539]. This scenario is particularly pronounced in low-income countries [527], which in addition are often excluded from most migraine clinical trials. This exclusion not only hampers the ability to generalize results to the majority of individuals living with migraine worldwide [540] but also reduces the chances of developing headache research in these countries.
Concluding remarks
In summary, addressing the needs of underserved populations in migraine care requires a multifaceted approach that includes increasing access to healthcare, raising awareness, and implementing innovative strategies (Table 5). By focusing on the unique challenges faced by these groups, we can improve diagnosis, treatment, and quality of life for all individuals living with migraines. Ensuring equitable care and resources for underserved populations is not only a matter of social justice but also a critical step toward reducing the overall burden of migraine on society.
Conclusions
The most relevant hallmark of migraine is that it has common and individual features together. The most relevant commonality is connected to the clinical manifestation of migraine, i.e. unilateral location of headache, with pulsating quality, pain of moderate/severe intensity, which is aggravated by physical activity and associated to either nausea, photophobia or phonophobia [33]. Excluding this, the presentation is heterogeneous with regard to frequency of attacks, presence of aura, response to therapy, associated comorbidities or other symptoms. This likely reflects the heterogeneity of migraine with regard to its genetic and molecular basis. Several variants have been identified, which mostly underline vascular and neuronal tissues activity, and large evidence exist on the role of neuromodulators and neurotransmitters like 5HT, CGRP, amylin, PACAP and NO, which led to the development of migraine-specific therapies both for acute treatment and for prophylaxis, such as triptans or mAbs targeting GCRP.
The amount of therapies for treating migraine in acute and for prophylaxis is really wide, to the extent that one of the difficulties is with finding the best treatment for the single patient, considering its clinical features, comorbidity profile, but also by making considerations about non-response to treatment or presence of refractory migraine are needed [267, 541, 542]. Yet, this is not enough: patients have different lives, in which carry out different activities, and might show lifestyle habits which are not entirely adequate to manage migraine. These include, for example, skipping meals, being poorly hydrated, having insufficient sleep: therefore, patient education on lifestyle issues, as well as on ways to avoid triggers, should be part of the treatment [543]. Education will be more and more important in the future, as a strategy of brain health promotion [7, 11, 544], because this will enable reducing the amount of subjects who will need specialty care for migraine, thus leaving it to those who are in need in reason of refractory condition or presence of comorbidities.
The information herein presented is not intended to be systematic or entirely exhaustive. The approach was narrative and each of the authors, in preparing the section assigned to them, included the most updated, comprehensive and precise evidence possible addressing migraine specific features, from genetic and molecular basis up to the impact at societal level. The most relevant consequence of this is that the contents represent the opinions of the authors, and not the results of a systematic review of the available evidence. Nevertheless, the cultural worth, especially for non-migraine experts, represents a relevant strength of this paper.
Recognizing the hallmarks of migraine and the features of patients’ daily lives thus enables not only to prescribe specific pharmacological treatments, but also non-pharmacological ones, such as physical therapies, non-invasive neuromodulation, nutraceuticals, and behavioural treatments. Providing the best possible treatment, considering social and cultural peculiarities [7, 11], will be more and more of importance to reduce the impact of a heterogeneous condition which affects approximately 1.16 billion people worldwide [1] and is the second cause of health loss among young adults, accounting for 7% of all-cause YLDs (7.5% among females) [545].
Medical research on headaches today particularly suffers from the syndrome of single-disease niche sub-specialties, epitomizing a poor propensity for comparison among different disciplines, thus emitting a monolithic type of scientific light [546,547,548]. This leads to a limitation of results that are brilliant yet lack a systematic project articulation. This is evident in the widespread vagueness of the discussion in many contemporary original research papers, with little to no interpretative vision, resulting in a reduced attractiveness of funding [549].
Without resorting to the now imaginative, old-fashioned holistic vision of the able generalist physician, it is more necessary than ever today for researchers to have a cross-sectional and joint vision with other niche disease specialties and stop relegating themselves to the role of fine technicians in the service of an organ or a function. This is one of the goals of this trilogy of Hallmarks of Primary Headaches, like a fil rouge that can lead from one end to the other, from the extreme, distal one dedicated to researchers, to the proximal, practical one dedicated to clinicians.
Let’s start with migraine.
Data availability
No datasets were generated or analysed during the current study.
Abbreviations
- 5-HT:
-
Serotonin
- ACT:
-
Acceptance and Commitment Therapy
- AEs:
-
Adverse Events
- AMY1:
-
Amylin 1
- BBB:
-
Blood–Brain Barrier
- BKCa:
-
Calcium-Activated Potassium
- BOLD:
-
Blood Oxygenation Level Dependent
- CADASIL:
-
Cerebral Autosomal Dominant Arteriopathy with Subcortical Infarcts and Leukoencephalopathy
- cAMP:
-
Cyclic Adenosine Monophosphate
- CBF:
-
Cerebral Blood Flow
- CBT:
-
Cognitive Behavioral Therapy
- cGMP:
-
Cyclic Guanosine Monophosphate
- CGRP:
-
Calcitonin Gene-Related Peptide
- CLR:
-
Calcitonin-Like Receptor
- CM:
-
Chronic Migraine
- CNS:
-
Central Nervous System
- CoQ10:
-
Coenzyme Q10
- CPM:
-
Conditioned Pain Modulation
- CSD:
-
Cortical Spreading Depression
- CTR:
-
Calcitonin Receptor
- EM:
-
Episodic Migraine
- FAERS:
-
Food and Drug Administration Adverse Event Reporting System
- FHM:
-
Familial Hemiplegic Migraine
- fMRI:
-
Functional Magnetic Resonance Imaging
- GABA:
-
γ-Aminobutyric Acid
- GBD:
-
Global Burden of Disease Study
- GPCR:
-
G-Protein Coupled Receptor
- GWAS:
-
Genome-Wide Association Studies
- HM:
-
Hemiplegic Migraine
- HRV:
-
Heart Rate Variability
- KD:
-
Ketogenic Diet
- Kir:
-
Inward rectifier potassium
- mAbs:
-
Monoclonal Antibodies
- MBSR:
-
Mindfulness-Based Stress Reduction
- MHD:
-
Monthly Headache Days
- MI:
-
Musculoskeletal Impairment
- MMD:
-
Monthly Migraine Days
- MOH:
-
Medication Overuse Headache
- MRS:
-
MR Spectroscopy
- MT:
-
Manual Therapy
- NO:
-
Nitric Oxide
- NP:
-
Neck Pain
- ONS:
-
Occipital Nerve Stimulation
- OPFAs:
-
Omega-3 polyunsaturated fatty acids
- PA:
-
Physical Activity
- PACAP:
-
Pituitary Adenylate Cyclase Activating Polypeptide
- PALYs:
-
Productivity-Adjusted Life Years
- PDEs:
-
Phosphodiesterases
- PEA:
-
Palmitoylethanolamide
- PKC:
-
Protein Kinase C
- PMR:
-
Progressive Muscle Relaxation
- PNE:
-
Pain Neuroscience Education
- PNS:
-
Peripheral Nervous System
- PRS:
-
Polygenic Risk Score
- PSDs:
-
Psychosocial Difficulties
- QALYs:
-
Quality-Adjusted Life Years
- RAMP1:
-
Receptor Activity-Modifying Protein 1
- RCT:
-
Randomized Clinical Trial
- SNPs:
-
Single Nucleotide Polymorphisms
- SNS:
-
Supraorbital Nerve Stimulation
- SUR:
-
Sulfonylurea Receptor
- tACS:
-
Transcranial Alternate Current Stimulation
- tDCS:
-
Transcranial Direct Current Stimulation
- TG:
-
Trigeminal Ganglion
- TMS:
-
Transcranial Magnetic Stimulation
- TNC:
-
Trigeminal Subnucleus Caudalis
- TRP:
-
Transient Receptor Potential
- TVS:
-
Trigeminovascular System
- VIP:
-
Vasoactive Intestinal Peptide
- VNS:
-
Vagus Nerve Stimulation
- WHO:
-
World Health Organization
- WPAI:
-
Work Productivity and Activity Impairment
- YLDs:
-
Years Lived with Disability
References
GBD (2021) Nervous System Disorders Collaborators (2024) Global, regional, and national burden of disorders affecting the nervous system, 1990–2021: a systematic analysis for the Global Burden of Disease Study 2021. Lancet Neurol 23:344–381
Stovner LJ, Hagen K, Linde M et al (2022) The global prevalence of headache: an update, with analysis of the influences of methodological factors on prevalence estimates. J Headache Pain 23:34
Dodick DW (2018) Migraine. Lancet 391:1315–1330
Caponnetto V, Deodato M, Robotti M et al (2021) Comorbidities of primary headache disorders: a literature review with meta-analysis. J Headache Pain 22:71
Leonardi M, Raggi A (2019) A narrative review on the burden of migraine: when the burden is the impact on people’s life. J Headache Pain 20:41
Waliszewska-Prosół M, Montisano DA, Antolak M et al (2024) The impact of primary headaches on disability outcomes: a literature review and meta-analysis to inform future iterations of the Global Burden of Disease study. J Headache Pain 25:27
Martelletti P, Leonardi M, Ashina M et al (2023) Rethinking headache as a global public health case model for reaching the SDG 3 HEALTH by 2030. J Headache Pain 24:140
Steiner TJ, Husøy A, Thomas H et al (2023) The HARDSHIP databases: a forthcoming free good from the Global Campaign against Headache. J Headache Pain 24:21
Viana M, Khaliq F, Zecca C et al (2020) Poor patient awareness and frequent misdiagnosis of migraine: findings from a large transcontinental cohort. Eur J Neurol 27:536–541
Caronna E, Gallardo VJ, Alpuente A et al (2022) Epidemiology, work and economic impact of migraine in a large hospital cohort: time to raise awareness and promote sustainability. J Neurol 269:1456–1462
Leonardi M, Martelletti P, Burstein R et al (2024) The World Health Organization Intersectoral Global Action Plan on Epilepsy and Other Neurological Disorders and the headache revolution: from headache burden to a global action plan for headache disorders. J Headache Pain 25:4
Tinelli M, Leonardi M, Paemeleire K et al (2021) Structured headache services as the solution to the ill-health burden of headache. 3. Modelling effectiveness and cost-effectiveness of implementation in Europe: findings and conclusions. J Headache Pain 22:90
Steiner TJ, Jensen R, Katsarava Z et al (2019) Aids to management of headache disorders in primary care (2nd edition): on behalf of the European Headache Federation and Lifting The Burden: the Global Campaign against Headache. J Headache Pain 20:57
Pascual J, Panni T, Dell’Agnello G et al (2023) Preventive treatment patterns and treatment satisfaction in migraine: results of the OVERCOME (EU) study. J Headache Pain 24:88
Lampl C, MaassenVanDenBrink A, Deligianni CI et al (2023) The comparative effectiveness of migraine preventive drugs: a systematic review and network meta-analysis. J Headache Pain 24:56
Cheng YC, Zeng BY, Hung CM et al (2022) Effectiveness and acceptability of noninvasive brain and nerve stimulation techniques for migraine prophylaxis: a network meta-analysis of randomized controlled trials. J Headache Pain 23:28
Bae JY, Sung HK, Kwon NY et al (2021) Cognitive Behavioral Therapy for Migraine Headache: A Systematic Review and Meta-Analysis. Medicina (Kaunas) 58:44
Russell MB, Hilden J, Sørensen SA et al (1993) Olesen J. Familial occurrence of migraine without aura and migraine with aura. Neurology 43:1369–1373
Russell MB, Olesen J (1995) Increased familial risk and evidence of genetic factor in migraine. BMJ 311:541–544
Ophoff RA, Terwindt GM, Vergouwe MN et al (1996) Familial hemiplegic migraine and episodic ataxia type-2 are caused by mutations in the Ca2+ channel gene CACNL1A4. Cell 87:543–552
Vanmolkot KR, Kors EE, Hottenga JJ et al (2003) Novel mutations in the Na+, K+-ATPase pump gene ATP1A2 associated with familial hemiplegic migraine and benign familial infantile convulsions. Ann Neurol 54:360–366
Auffenberg E, Hedrich UB, Barbieri R et al (2021) Hyperexcitable interneurons trigger cortical spreading depression in an Scn1a migraine model. J Clin Invest 131:e142202
Riant F, Roos C, Roubertie A et al (2022) Hemiplegic migraine associated with PRRT2 variations: a clinical and genetic study. Neurology 98:e51–e61
Adib-Samii P, Brice G, Martin RJ, Markus HS et al (2010) Clinical spectrum of CADASIL and the effect of cardiovascular risk factors on phenotype: study in 200 consecutively recruited individuals. Stroke 41:630–634
Hautakangas H, Winsvold BS, Ruotsalainen SE et al (2022) Genome-wide analysis of 102,084 migraine cases identifies 123 risk loci and subtype-specific risk alleles. Nat Genet 54:152–160
Häppölä P, Gormley P, Nuottamo ME et al (2022) Polygenic risk provides biological validity for the ICHD-3 criteria among Finnish migraine families. Cephalalgia 42:345–356
Yang Y, Zhao H, Boomsma DI et al (2018) Molecular genetic overlap between migraine and major depressive disorder. Eur J Hum Genet 26:1202–1216
Guo Y, Rist PM, Daghlas I et al (2020) A genome-wide cross-phenotype meta-analysis of the association of blood pressure with migraine. Nat Commun 11:3368
Charles AC, Baca SM (2013) Cortical spreading depression and migraine. Nat Rev Neurol 9:637–644
Hiekkala ME, Vuola P, Artto V et al (2018) The contribution of CACNA1A, ATP1A2 and SCN1A mutations in hemiplegic migraine: a clinical and genetic study in Finnish migraine families. Cephalalgia 38:1849–1863
Russell MB, Ducros A (2011) Sporadic and familial hemiplegic migraine: pathophysiological mechanisms, clinical characteristics, diagnosis, and management. Lancet Neurol 10:457–470
Lykke Thomsen L, Kirchmann Eriksen M, Faerch Romer S et al (2002) An epidemiological survey of hemiplegic migraine. Cephalalgia 22:361–375
Headache Classification Committee of the International Headache Society (IHS) (2018) The International Classification of Headache Disorders, 3rd edition. Cephalalgia 38:1–211
Thomsen LL, Olesen J, Russell MB (2003) Increased risk of migraine with typical aura in probands with familial hemiplegic migraine and their relatives. Eur J Neurol 10:421–427
Friedrich T, Tavraz NN, Junghans C (2016) ATP1A2 Mutations in Migraine: Seeing through the Facets of an Ion Pump onto the Neurobiology of Disease. Front Physiol 7:239
Guey S, Hervé D (2022) Main features of COL4A1-COL4A2 related cerebral microangiopathies. Cereb Circ Cogn Behav 3:100140
Wilms AE, de Boer I, Terwindt GM (2022) Retinal Vasculopathy with Cerebral Leukoencephalopathy and Systemic manifestations (RVCL-S): An update on basic science and clinical perspectives. Cereb Circ Cogn Behav 3:100046
Lafrenière RG, Cader MZ, Poulin JF et al (2010) A dominant-negative mutation in the TRESK potassium channel is linked to familial migraine with aura. Nat Med 16:1157–1160
Brennan KC, Bates EA, Shapiro RE et al (2013) Casein kinase iδ mutations in familial migraine and advanced sleep phase. Sci Transl Med 5:183ra56, 1–11
Williams LB, Javed A, Sabri A et al (2019) ALPK1 missense pathogenic variant in five families leads to ROSAH syndrome, an ocular multisystem autosomal dominant disorder. Genet Med 21:2103–2115
Gormley P, Anttila V, Winsvold BS et al (2016) Meta-analysis of 375,000 individuals identifies 38 susceptibility loci for migraine. Nat Genet 48:856–866
Choquet H, Yin J, Jacobson AS et al (2021) New and sex-specific migraine susceptibility loci identified from a multiethnic genome-wide meta-analysis. Commun Biol 4:864
Gormley P, Kurki MI, Hiekkala ME et al (2018) Common variant burden contributes to the familial aggregation of migraine in 1,589 families. Neuron 98:743-753.e744
Malik R, Freilinger T, Winsvold BS et al (2015) Shared genetic basis for migraine and ischemic stroke: a genome-wide analysis of common variants. Neurology 84:2132–2145
Daghlas I, Guo Y, Chasman DI (2020) Effect of genetic liability to migraine on coronary artery disease and atrial fibrillation: a Mendelian randomization study. Eur J Neurol 27:550–556
Daghlas I, Vgontzas A, Guo Y et al (2020) Habitual sleep disturbances and migraine: a Mendelian randomization study. Ann Clin Transl Neurol 7:2370–2380
Adewuyi EO, Sapkota Y, International Endogene Consortium Iec et al (2020) Shared molecular genetic mechanisms underlie endometriosis and migraine comorbidity. Genes (Basel) 11:268
Kamm K (2022) CGRP and migraine: what have we learned from measuring CGRP in migraine patients so far? Front Neurol 13:930383
Gallai V, Alberti A, Gallai B et al (2003) Glutamate and nitric oxide pathway in chronic daily headache: evidence from cerebrospinal fluid. Cephalalgia 23:166–174
Lassen LH, Haderslev PA, Jacobsen VB et al (2002) CGRP may play a causative role in migraine. Cephalalgia 22:54–61
Eftekhari S, Warfvinge K, Blixt FW et al (2013) Differentiation of nerve fibers storing CGRP and CGRP receptors in the peripheral trigeminovascular system. J Pain 14:1289–1303
De Logu F, Nassini R, Hegron A et al (2022) Schwann cell endosome CGRP signals elicit periorbital mechanical allodynia in mice. Nat Commun 13:646
Rees TA, Labastida-Ramírez A, Rubio-Beltrán E (2023) Calcitonin/PAC1 receptor splice variants: a blind spot in migraine research. Trends Pharmacol Sci 44:651–663
Edvinsson L (2017) The Trigeminovascular Pathway: Role of CGRP and CGRP Receptors in Migraine. Headache 57(Suppl 2):47–55
Kilinc E, Guerrero-Toro C, Zakharov A et al (2017) Serotonergic mechanisms of trigeminal meningeal nociception: Implications for migraine pain. Neuropharmacology 116:160–173
Della Pietra A, Gómez Dabó L, Mikulenka P et al (2024) Mechanosensitive receptors in migraine: a systematic review. J Headache Pain 25:6
Ghanizada H, Al-Karagholi MA, Walker CS et al (2021) Amylin analog pramlintide induces migraine-like attacks in patients. Ann Neurol 89:1157–1171
Labastida-Ramírez A, Runio-Beltran E, Holland PR, et al. (2024) Sexually dimorphic effects of Amylin 1 receptor activation in trigeminovascular neurons. bioRxiv. 2024.01.12.575235. https://doiorg.publicaciones.saludcastillayleon.es/10.1101/2024.01.12.575235
Rees TA, Russo AF, O’Carroll SJ et al (2022) CGRP and the calcitonin receptor are co-expressed in mouse, rat and human trigeminal ganglia neurons. Front Physiol 13:860037
Al-Karagholi MA (2023) Involvement of potassium channel signalling in migraine pathophysiology. Pharmaceuticals (Basel) 16:438
Fahrenkrug J, Goetzl EJ, Gozes I, et al (2023) VIP and PACAP receptors in GtoPdb v.2023.1. Available from: https://journals.ed.ac.uk/gtopdb-cite/article/view/8714. Accessed 12 July 2024
Harmar AJ, Fahrenkrug J, Gozes I et al (2012) Pharmacology and functions of receptors for vasoactive intestinal peptide and pituitary adenylate cyclase-activating polypeptide: IUPHAR review 1. Br J Pharmacol 166:4–17
Schytz HW, Birk S, Wienecke T et al (2009) PACAP38 induces migraine-like attacks in patients with migraine without aura. Brain 132:16–25
Ghanizada H, Al-Karagholi MA, Arngrim N et al (2020) PACAP27 induces migraine-like attacks in migraine patients. Cephalalgia 40:57–67
Rubio-Beltrán E, Correnti E, Deen M et al (2018) PACAP38 and PAC1 receptor blockade: a new target for headache? J Headache Pain 19:64
Tasma Z, Siow A, Harris PWR et al (2022) Characterisation of agonist signalling profiles and agonist-dependent antagonism at PACAP-responsive receptors: Implications for drug discovery. Br J Pharmacol 179:435–453
Olesen J (2024) Provocation of attacks to discover migraine signaling mechanisms and new drug targets: early history and future perspectives - a narrative review. J Headache Pain 25:105
Christiansen I, Thomsen LL, Daugaard D et al (1999) Glyceryl trinitrate induces attacks of migraine without aura in sufferers of migraine with aura. Cephalalgia 19:660–667
Lassen LH, Ashina M, Christiansen I et al (1997) Nitric oxide synthase inhibition in migraine. Lancet 349:401–402
Birk S, Kruuse C, Petersen KA et al (2004) The phosphodiesterase 3 inhibitor cilostazol dilates large cerebral arteries in humans without affecting regional cerebral blood flow. J Cereb Blood Flow Metab 24:1352–1358
Schankin CJ, Kruse LS, Reinisch VM et al (2010) Nitric oxide-induced changes in endothelial expression of phosphodiesterases 2, 3, and 5. Headache 50:431–441
Nordgaard JC, Kruse LS, Møller M et al (2014) Phosphodiesterases 3 and 5 express activity in the trigeminal ganglion and co-localize with calcitonin gene-related peptide. Cephalalgia 34:503–513
Guo S, Olesen J, Ashina M (2014) Phosphodiesterase 3 inhibitor cilostazol induces migraine-like attacks via cyclic AMP increase. Brain 137:2951–2959
Falkenberg K, Bjerg HR, Olesen J (2020) Subcutaneous sumatriptan reduces cilostazol induced headache in migraine patients. Cephalalgia 40:842–850
Younis S, Christensen CE, Toft NM et al (2019) Investigation of distinct molecular pathways in migraine induction using calcitonin gene-related peptide and sildenafil. Cephalalgia 39:1776–1788
Raffaelli B, Do TP, Ashina H et al (2024) Induction of cGMP-mediated migraine attacks is independent of CGRP receptor activation. Cephalalgia 44:3331024241259489
Silvestro M, Iannone LF, Orologio I et al (2023) Migraine Treatment: Towards New Pharmacological Targets. Int J Mol Sci 24:12268
Shyng S, Nichols CG (1997) Octameric stoichiometry of the KATP channel complex. J Gen Physiol 110:655–664
Kokoti L, Al-Karagholi MA, Zhuang ZA et al (2024) Non-vascular ATP-sensitive potassium channel activation does not trigger migraine attacks: a randomized clinical trial. Cephalalgia 44:3331024241248211
Dyhring T, Jansen-Olesen I, Christophersen P et al (2023) Pharmacological profiling of KATP channel modulators: an outlook for new treatment opportunities for migraine. Pharmaceuticals (Basel) 16:225
Al-Karagholi MA, Ghanizada H, Nielsen CAW et al (2021) Opening of ATP sensitive potassium channels causes migraine attacks with aura. Brain 144:2322–2332
Al-Karagholi MA, Hansen JM, Guo S et al (2019) Opening of ATP-sensitive potassium channels causes migraine attacks: a new target for the treatment of migraine. Brain 142:2644–2654
Al-Karagholi MA, Ghanizada H, Waldorff Nielsen CA et al (2021) Opening of BKCa channels causes migraine attacks: a new downstream target for the treatment of migraine. Pain 162:2512–2520
Labastida-Ramírez A, Rubio-Beltrán E, Haanes KA et al (2020) Lasmiditan inhibits calcitonin gene-related peptide release in the rodent trigeminovascular system. Pain 161:1092–1099
Classey JD, Bartsch T, Goadsby PJ (2010) Distribution of 5-HT(1B), 5-HT(1D) and 5-HT(1F) receptor expression in rat trigeminal and dorsal root ganglia neurons: relevance to the selective anti-migraine effect of triptans. Brain Res 1361:76–85
Iannone LF, De Logu F, Geppetti P et al (2022) The role of TRP ion channels in migraine and headache. Neurosci Lett 768:136380
Messina R, Cetta I, Colombo B et al (2022) Tracking the evolution of non-headache symptoms through the migraine attack. J Headache Pain 23:149
Dodick DW (2018) A phase-by-phase review of migraine pathophysiology. Headache 58(Suppl 1):4–16
Vincent M, Viktrup L, Nicholson RA et al (2022) The not so hidden impact of interictal burden in migraine: a narrative review. Front Neurol 13:1032103
Gago-Veiga AB, Vivancos J, Sobrado M (2021) The premonitory phase: A crucial stage in migraine. Neurologia (Engl Ed) 36:298–304
Eigenbrodt AK, Christensen RH, Ashina H et al (2022) Premonitory symptoms in migraine: a systematic review and meta-analysis of observational studies reporting prevalence or relative frequency. J Headache Pain 23:140
Karsan N, Goadsby PJ (2023) Neuroimaging in the pre-ictal or premonitory phase of migraine: a narrative review. J Headache Pain 24:106
Lipton RB, Lanteri-Minet M, Leroux E et al (2023) Pre- and post-headache phases of migraine: multi-country results from the CaMEO - International Study. J Headache Pain 24:151
May A (2017) Understanding migraine as a cycling brain syndrome: reviewing the evidence from functional imaging. Neurol Sci 38(Suppl 1):125–130
Lauritzen M (1994) Pathophysiology of the migraine aura. The spreading depression theory. Brain 117:199–210
Hadjikhani N, Sanchez Del Rio M, Wu O et al (2001) Mechanisms of migraine aura revealed by functional MRI in human visual cortex. Proc Natl Acad Sci U S A 98:4687–4692
Lee MJ, Park BY, Cho S et al (2019) Dynamic functional connectivity of the migraine brain: a resting-state functional magnetic resonance imaging study. Pain 160:2776–2786
Karatas H, Erdener SE, Gursoy-Ozdemir Y et al (2013) Spreading depression triggers headache by activating neuronal Panx1 channels. Science 339:1092–1095
Noseda R, Burstein R (2013) Migraine pathophysiology: anatomy of the trigeminovascular pathway and associated neurological symptoms, cortical spreading depression, sensitization, and modulation of pain. Pain 154(Suppl 1):S44-53
Pietrobon D, Moskowitz MA (2013) Pathophysiology of migraine. Annu Rev Physiol 75:365–391
Vila-Pueyo M, Hoffmann J, Romero-Reyes M et al (2019) Brain structure and function related to headache: Brainstem structure and function in headache. Cephalalgia 39:1635–1660
Christensen RH, Eigenbrodt AK, Ashina H et al (2023) What proportion of people with migraine report postdromal symptoms? A systematic review and meta-analysis of observational studies. Cephalalgia 43:3331024231206376
Thuraiaiyah J, Ashina H, Christensen RH et al (2024) Postdromal symptoms in migraine: a REFORM study. J Headache Pain 25:25
Brighina F, Cosentino G, Fierro B (2016) Habituation or lack of habituation: What is really lacking in migraine? Clin Neurophysiol 127:19–20
Di Antonio S, Castaldo M, Ponzano M et al (2021) Disability, burden, and symptoms related to sensitization in migraine patients associate with headache frequency. Scand J Pain 21:766–777
Han SM, Kim KM, Cho SJ et al (2021) Prevalence and characteristics of cutaneous allodynia in probable migraine. Sci Rep 11:2467
Coppola G, Parisi V, Di Renzo A et al (2020) Cortical pain processing in migraine. J Neural Transm (Vienna) 127:551–566
Aguggia M, Saracco MG, Cavallini M et al (2013) Sensitization and pain. Neurol Sci 34(Suppl 1):S37-40
Burstein R, Jakubowski M, Garcia-Nicas E et al (2010) Thalamic sensitization transforms localized pain into widespread allodynia. Ann Neurol 68:81–91
Martinelli D, Pocora MM, De Icco R et al (2022) Triggers of migraine: where do we stand? Curr Opin Neurol 35:360–366
Fischer-Schulte LH (2023) Peng KP (2023) Migraine prodromes and migraine triggers. Handb Clin Neurol 198:135–148
Russo A, Tessitore A, Silvestro M et al (2019) Advanced visual network and cerebellar hyperresponsiveness to trigeminal nociception in migraine with aura. J Headache Pain 20:46
Burke MJ, Joutsa J, Cohen AL et al (2020) Mapping migraine to a common brain network. Brain 143:541–553
Hadjikhani N, Del Rio MS, Wu O et al (2001) Mechanisms of migraine aura revealed by functional MRI in human visual cortex. Proc Natl Acad Sci 98:4687–4692
Maniyar FH, Sprenger T, Schankin C et al (2014) Photic hypersensitivity in the premonitory phase of migraine – a positron emission tomography study. Eur J Neurol 21:1178–1183
Puledda F, Ffytche D, O’Daly O et al (2019) Imaging the Visual Network in the Migraine Spectrum. Front Neurol 10:1325
Schulte LH, May A (2016) The migraine generator revisited: continuous scanning of the migraine cycle over 30 days and three spontaneous attacks. Brain 139:1987–1993
Schulte LH, Mehnert J, May A (2020) Longitudinal neuroimaging over 30 days: temporal characteristics of migraine. Ann Neurol 87:646–651
Schwedt TJ, Chiang C-C, Chong CD et al (2015) Functional MRI of migraine. Lancet Neurol 14:81–91
Moulton EA, Becerra L, Maleki N et al (2011) Painful heat reveals hyperexcitability of the temporal pole in interictal and ictal migraine States. Cereb Cortex 21:435–448
Schwedt TJ, Chong CD, Chiang CC et al (2014) Enhanced pain-induced activity of pain-processing regions in a case-control study of episodic migraine. Cephalalgia 34:947–958
Kim JH, Kim S, Suh S-I et al (2010) Interictal metabolic changes in episodic migraine: a voxel-based FDG-PET study. Cephalalgia 30:53–61
Tessitore A, Russo A, Conte F et al (2015) Abnormal connectivity within executive resting-state network in migraine with aura. Headache 55:794–805
Chong CD, Gaw N, Fu Y et al (2017) Migraine classification using magnetic resonance imaging resting-state functional connectivity data. Cephalalgia 37:828–844
Szabo N, Kincses ZT, Párdutz Á et al (2012) White matter microstructural alterations in migraine: a diffusion-weighted MRI study. Pain 153:651–656
Amin FM, Hougaard A, Magon S et al (2016) Change in brain network connectivity during PACAP38-induced migraine attacks: a resting-state functional MRI study. Neurology 86:180–187
Ellingson BM, Hesterman C, Johnston M et al (2019) Advanced imaging in the evaluation of migraine headaches. Neuroimaging Clin N Am 29:301–324
Karsan N, Silva E, Goadsby PJ (2023) Evaluating migraine with typical aura with neuroimaging. Front Hum Neurosci 17:1112790
Gaist D, Hougaard A, Garde E et al (2018) Migraine with visual aura associated with thicker visual cortex. Brain 141:776–785
Bridge H, Stagg CJ, Near J et al (2015) Altered neurochemical coupling in the occipital cortex in migraine with visual aura. Cephalalgia 35:1025–1030
Tedeschi G, Russo A, Conte F et al (2016) Increased interictal visual network connectivity in patients with migraine with aura. Cephalalgia 36:139–147
Datta R, Aguirre GK, Hu S et al (2013) Interictal cortical hyperresponsiveness in migraine is directly related to the presence of aura. Cephalalgia 33:365–374
Faragó P, Tóth E, Kocsis K et al (2019) Altered resting state functional activity and microstructure of the white matter in migraine with aura. Front Neurol 10:1039
Hougaard A, Amin F, Larsson H et al (2017) Increased intrinsic brain connectivity between pons and somatosensory cortex during attacks of migraine with aura. Hum Brain Mapp 38:2635–2642
Coppola G, Corbelli I, Di Renzo A et al (2022) Visual stimulation and frequency of focal neurological symptoms engage distinctive neurocognitive resources in migraine with aura patients: a study of resting-state functional networks. J Headache Pain 23:80
Lauritzen M, Olsen TS, Lassen NA et al (1983) Changes in regional cerebral blood flow during the course of classic migraine attacks. Ann Neurol 13:633–641
Fu T, Liu L, Huang X et al (2022) Cerebral blood flow alterations in migraine patients with and without aura: an arterial spin labeling study. J Headache Pain 23:131
Christensen RH, Gollion C, Amin FM et al (2022) Imaging the inflammatory phenotype in migraine. J Headache Pain 23:60
Silvestro M, Tessitore A, Di Nardo F et al (2022) Functional connectivity changes in complex migraine aura: beyond the visual network. Eur J Neurol 29:295–304
Petrusic I, Viana M, Dakovic M et al (2019) Application of the migraine aura complexity score (MACS): clinical and neuroimaging study. Front Neurol 10:1112
Coppola G, Di Renzo A, Tinelli E et al (2021) Thalamo-cortical networks in subtypes of migraine with aura patients. J Headache Pain 22:58
Abagnale C, Di Renzo A, Sebastianelli G et al (2023) Whole brain surface-based morphometry and tract-based spatial statistics in migraine with aura patients: difference between pure visual and complex auras. Front Hum Neurosci 17:1146302
Mitrović K, Petrušić I, Radojičić A et al (2023) Migraine with aura detection and subtype classification using machine learning algorithms and morphometric magnetic resonance imaging data. Front Neurol 14:1106612
Mitrović K, Savić AM, Radojičić A (2023) Machine learning approach for Migraine Aura Complexity Score prediction based on magnetic resonance imaging data. J Headache Pain 24:169
Lee MJ, Park BY, Cho S et al (2019) Increased connectivity of pain matrix in chronic migraine: a resting-state functional MRI study. J Headache Pain 20:29
Coppola G, Petolicchio B, Di Renzo A et al (2017) Cerebral gray matter volume in patients with chronic migraine: correlations with clinical features. J Headache Pain 18:115
Lai TH, Chou KH, Fuh JL et al (2016) Gray matter changes related to medication overuse in patients with chronic migraine. Cephalalgia 36:1324–1333
Li X, Liu M, Fan W et al (2023) Altered cerebral neurovascular coupling in medication-overuse headache: a study combining multi-modal resting-state fMRI with 3D PCASL. Front Neurosci 17:1139086
Russo A, Coppola G, Pierelli F et al (2018) Pain perception and migraine. Front Neurol 9:576
Sandrini G, Rossi P, Milanov I et al (2006) Abnormal modulatory influence of diffuse noxious inhibitory controls in migraine and chronic tension-type headache patients. Cephalalgia 26:782–789
Morrison DP (1990) Abnormal perceptual experiences in migraine. Cephalalgia 10:273–277
Shechter A, Stewart WF, Silberstein SD et al (2002) Migraine and autonomic nervous system function: a population-based, case-control study. Neurology 58:422–427
Koenig J, Williams DP, Kemp AH et al (2016) Vagally mediated heart rate variability in headache patients–a systematic review and meta-analysis. Cephalalgia 36:265–278
Chuang CH, Li JY, King JT et al (2023) Abnormal heart rate variability and its application in predicting treatment efficacy in patients with chronic migraine: an exploratory study. Cephalalgia 43:3331024231206781
Hvedstrup J, Kolding LT, Ashina M et al (2020) Increased neck muscle stiffness in migraine patients with ictal neck pain: a shear wave elastography study. Cephalalgia 40:565–574
Do TP, Heldarskard GF, Kolding LT et al (2018) Myofascial trigger points in migraine and tension-type headache. J Headache Pain 19:84
Leistad RB, Sand T, Westgaard RH et al (2006) Stress-induced pain and muscle activity in patients with migraine and tension-type headache. Cephalalgia 26:64–73
Luedtke K, Mehnert J, May A (2018) Altered muscle activity during rest and during mental or physical activity is not a trait symptom of migraine - a neck muscle EMG study. J Headache Pain 19:26
Zhang X, Levy D, Kainz V et al (2011) Activation of central trigeminovascular neurons by cortical spreading depression. Ann Neurol 69:855–865
Costa C, Tozzi A, Rainero I et al (2013) Cortical spreading depression as a target for anti-migraine agents. J Headache Pain 14:62
Coppola G, Di Lorenzo C, Schoenen J et al (2013) Habituation and sensitization in primary headaches. J Headache Pain 14:65
Coppola G, Pierelli F, Schoenen J (2009) Habituation and migraine. Neurobiol Learn Mem 92:249–259
Chen WT, Hsiao FJ, Ko YC et al (2018) Comparison of somatosensory cortex excitability between migraine and “strict-criteria” tension-type headache: a magnetoencephalographic study. Pain 159:793–803
Abagnale C, Ranieri F, Di Renzo A et al (2021) Impaired short-term visual paired associative plasticity in patients with migraine between attacks. Pain 162:803–810
Puledda F, Vigano A, Sebastianelli G et al (2023) Electrophysiological findings in migraine may reflect abnormal synaptic plasticity mechanisms: a narrative review. Cephalalgia 43:3331024231195780
Hsiao FJ, Chen WT, Liu HY et al (2021) Migraine chronification is associated with beta-band connectivity within the pain-related cortical regions: a magnetoencephalographic study. Pain 162:2590–2598
Hsiao FJ, Chen WT, Pan LH et al (2022) Resting-state magnetoencephalographic oscillatory connectivity to identify patients with chronic migraine using machine learning. J Headache Pain 23:130
Hsiao FJ, Chen WT, Wu YT et al (2023) Characteristic oscillatory brain networks for predicting patients with chronic migraine. J Headache Pain 24:139
Tessitore A, Russo A, Giordano A et al (2013) Disrupted default mode network connectivity in migraine without aura. J Headache Pain 14:89
Skorobogatykh K, van Hoogstraten WS, Degan D et al (2019) Functional connectivity studies in migraine: what have we learned? J Headache Pain 20:108
Schramm S, Borner C, Reichert M et al (2023) Functional magnetic resonance imaging in migraine: a systematic review. Cephalalgia 43:3331024221128278
Porcaro C, Di Lorenzo G, Seri S et al (2017) Impaired brainstem and thalamic high-frequency oscillatory EEG activity in migraine between attacks. Cephalalgia 37:915–926
Hsiao FJ, Chen WT, Pan LH et al (2022) Dynamic brainstem and somatosensory cortical excitability during migraine cycles. J Headache Pain 23:21
GBD (2016) Nervous System Disorders Collaborators (2019) Global, regional, and national burden of neurological disorders, 1990–2016: a systematic analysis for the Global Burden of Disease Study 2016. Lancet Neurol 18:459–480
GBD (2019) Nervous System Disorders Collaborators (2020) Global burden of 369 diseases and injuries in 204 countries and territories, 1990–2019: a systematic analysis for the Global Burden of Disease Study 2019. Lancet 396:1204–1222
Rosignoli C, Ornello R, Onofri A et al (2022) Applying a biopsychosocial model to migraine: rationale and clinical implications. J Headache Pain 23:100
Eigenbrodt AK, Ashina H, Khan S et al (2021) Diagnosis and management of migraine in ten steps. Nat Revi Neurol 17:501–514
Leppert MH, Poisson SN, Scarbro S et al (2024) Association of traditional and nontraditional risk factors in the development of strokes among young adults by sex and age group: a retrospective case-control study. Circ Cardiovasc Qual Outcomes 17:e010307
Kurth T, Chabriat H, Bousser MG (2012) Migraine and stroke: a complex association with clinical implications. Lancet Neurol 11:92–100
Kruit MC, Launer LJ, Ferrari MD et al (2005) Infarcts in the posterior circulation territory in migraine. The population-based MRI CAMERA study. Brain 128:2068–2077
Kruit MC, van Buchem MA, Hofman PA et al (2004) Migraine as a risk factor for subclinical brain lesions. JAMA 291:427–434
Kruit MC, van Buchem MA, Launer LJ et al (2010) Migraine is associated with an increased risk of deep white matter lesions, subclinical posterior circulation infarcts and brain iron accumulation: the population-based MRI CAMERA study. Cephalalgia 30:129–136
Iwasaki A, Suzuki K, Takekawa H et al (2017) The relationship between right-to-left shunt and brain white matter lesions in Japanese patients with migraine: a single center study. J Headache Pain 18:3
Zhao Q, Liu R, Zhou J et al (2021) Prevalence and grade of RLS in migraine: A prospective study of 251 migraineurs by synchronous test of c-TTE and c-TCD. Medicine (Baltimore) 100:e24175
Meilán A, Larrosa D, Ramón C et al (2020) No association between migraine frequency, white matter lesions and silent brain infarctions: a study in a series of women with chronic migraine. Eur J Neurol 27:1689–1696
Adelborg K, Szépligeti SK, Holland-Bill L et al (2018) Migraine and risk of cardiovascular diseases: Danish population based matched cohort study. BMJ 360:k96
Guey S, Mawet J, Hervé D et al (2016) Prevalence and characteristics of migraine in CADASIL. Cephalalgia 36:1038–1047
Rist PM, Winter AC, Buring JE et al (2018) Migraine and the risk of incident hypertension among women. Cephalalgia 38:1817–1824
Scher AI, Terwindt GM, Picavet HS et al (2005) Cardiovascular risk factors and migraine: the GEM population-based study. Neurology 64:614–620
Øie LR, Kurth T, Gulati S et al (2020) Migraine and risk of stroke. J Neurol Neurosurg Psychiatry 91:593–604
Filippopulos FM, Schoeberl F, Becker HC et al (2019) Coronary artery calcification score in migraine patients. Sci Rep 9:14069
Wilson SL, Poulter NR (2006) The effect of non-steroidal anti-inflammatory drugs and other commonly used non-narcotic analgesics on blood pressure level in adults. J Hypertens 24:1457–1469
Petersen CL, Hougaard A, Gaist D et al (2024) Risk of Stroke and Myocardial Infarction Among Initiators of Triptans. JAMA Neurol 81:248–254
Mulder IA, Li M, de Vries T et al (2020) Anti-migraine calcitonin gene-related peptide receptor antagonists worsen cerebral ischemic outcome in mice. Ann Neurol 88:771–784
Wang K, Fenton BT, Dao VX et al (2023) Trajectory of blood pressure after initiating anti-calcitonin gene-related peptide treatment of migraine: a target trial emulation from the veterans health administration. J Headache Pain 24:108
Battle DE (2013) Diagnostic and Statistical Manual of Mental Disorders (DSM). Codas 25:191–192
Kendler KS (1997) The genetic epidemiology of psychiatric disorders: a current perspective. Soc Psychiatry Psychiatr Epidemiol 32:5–11
Merrill RM, Gibbons IS (2023) Comorbidity of sleep disorders, mental illness, and migraine or headaches. SN Compr Clin Med 5:283
Dresler T, Caratozzolo S, Guldolf K et al (2019) Understanding the nature of psychiatric comorbidity in migraine: a systematic review focused on interactions and treatment implications. J Headache Pain 20:51
Minen MT, Begasse De Dhaem O et al (2016) Migraine and its psychiatric comorbidities. J Neurol Neurosurg Psychiatry 87:741–749
Giri S, Tronvik EA, Hagen K (2022) The bidirectional temporal relationship between headache and affective disorders: longitudinal data from the HUNT studies. J Headache Pain 23:14
Burch RC, Buse DC, Lipton RB (2019) Migraine: epidemiology, burden, and comorbidity. Neurol Clin 37:631–649
Lampl C, Thomas H, Tassorelli C et al (2016) Headache, depression and anxiety: associations in the Eurolight project. J Headache Pain 17:59
Liu HY, Chou KH, Chen WT (2018) Migraine and the Hippocampus. Curr Pain Headache Rep 22:13
Burch R (2019) Antidepressants for preventive treatment of migraine. Current Treat Options Neurol 21:18
Jannini TB, Lorenzo GD, Bianciardi E et al (2022) Off-label Uses of Selective Serotonin Reuptake Inhibitors (SSRIs). Curr Neuropharmacol 20:693–712
Bromis K, Calem M, Reinders A et al (2018) Meta-analysis of 89 structural MRI studies in posttraumatic stress disorder and comparison with major depressive disorder. Am J Psychiatry 175:989–998
Schoonman GG, Evers DJ, Terwindt GM et al (2006) (2006) The prevalence of premonitory symptoms in migraine: a questionnaire study in 461 patients. Cephalalgia 26:1209–1213
Giffin NJ, Ruggiero L, Lipton RB et al (2003) Premonitory symptoms in migraine: an electronic diary study. Neurology 60:935–940
Gago-Veiga AB, Pagán J, Henares K et al (2018) To what extent are patients with migraine able to predict attacks? J Pain Res 11:2083–2094
Xie YJ, Lin M, Wong YT et al (2022) Migraine attacks and relevant trigger factors in undergraduate nursing students in hong kong: a cross-sectional study. J Pain Res 15:701–713
Lipton RB, Pavlovic JM, Haut SR et al (2014) Methodological issues in studying trigger factors and premonitory features of migraine. Headache 54:1661–1669
Schulte LH, Jürgens TP, May A (2015) Photo-, osmo- and phonophobia in the premonitory phase of migraine: mistaking symptoms for triggers? J Headache Pain 16:14
Wang X, Yin Z, Lian Y et al (2021) Premonitory symptoms in migraine from China: a multi-clinic study of 4821 patients. Cephalalgia 41:991–1003
Ferrari MD, Goadsby PJ, Burstein R et al (2022) Migraine Nat Rev Dis Primers 8:2
Puledda F, Silva EM, Suwanlaong K et al (2023) Migraine: from pathophysiology to treatment. J Neurol 270:3654–3666
Tu YH, Wang YF, Yuan H et al (2022) Most bothersome symptoms in patients with migraine: a hospital-based study in Taiwan. Headache 62:596–603
Munjal S, Singh P, Reed ML et al (2020) Most Bothersome symptom in persons with migraine: results from the Migraine in America Symptoms and Treatment (MAST) Study. Headache 60:416–429
Leao AAP (1944) Spreading depression of activity in the cerebral cortex. J Neurophysiol 7:359–390
Gerstein MT, McGinley JS, Houts CR et al (2022) Non-ICHD-3 defning migraine symptoms by phase of the migraine cycle: results of the MiCOAS qualitative study. Headache 62(suppl 1):25
Bose P, Goadsby PJ (2016) The migraine postdrome. Curr Opin Neurol 29:299–301
Karsan N, Bose P, Goadsby PJ (2018) The migraine postdrome. Continuum (Minneap Minn) 24:996–1008
Recobe A (2021) Pathophysiology of migraine. Continuum (Minneap Minn) 27:586–596
Lipton RB, Dodick DW, Ailani J et al (2019) Effect of ubrogepant vs placebo on pain and the most bothersome associated symptom in the acute treatment of migraine: the ACHIEVE II randomized clinical trial. JAMA 322:1887–1898
Villalón CM, VanDenBrink AM (2017) The role of 5-hydroxytryptamine in the pathophysiology of migraine and its relevance to the design of novel treatments. Mini Rev Med Chem 17:928–938
Rubio-Beltrán E, Labastida-Ramírez A, Villalón CM et al (2018) Is selective 5-HT(1F) receptor agonism an entity apart from that of the triptans in antimigraine therapy? Pharmacol Ther 186:88–97
Doenicke A, Brand J, Perrin VL (1988) Possible benefit of GR43175, a novel 5-HT1-like receptor agonist, for the acute treatment of severe migraine. Lancet 1:1309–1311
Rubio-Beltrán E, Labastida-Ramírez A, Haanes KA et al (2019) Characterization of binding, functional activity, and contractile responses of the selective 5-HT(1F) receptor agonist lasmiditan. Br J Pharmacol 176:4681–4695
Loder E (2010) Triptan therapy in migraine. N Engl J Med 363:63–70
Benemei S, Cortese F, Labastida-Ramírez A et al (2017) Triptans and CGRP blockade - impact on the cranial vasculature. J Headache Pain 18:103
Cameron C, Kelly S, Hsieh SC et al (2015) Triptans in the acute treatment of migraine: a systematic review and network meta-analysis. Headache 55(Suppl 4):221–235
Ferrari MD, Roon KI, Lipton RB et al (2001) Oral triptans (serotonin 5-HT(1B/1D) agonists) in acute migraine treatment: a meta-analysis of 53 trials. Lancet 358:1668–1675
VanderPluym JH, Halker Singh RB, Urtecho M et al (2021) Acute treatments for episodic migraine in adults: a systematic review and meta-analysis. JAMA 325:2357–2369
Nappi G, Sandrini G, Sances G (2003) Tolerability of the triptans: clinical implications. Drug Saf 26:93–107
Roberto G, Raschi E, Piccinni C et al (2015) Adverse cardiovascular events associated with triptans and ergotamines for treatment of migraine: systematic review of observational studies. Cephalalgia 35:118–131
Jamieson DG (2002) The safety of triptans in the treatment of patients with migraine. Am J Med 112:135–140
Mitsikostas DD, Waeber C, Sanchez-Del-Rio M et al (2023) The 5-HT(1F) receptor as the target of ditans in migraine - from bench to bedside. Nat Rev Neurol 19:489–505
Mecklenburg J, Raffaelli B, Neeb L et al (2020) The potential of lasmiditan in migraine. Ther Adv Neurol Disord 13:1756286420967847
Raffaelli B, Israel H, Neeb L et al (2017) The safety and efficacy of the 5-HT 1F receptor agonist lasmiditan in the acute treatment of migraine. Expert Opin Pharmacother 18:1409–1415
Goadsby PJ, Duckworth JW (1987) Effect of stimulation of trigeminal ganglion on regional cerebral blood flow in cats. Am J Physiol 253:270–274
Goadsby PJ, Edvinsson L, Ekman R (1990) Vasoactive peptide release in the extracerebral circulation of humans during migraine headache. Ann Neurol 28:183–187
Levy D, Labastida-Ramirez A, MaassenVanDenBrink A (2019) Current understanding of meningeal and cerebral vascular function underlying migraine headache. Cephalalgia 39:1606–1622
Edvinsson L, Haanes KA, Warfvinge K (2018) CGRP as the target of new migraine therapies - successful translation from bench to clinic. Nat Rev Neurol 14:338–350
Olesen J, Diener H-C, Husstedt IW et al (2004) Calcitonin gene–related peptide receptor antagonist BIBN 4096 BS for the acute treatment of migraine. N Engl J Med 350:1104–1110
Hargreaves R, Olesen J (2019) Calcitonin gene-related peptide modulators - the history and renaissance of a new migraine drug class. Headache 59:951–970
Messina R, Huessler EM, Puledda F et al (2023) Safety and tolerability of monoclonal antibodies targeting the CGRP pathway and gepants in migraine prevention: a systematic review and network meta-analysis. Cephalalgia 43:3331024231152169
Labastida-Ramírez A, Caronna E, Gollion C et al (2023) Mode and site of action of therapies targeting CGRP signaling. J Headache Pain 24:125
Garelja ML, Walker CS, Hay DL (2022) CGRP receptor antagonists for migraine. Are they also AMY(1) receptor antagonists? Br J Pharmacol 179:454–459
Edvinsson L, Grell AS, Warfvinge K (2020) Expression of the CGRP family of neuropeptides and their receptors in the trigeminal ganglion. J Mol Neurosci 70:930–944
Yang CP, Liang CS, Chang CM et al (2021) Comparison of new pharmacologic agents with triptans for treatment of migraine: a systematic review and meta-analysis. JAMA Netw Open 4:e2128544
Puledda F, Younis S, Huessler E-M et al (2023) Efficacy, safety and indirect comparisons of lasmiditan, rimegepant, and ubrogepant for the acute treatment of migraine: a systematic review and network meta-analysis of the literature. Cephalalgia 43:03331024231151419
Li D, Abreu J, Tepper SJ (2023) A brief review of gepants. Curr Pain Headache Rep 27:479–488
Maasumi K, Michael RL, Rapoport AM (2018) CGRP and migraine: the role of blocking calcitonin gene-related peptide ligand and receptor in the management of migraine. Drugs 78:913–928
Tajti J, Uddman R, Möller S et al (1999) Messenger molecules and receptor mRNA in the human trigeminal ganglion. J Auton Nerv Syst 76:176–183
Ho TW, Edvinsson L, Goadsby PJ (2010) CGRP and its receptors provide new insights into migraine pathophysiology. Nat Rev Neurol 6:573–582
Tfelt-Hansen P, Le H (2009) Calcitonin gene-related peptide in blood: is it increased in the external jugular vein during migraine and cluster headache? A review. J Headache Pain 10:137–143
Hansen JM, Hauge AW, Olesen J et al (2010) Calcitonin gene-related peptide triggers migraine-like attacks in patients with migraine with aura. Cephalalgia 30:1179–1186
Ashina M (2020) Migraine. N Engl J Med 383:1866–1876
Lipton RB, Croop R, Stock EG et al (2019) Rimegepant, an oral calcitonin gene-related peptide receptor antagonist, for migraine. N Engl J Med 381:142–149
Croop R, Lipton RB, Kudrow D et al (2021) Oral rimegepant for preventive treatment of migraine: a phase 2/3, randomised, double-blind, placebo-controlled trial. Lancet 397:51–60
Ailani J, Lipton RB, Goadsby PJ et al (2021) Atogepant for the preventive treatment of migraine. N Engl J Med 385:695–706
Pozo-Rosich P, Ailani J, Ashina M et al (2023) Atogepant for the preventive treatment of chronic migraine (PROGRESS): a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet 402:775–785
Lipton RB, Croop R, Stock DA et al (2023) Safety, tolerability, and efficacy of zavegepant 10 mg nasal spray for the acute treatment of migraine in the USA: a phase 3, double-blind, randomised, placebo-controlled multicentre trial. Lancet Neurol 22:209–217
Haghdoost F, Puledda F, Garcia-Azorin D et al (2023) Evaluating the efficacy of CGRP mAbs and gepants for the preventive treatment of migraine: a systematic review and network meta-analysis of phase 3 randomised controlled trials. Cephalalgia 43:3331024231159366
Schwedt TJ, Myers Oakes TM, Martinez JM et al (2024) Comparing the efficacy and safety of galcanezumab versus rimegepant for prevention of episodic migraine: results from a randomized, controlled clinical trial. Neurol Ther 13:85–105
Hong JB, Lange KS, Overeem LH et al (2023) A Scoping Review and Meta-Analysis of Anti-CGRP Monoclonal Antibodies: Predicting Response. Pharmaceuticals (Basel) 16:934
Raffaelli B, Fitzek M, Overeem LH et al (2023) Clinical evaluation of super-responders vs. non-responders to CGRP(-receptor) monoclonal antibodies: a real-world experience. J Headache Pain 24:16
Barbanti P, Aurilia C, Egeo G et al (2023) Late response to anti-CGRP monoclonal antibodies in migraine: a multicenter prospective observational study. Neurology 101:482–488
Waliszewska-Prosół M, Vuralli D, Martelletti P (2023) What to do with non-responders to CGRP(r) monoclonal antibodies: switch to another or move to gepants? J Headache Pain 24:163
Miyata A, Arimura A, Dahl RR et al (1989) Isolation of a novel 38 residue-hypothalamic polypeptide which stimulates adenylate cyclase in pituitary cells. Biochem Biophys Res Commun 164:567–574
Miyata A, Jiang L, Dahl RD et al (1990) 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 170:643–648
Vaudry D, Falluel-Morel A, Bourgault S et al (2009) Pituitary adenylate cyclase-activating polypeptide and its receptors: 20 years after the discovery. Pharmacol Rev 61:283–357
Moller K, Zhang YZ, Håkanson R et al (1993) Pituitary adenylate cyclase activating peptide is a sensory neuropeptide: immunocytochemical and immunochemical evidence. Neuroscience 57:725–732
Mulder H, Uddman R, Moller K et al (1994) Pituitary adenylate cyclase activating polypeptide expression in sensory neurons. Neuroscience 63:307–312
Uddman R, Tajti J, Möller S et al (1999) Neuronal messengers and peptide receptors in the human sphenopalatine and otic ganglia. Brain Res 826:193–199
Uddman R, Tajti J, Hou M et al (2002) Neuropeptide expression in the human trigeminal nucleus caudalis and in the cervical spinal cord C1 and C2. Cephalalgia 22:112–116
Rawlings SR (1994) PACAP, PACAP receptors, and intracellular signalling. Mol Cell Endocrinol 101:C5–C9
Knutsson M, Edvinsson L (2002) Distribution of mRNA for VIP and PACAP receptors in human cerebral arteries and cranial ganglia. NeuroReport 13:507–509
Chan KY, Baun M, De Vries R et al (2011) Pharmacological characterization of VIP and PACAP receptors in the human meningeal and coronary artery. Cephalalgia 31:181–189
Waschek JA, Baca SM, Akerman S (2018) PACAP and migraine headache: immunomodulation of neural circuits in autonomic ganglia and brain parenchyma. J Headache Pain 19:23
Wienholtz NKF, Christensen CE, Zhang DG et al (2021) Early treatment with sumatriptan prevents PACAP38-induced migraine: a randomised clinical trial. Cephalalgia 41:731–748
Tuka B, Helyes Z, Markovics A et al (2013) Alterations in PACAP-38-like immunoreactivity in the plasma during ictal and interictal periods of migraine patients. Cephalalgia. 33:1085–1095
Zagami AS, Edvinsson L, Goadsby PJ (2014) Pituitary adenylate cyclase activating polypeptide and migraine. Ann Clin Transl Neurol 1:1036–1040
Guo S, Vollesen ALH, Hansen YB et al (2017) Part II: biochemical changes after pituitary adenylate cyclase-activating polypeptide-38 infusion in migraine patients. Cephalalgia 37:136–147
Sbei S, Moncrief T, Limjunyawong N et al (2023) PACAP activates MRGPRX2 on meningeal mast cells to drive migraine-like pain. Sci Rep 13:12302
Ashina M, Doležil D, Bonner JH et al (2021) A phase 2, randomized, double-blind, placebo-controlled trial of AMG 301, a pituitary adenylate cyclase-activating polypeptide PAC1 receptor monoclonal antibody for migraine prevention. Cephalalgia 41:33–44
Rasmussen NB, Deligianni C, Christensen CE et al (2023) The effect of Lu AG09222 on PACAP38- and VIP-induced vasodilation, heart rate increase, and headache in healthy subjects: an interventional, randomized, double-blind, parallel-group, placebo-controlled study. J Headache Pain 24:60
Grazzi L, Sansone M, Rizzoli P (2018) A short review of the Non-invasive transcutaneous pericranial electrical stimulation techniques and their application in headache. Curr Pain Head Rep 22:4
Tiwari V, Agrawal S (2022) Migraine and neuromodulation: a literature review. Cureus 14:e31223
Viganò A, Toscano M, Puledda F et al (2019) Treating chronic migraine with neuromodulation: the role of neurophysiological abnormalities and maladaptive plasticity. Front Pharmacol 10:32
Hallett M (2007) Transcranial magnetic stimulation: a primer. Neuron 55:187–199
Andreou AP, Holland PR, Akerman S et al (2016) Transcranial magnetic stimulation and potential cortical and trigeminothalamic mechanisms in migraine. Brain 139:2002–2014
Cosentino G, Fierro B, Vigneri S et al (2014) Cyclical changes of cortical excitability and metaplasticity in migraine: evidence from a repetitive transcranial magnetic stimulation study. Pain 155:1070–1078
Moisset X, Pereira B, Ciampi De Andrade D et al (2020) Neuromodulation techniques for acute and preventive migraine treatment: a systematic review and meta-analysis of randomized controlled trials. J Headache Pain 21:142
Shen M, Li C, Wei X et al (2023) Transcranial magnetic stimulation as a therapy for migraine: an overview of systematic reviews. J Pain Res 16:3133–3144
Lipton RB, Dodick DW, Silberstein SD et al (2010) Single-pulse transcranial magnetic stimulation for acute treatment of migraine with aura: a randomised, double-blind, parallel-group, sham-controlled trial. Lancet Neurol 9:373–380
De Couck M, Nijs J, Gidron Y (2014) You may need a nerve to treat pain: the neurobiological rationale for vagal nerve activation in pain management. Clin J Pain 30:1099–1105
Henssen DJHA, Derks B, van Doorn M et al (2019) Vagus nerve stimulation for primary headache disorders: an anatomical review to explain a clinical phenomenon. Cephalalgia 39:1180–1194
Grazzi L, Tassorelli C, de Tommaso M et al (2018) Practical and clinical utility of non-invasive vagus nerve stimulation (nVNS) for the acute treatment of migraine: a post hoc analysis of the randomized, sham-controlled, double-blind PRESTO trial. J Headache Pain 19:98
Tassorelli C, Grazzi L, de Tommaso M et al (2018) Noninvasive vagus nerve stimulation as acute therapy for migraine: the randomized PRESTO study. Neurology 91:e364–e373
Grazzi L, Egeo G, Calhoun AH et al (2016) Non-invasive Vagus Nerve Stimulation (nVNS) as mini-prophylaxis for menstrual/menstrually related migraine: an open-label study. J Headache Pain 17:91
Song D, Li P, Wang Y et al (2023) Noninvasive vagus nerve stimulation for migraine: a systematic review and meta-analysis of randomized controlled trials. Front Neurol 14:1190062
Miller S, Sinclair AJ, Davies B et al (2016) Neurostimulation in the treatment of primary headaches. Pract Neurol 16:362–375
Ordas CM, Cuadrado ML, Pareja JA et al (2020) Transcutaneous supraorbital stimulation as a preventive treatment for chronic migraine: a prospective. Open-Label Study Pain Med 21:415–422
Reed KL (2013) Peripheral neuromodulation and headaches: clinical approach and considerations on underlying mechanisms. Curr Pain Head Rep 217:305
Magis D (2015) Neuromodulation in migraine: state of the art and perspectives. Expert Rev Med Devices 12:329–339
Subramonian A, Argaez C (2020) Non-invasive Nerve Stimulation Modalities for Migraine Pain: A Review of Clinical Effectiveness and Cost-effectiveness. Ottawa (ON): Canadian Agency for Drugs and Technologies in Health
Thair H, Holloway AL, Newport R et al (2017) Transcranial Direct Current Stimulation (tDCS): a beginner’s guide for designe and implementation. Front Neurosci 11:641
Vigneri S, Bonventre S, Inviati A et al (2014) Effects of transcranial direct current stimulation on esophageal motility in patients with gastroesophageal reflux disease. Clin Neurophysiol 125:1840–1846
Cai G, Xia Z, Charvet L et al (2021) A systematic review and meta-analysis on the efficacy of repeated transcranial direct current stimulation for migraine. J Pain Res 14:1171–1183
Hong P, Liu Y, Wan Y et al (2022) Transcranial direct current stimulation for migraine: a systematic review and meta-analysis of randomized controlled trials. CNS Neurosci Ther 28:992–998
Elyamany O, Leicht G, Herrmann CS et al (2021) Transcranial alternating current stimulation (tACS): From basic mechanisms towards first application in psychiatry. Eur Arch Psychiatry Clin Neurosci 271:135–156
Antal A, Bischoff R, Stephani C et al (2020) Low intensity, transcranial, alternating current stimulation reduces migraine attack burden in a home application set-up: a double-blinded, randomized feasibility study. Brain Sci 10:888
Weeks RE (2013) Application of behavioral therapies in adult and adolescent patients with chronic migraine. Ital J Neurol Sci 34(Suppl 1):S11–S17
Faedda N, Cerutti R, Verdecchia P et al (2016) Behavioral management of headache in children and adolescents. J Headache Pain 17:80
Kabbouche MA, Gilman DK (2008) Management of migraine in adolescents. Neuropsychiatr Dis Treat 4:535–548
Harris P, Loveman E, Clegg A et al (2015) Systematic review of cognitive behavioural therapy for the management of headaches and migraines in adults. Br J Pain 2015:9213–9224
Kroner JW, Peugh J, Kashikar-Zuck SM et al (2017) Trajectory of improvement in children and adolescents with chronic migraine: results from the cognitive behavioral therapy and amitriptyline trial. J Pain 18:637–644
Powers SW, Kashikar-Zuck SM, Allen JR et al (2013) Cognitive behavioral therapy plus amitriptyline for chronic migraine in children and adolescents. JAMA 310:2622–2630
Lake AE 3rd (2001) Behavioral and nonpharmacologic treatments of headache. Med Clin North Am 85:1055–1075
Carpino E, Segal S, Logan D et al (2014) The interplay of pain-related self-efficacy and fear on functional outcomes among youth with headache. J Pain 15:527–534
Short AL (2019) Enhancing migraine self-efficacy and reducing disability through a self-management program. J Am Assoc Nurse Pract 33:20–28
Mérelle SY, Sorbi MJ, Duivenvoorden HJ et al (2010) Qualities and health of lay trainers with migraine for behavioral attack prevention. Headache 50:613–625
Allen LL, Haririfar M, Cohen J et al (2000) Quality of life and locus of control of migraineurs. Clin Excell Nurse Pract 4:41–49
Wells RE, O’Connell N, Pierce CR et al (2021) Effectiveness of mindfulness meditation vs headache education for adults with migraine: a randomized clinical trial. JAMA Intern Med 181:317–328
Grazzi L, Montisano DA, Rizzoli P et al (2023) A single-group study on the effect of onabotulinumtoxina in patients with chronic migraine associated with medication overuse headache: pain catastrophizing plays a role. Toxins (Basel) 15:86
Kleiboer A, Sorbi M, van Silfhout M et al (2014) Short-term effectiveness of an online behavioral training in migraine self-management: a randomized controlled trial. Behav Res Ther 61:61–69
Raggi A, Grignani E, Leonardi M et al (2018) Behavioral approaches for primary headaches: recent advances. Headache 56:913–925
Grazzi L, Montisano DA, D’Amico D, et al (2024) Multimodal digital health treatments for Chronic Migraine associated with Medication Overuse Headache: a literature appraisal and results of a single-arm open trial (the BE-HOME program). Neurol Sci. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s10072-024-07568-8. Epub ahead of print, May 21, 2024
Licina E, Radojicic A, Jeremic M et al (2023) Non-pharmacological treatment of primary headaches-a focused review. Brain Sci 13:1432
Meyer B, Keller A, Wöhlbier HG et al (2016) Progressive muscle relaxation reduces migraine frequency and normalizes amplitudes of contingent negative variation (CNV). J Headache Pain 9:213–224
Dobos D, Szabó E, Baksa D et al (2022) Regular practice of autogenic training reduces migraine frequency and is associated with brain activity changes in response to fearful visual stimuli. Front behav neurosci 15:780081
Aemaz Ur Rehman M, Waseem R, Habiba U et al (2022) Efficacy of mindfulness-based intervention for the treatment of chronic headaches: a systematic review and meta-analysis. Ann Med Surg (Lond) 78:103862
Andrasik F, Grazzi L, D’Amico D et al (2016) Mindfulness and headache: a “new” old treatment, with new findings. Cephalalgia 36:1192–1205
Teasdale JD, Segal Z, Williams JM (1995) How does cognitive therapy prevent depressive relapse and why should attentional control (mindfulness) training help? Behav Res Ther 33:25–39
Kabat-Zinn J (2013) Full catastrophe living: using the wisdom of your body and mind to face stress, pain, and illness. Bantam Books, New York
Sharma M, Rush SE (2014) Mindfulness-based stress reduction as a stress management intervention for healthy individuals: a systematic review. J Evid Based Complementary Altern Med 19:271–286
Zhang CQ, Leeming E, Smith P et al (2018) Acceptance and commitment therapy for health behavior change: a contextually-driven approach. Front Psychol 8:2350
Bernstein CA, Connor JP, Vilsmark ES et al (2022) Acceptance and commitment therapy for episodic migraine: rationale and design of a pilot randomized controlled trial. Contemp Clin Trials 121:106907
Grazzi L, Andrasik F, Rizzoli P et al (2021) Acceptance and commitment therapy for high frequency episodic migraine without aura: findings from a randomized pilot investigation. Headache 61:895–905
Grazzi L, D’Amico D, Guastafierro E et al (2023) Efficacy of mindfulness added to treatment as usual in patients with chronic migraine and medication overuse headache: a phase-III single-blind randomized-controlled trial (the MIND-CM study). J Headache Pain 24:86
Fedeli D, Ciullo G, Demichelis G et al (2024) Longitudinal neurofunctional changes in medication overuse headache patients after mindfulness practice in a randomized controlled trial (the MIND-CM study). J Headache Pain 25:97
Faedda N, Natalucci G, Baglioni V et al (2019) Behavioral therapies in headache: focus on mindfulness and cognitive behavioral therapy in children and adolescents. Expert Rev Neurother 19:1219–1228
Gazerani P (2021) A bidirectional view of migraine and diet relationship. Neuropsychiatr Dis Treat 17:435–451
Gazerani P (2023) Diet and migraine: what is proven? Curr Opin Neurol 36:615–621
Gazerani P (2020) Migraine and diet. Nutrients 12:1658
Orr SL (2016) Diet and nutraceutical interventions for headache management: a review of the evidence. Cephalalgia 36(12):1112–1133. https://doiorg.publicaciones.saludcastillayleon.es/10.1177/0333102415590239
Grazzi L, Rizzoli P (2021) Lessons from lockdown — behavioural interventions in migraine. Nat Rev Neurol 17:195–196
Grazzi L, Toppo C, D’Amico D et al (2021) Non-pharmacological approaches to headaches: non-invasive neuromodulation, nutraceuticals, and behavioral approaches. Int J Environ Res Public Health 18:1503
Ariyanfar S, Razeghi Jahromi S, Togha M et al (2022) Review on headache related to dietary supplements. Curr Pain Headache Rep 26:193–218
D’Onofrio F, Raimo S, Spitaleri D et al (2017) Usefulness of nutraceuticals in migraine prophylaxis. Neurol Sci 38(Suppl 1):117–120
Kaur K, Hernandez V, Al Hajaj SW et al (2021) The efficacy of herbal supplements and nutraceuticals for prevention of migraine: can they help? Cureus 13:e14868
Papetti L, Moavero R, Ferilli MAN et al (2021) Truths and myths in pediatric migraine and nutrition. Nutrients 13:2714
Di Lorenzo C, Coppola G, Bracaglia M et al (2019) A ketogenic diet normalizes interictal cortical but not subcortical responsivity in migraineurs. BMC Neurol 19:136
Di Lorenzo C, Pinto A, Ienca R et al (2019) A randomized double-blind, cross-over trial of very low-calorie diet in overweight migraine patients: a possible role for ketones? Nutrients 11:1742
Rajapakse T, Pringsheim T (2016) Nutraceuticals in migraine: a summary of existing guidelines for use. Headache 56:808–816
Schoenen J, Jacquy J, Lenaerts M (1998) Effectiveness of high-dose riboflavin in migraine prophylaxis. A randomized controlled trial. Neurology 50:466–470
Sándor PS, Di Clemente L, Coppola G et al (2005) Efficacy of coenzyme Q10 in migraine prophylaxis: a randomized controlled trial. Neurology 64:713–715
Parohan M, Sarraf P, Javanbakht MH et al (2020) Effect of coenzyme Q10 supplementation on clinical features of migraine: a systematic review and dose-response meta-analysis of randomized controlled trials. Nutr Neurosci 23:868–875
Teigen L, Boes CJ (2015) An evidence-based review of oral magnesium supplementation in the preventive treatment of migraine. Cephalalgia 35:912–922
Lipton RB, Göbel H, Einhäupl KM et al (2004) Petasites hybridus root (butterbur) is an effective preventive treatment for migraine. Neurology 63:2240–2244
Diener HC, Freitag FG, Danesch U (2018) Safety profile of a special butterbur extract from Petasites hybridus in migraine prevention with emphasis on the liver. Cephalalgia Reports 1:2515816318759304
Pittler MH, Ernst E (2004) Feverfew for preventing migraine. Cochrane Database Syst Rev. 2004:Cd002286
Pradalier A, Bakouche P, Baudesson G et al (2001) Failure of omega-3 polyunsaturated fatty acids in prevention of migraine: a double-blind study versus placebo. Cephalalgia 21:818–822
Ramsden CE, Zamora D, Faurot KR et al (2021) Dietary alteration of n-3 and n-6 fatty acids for headache reduction in adults with migraine: randomized controlled trial. BMJ 374:n1448
Wang HF, Liu WC, Zailani H et al (2024) A 12-week randomized double-blind clinical trial of eicosapentaenoic acid intervention in episodic migraine. Brain Behav Immun 118:459–467
Chirchiglia D, Cione E, Caroleo MC et al (2018) Effects of add-on ultramicronized N-palmitol ethanol amide in patients suffering of migraine with aura: a pilot study. Front Neurol 9:674
Papetti L, Sforza G, Tullo G et al (2020) Tolerability of palmitoylethanolamide in a pediatric population suffering from migraine: a pilot study. Pain Res Manag 2020:3938640
Usai S, Grazzi L, Bussone G (2011) Gingkolide B as migraine preventive treatment in young age: results at 1-year follow-up. Neurol Sci 32(Suppl 1):S197–S199
Holdridge A, Donnelly M, Kuruvilla DE (2022) Integrative, interventional, and non-invasive approaches for the treatment for migraine during pregnancy. Curr Pain Headache Rep 26:323–330
Makrides M, Crosby DD, Bain E et al (2014) Magnesium supplementation in pregnancy. Cochrane Database Syst Rev. 2014:Cd000937
Institute of Medicine Standing Committee on the Scientific Evaluation of Dietary Reference I, its Panel on Folate OBV, Choline (1998) Dietary Reference Intakes for Thiamin, Riboflavin, Niacin, Vitamin B(6), Folate, Vitamin B(12), Pantothenic Acid, Biotin, and Choline. Washington (DC): National Academies Press. Available at: https://www.ncbi.nlm.nih.gov/books/NBK114310/. Accessed 18 July 2024
Teran E, Hernandez I, Nieto B et al (2009) Coenzyme Q10 supplementation during pregnancy reduces the risk of pre-eclampsia. Int J Gynaecol Obstet 105:43–45
Guilbot A, Bangratz M, Ait Abdellah S et al (2017) A combination of coenzyme Q10, feverfew and magnesium for migraine prophylaxis: a prospective observational study. BMC Complement Altern Med 17:433
Negro A, Delaruelle Z, Ivanova TA et al (2017) Headache and pregnancy: a systematic review. J Headache Pain 18:106
Papetti L, Ursitti F, Moavero R et al (2019) Prophylactic treatment of pediatric migraine: is there anything new in the last decade? Front Neurol 10:771
Wells RE, Bertisch SM, Buettner C et al (2011) Complementary and alternative medicine use among adults with migraines/severe headaches. Headache 51:1087–1097
Onan D, Ekizoğlu E, Arıkan H et al (2023) The efficacy of physical therapy and rehabilitation approaches in chronic migraine: a systematic review and meta-analysis. J Integr Neurosci 22:126
Onan D, Arıkan H, Martelletti P (2023) The effect of onabotulinumtoxina on headache intensity and number of monthly headache days in individuals with chronic migraine with different levels of neck disability. Toxins (Basel) 15:685
Woldeamanuel YW, Oliveira ABD (2022) What is the efficacy of aerobic exercise versus strength training in the treatment of migraine? A systematic review and network meta-analysis of clinical trials. J Headache Pain 23:134
Sari Aslani P, Hassanpour M, Razi O et al (2022) Resistance training reduces pain indices and improves quality of life and body strength in women with migraine disorders. Sport Sci Health 18:433–443
Benatto MT, Florencio LL, Bragatto MM et al (2022) Neck-specific strengthening exercise compared with placebo sham ultrasound in patients with migraine: a randomized controlled trial. BMC Neurol 22:126
Sun L, Li G, Liu F et al (2022) Resistance exercise relieves symptoms of vestibular migraine patients with MRI diagnosis: a randomized parallel-controlled single-blind clinical trial. Rev Neurol (Paris) 178:370–376
La Touche R, Fierro-Marrero J, Sánchez-Ruíz I et al (2023) Prescription of therapeutic exercise in migraine, an evidence-based clinical practice guideline. J Headache Pain 24:68
Rainero I, Rubino E, Gallone S et al (2011) Evidence for an association between migraine and the hypocretin receptor 1 gene. J Headache Pain 12:193–199
Watanabe H, Kuwabara T, Ohkubo M et al (1996) Elevation of cerebral lactate detected by localized 1H-magnetic resonance spectroscopy in migraine during the interictal period. Neurology 47:1093–1095
Jonhagen S, Ackermann P, Saartok T et al (2006) Calcitonin gene related peptide and neuropeptide Y in skeletal muscle after eccentric exercise: a microdialysis study. Br J Sports Med 40:264–267
Dittrich SM, Günther V, Franz G et al (2008) Aerobic exercise with relaxation: influence on pain and psychological well-being in female migraine patients. Clin J Sport Med 18:363–365
Dinoff A, Herrmann N, Swardfager W et al (2016) The effect of exercise training on resting concentrations of peripheral Brain-Derived Neurotrophic Factor (BDNF): a meta-analysis. PLoS One 11:e0163037
Amin FM, Aristeidou S, Baraldi C et al (2018) The association between migraine and physical exercise. J Headache Pain 19:83
Sparling PB, Giuffrida A, Piomelli D et al (2003) Exercise activates the endocannabinoid system. NeuroReport 14:2209–2211
Demarquay G, Mawet J, Guégan-Massardier E et al (2021) Revised guidelines of the French headache society for the diagnosis and management of migraine in adults. Part 3: Non-pharmacological treatment. Rev Neurol (Paris) 177:753–759
Schytz HW, Amin FM, Jensen RH et al (2021) Reference programme: diagnosis and treatment of headache disorders and facial pain. Danish Headache Society, 3rd edition, 2020. J Headache Pain. 22:22
American Headache Society (2019) The American Headache Society position statement on integrating new migraine treatments into clinical practice. Headache 59:1–18
Reina-Varona Á, Madroñero-Miguel B, Gaul C et al (2023) Therapeutic exercise parameters, considerations, and recommendations for migraine treatment: an international delphi study. Phys Ther. 103:pzad080
Moore CS, Sibbritt DW, Adams J (2017) A critical review of manual therapy use for headache disorders: prevalence, profiles, motivations, communication and self-reported effectiveness. BMC Neurol 17:61
Rist PM, Hernandez A, Bernstein C et al (2019) The impact of spinal manipulation on migraine pain and disability: a systematic review and meta-analysis. Headache 59:532–542
Chaibi A, Tuchin PJ, Russell MB (2011) Manual therapies for migraine: a systematic review. J Headache Pain 12:127–133
Muñoz-Gómez E, Inglés M, Serra-Añó P et al (2021) Effectiveness of a manual therapy protocol based on articulatory techniques in migraine patients. A randomized controlled trial. Musculoskelet Sci Pract. 54:102386
Bialosky JE, Bishop MD, Price DD et al (2009) The mechanisms of manual therapy in the treatment of musculoskeletal pain: a comprehensive model. Man Ther 14:531–538
Muñoz-Gómez E, Serra-Añó P, Mollà-Casanova S et al (2022) Potential add-on effects of manual therapy techniques in migraine patients: a randomised controlled trial. J Clin Med 11:4686
Di Antonio S, Arendt-Nielsen L, Castaldo M (2023) Cervical musculoskeletal impairments and pain sensitivity in migraine patients. Musculoskelet Sci Pract 66:102817
Di Antonio S, Arendt-Nielsen L, Ponzano M et al (2023) Migraine patients with and without neck pain: differences in clinical characteristics, sensitization, musculoskeletal impairments, and psychological burden. Musculoskelet Sci Pract 66:102800
Szikszay TM, Hoenick S, von Korn K et al (2019) Which examination tests detect differences in cervical musculoskeletal impairments in people with migraine? A systematic review and meta-analysis. Phys Ther 99:549–569
Di Antonio S, Arendt-Nielsen L, Ponzano M et al (2022) Cervical musculoskeletal impairments in the 4 phases of the migraine cycle in episodic migraine patients. Cephalalgia 42:827–845
Tedeschi R, Pillastrini P, Pierangeli G et al (2024) Is physiotherapy in migraines known to sufferers? A cross-sectional study. Neurol Sci 45:1669–1674
Louw A, Diener I, Butler DS et al (2011) The effect of neuroscience education on pain, disability, anxiety, and stress in chronic musculoskeletal pain. Arch Phys Med Rehabil 92:2041–2056
Kindelan-Calvo P, Gil-Martínez A, Paris-Alemany A et al (2014) Effectiveness of therapeutic patient education for adults with migraine. A systematic review and meta-analysis of randomized controlled trials. Pain Med 15:1619–1636
Meise R, Carvalho GF, Thiel C et al (2023) Additional effects of pain neuroscience education combined with physiotherapy on the headache frequency of adult patients with migraine: a randomized controlled trial. Cephalalgia 43:3331024221144781
Fernández-de-Las-Peñas C, Navarro-Santana MJ, Curiel-Montero F et al (2022) Localized and widespread pressure pain hypersensitivity in patients with episodic or chronic migraine: a systematic review and meta-analysis. Cephalalgia 42:966–980
Di Antonio S, Arendt-Nielsen L, Ponzano M et al (2024) Profiling migraine patients according to clinical and psychophysical characteristics: clinical validity of distinct migraine clusters. Neurol Sci 45:1185–1200
Edwards RR, Dworkin RH, Turk DC et al (2016) Patient phenotyping in clinical trials of chronic pain treatments: IMMPACT recommendations. Pain 157:1851–1871
World Health Organization (2024) “What is Pharmacovigilance?”. Available at: https://www.who.int/teams/regulation-prequalification/regulation-and-safety/pharmacovigilance#:~:text=Pharmacovigilance%20is%20the%20science%20and,other%20medicine%2Fvaccine%20related%20problem. Accessed 18 July 2024
Gérard AO, Merino D, Van Obberghen EK et al (2022) Calcitonin gene-related peptide-targeting drugs and Raynaud’s phenomenon: a real-world potential safety signal from the WHO pharmacovigilance database. J Headache Pain 23:53
Woods RH (2022) Alopecia signals associated with calcitonin gene-related peptide inhibitors in the treatment or prophylaxis of migraine: a pharmacovigilance study. Pharmacotherapy 42:758–767
Roberto G, Piccinni C, D’Alessandro R et al (2014) Triptans and serious adverse vascular events: data mining of the FDA Adverse Event Reporting System database. Cephalalgia 34:5–13
Beau-Salinas F, Jonville-Béra AP, Cissoko H et al (2010) Drug dependence associated with triptans and ergot derivatives: a case/non-case study. Eur J Clin Pharmacol 66:413–417
Millson D, Frischer M, Croft P et al (2000) Are triptans with enhanced lipophilicity used for the acute treatment of migraine associated with an increased consulting rate for depressive illness? Cephalalgia 20:732–737
Conijn M, Maas V, van Tuyl M, et al (2024) Breastfeeding-related adverse drug reactions of triptans: a descriptive analysis using four pharmacovigilance databases. Breastfeed Med. https://doiorg.publicaciones.saludcastillayleon.es/10.1089/bfm.2024.0022. Epub ahead of print, Apr 2, 2024
Merino D, Gérard AO, Van Obberghen EK et al (2023) The neuropsychiatric safety profile of lasmiditan: a comparative disproportionality analysis with triptans. Neurotherapeutics 20:1305–1315
Wan X, Zhang S, Jiang C, et al (2024) Safety profile of lasmiditan: a retrospective post-marketing pharmacovigilance study based on the real-world data of FAERS database. Expert Opin Drug Saf. https://doiorg.publicaciones.saludcastillayleon.es/10.1080/14740338.2024.2371382 2024 Epub ahead of print, Jun 20, 2024
Battini V, Carnovale C, Clementi E et al (2023) Ubrogepant and rimegepant: signal detection using spontaneous reports of adverse events from the Food and Drug Administration Adverse Event Reporting System. Expert Opin Drug Saf 22:1105–1112
Cao B, Gu S, Shen Z et al (2024) Evaluating Ubrogepant-related adverse events using the FDA adverse event reporting system. Expert Opin Drug Saf 23:297–303
Blumenfeld AM, Kaur G, Mahajan A et al (2023) Effectiveness and safety of chronic migraine preventive treatments: a systematic literature review. Pain Ther 12:251–274
Holzer P, Holzer-Petsche U (2021) Constipation caused by anti-calcitonin gene-related peptide migraine therapeutics explained by antagonism of calcitonin gene-related peptide’s motor-stimulating and prosecretory function in the intestine. Front Physiol 12:820006
Ihara K, Takizawa T, Watanabe N et al (2024) Potential benefits and possible risks of CGRP-targeted multitherapy in migraine. Expert Opin Drug Metab Toxicol 20:1–4
Silberstein SD, Reshef S, Cohen JM et al (2023) Adverse events reported with therapies targeting the CGRP pathway during the first 6 months post-launch: a retrospective analysis using the FDA adverse events reporting system. Adv Ther 40:445–459
Robblee J, Harvey LK (2022) Cardiovascular disease and migraine: are the new treatments safe? Curr Pain Headache Rep 26:647–655
Chhabra N, Mead-Harvey C, Dodoo CA et al (2024) Blood pressure elevation in erenumab-treated patients with migraine: a retrospective real-world experience. Headache 64:233–242
Perino J, Corand V, Laurent E et al (2022) Myocardial infarction associated with erenumab: a case report. Pharmacotherapy 42:585–589
Noseda R, Bedussi F, Gobbi C et al (2021) Safety profile of erenumab, galcanezumab and fremanezumab in pregnancy and lactation: analysis of the WHO pharmacovigilance database. Cephalalgia 41:789–798
Noseda R, Bedussi F, Gobbi C et al (2023) Safety profile of monoclonal antibodies targeting the calcitonin gene-related peptide system in pregnancy: updated analysis in VigiBase®. Cephalalgia 43:03331024231158083
Shafei L, Mekki L, Maklad E et al (2023) Factors that influence patient and public adverse drug reaction reporting: a systematic review using the theoretical domains framework. Int J Clin Pharm 45:801–813
Hartnell NR, Wilson JP (2004) Replication of the weber effect using postmarketing adverse event reports voluntarily submitted to the United States food and drug administration. Pharmacotherapy 24:743–749
Negro A, Martelletti P (2011) Chronic migraine plus medication overuse headache: two entities or not? J Headache Pain 12:593–601
Committee HC, Olesen J, Bousser MG et al (2006) New appendix criteria open for a broader concept of chronic migraine. Cephalalgia 26:742–746
Welch K, Goadsby P (2002) Chronic daily headache: nosology and pathophysiology. Curr Opin Neurol 15:287–295
Torres-Ferrús M, Ursitti F, Alpuente A et al (2020) From transformation to chronification of migraine: pathophysiological and clinical aspects. J Headache Pain 21:42
Weatherall MW (2015) The diagnosis and treatment of chronic migraine. Ther Adv Chronic Dis 6:115–123
Gosalia H, Moreno-Ajona D, Goadsby PJ (2024) Medication-overuse headache: a narrative review. J Headache Pain 25:89
Ferrari A, Baraldi C, Sternieri E (2015) Medication overuse and chronic migraine: a critical review according to clinical pharmacology. Expert Opin Drug Metab Toxicol 11:1127–1144
Tana C, Raffaelli B, Souza MNP et al (2024) Health equity, care access and quality in headache - part 1. J Headache Pain 25:12
Raffaelli B, Rubio-Beltrán E, Cho SJ et al (2023) Health equity, care access and quality in headache - part 2. J Headache Pain 24:167
Shao SC, Hentz J, Shank P et al (2024) Functional impairment of chronic migraine with medication overuse: Secondary analysis from the Medication Overuse Treatment Strategy (MOTS) trial. Headache 64:632–642
Diener HC, Kropp P, Dresler T et al (2022) Management of medication overuse (MO) and medication overuse headache (MOH) S1 guideline. Neurol Res Pract 4:37
Tana C, Cipollone F, Giamberardino MA (2023) New therapeutic options for migraine. Curr Pharm Des 29:1964–1966
Tana C, Cipollone F, Giamberardino MA et al (2023) New drugs targeting calcitonin gene-related peptide for the management of migraines. Expert Opin Emerg Drugs 28:233–240
Lipton RB, Silberstein SD (2015) Episodic and chronic migraine headache: breaking down barriers to optimal treatment and prevention. Headache 55(Suppl 2):103–122
GBD (2021) Diseases and Injuries Collaborators (2024) Global incidence, prevalence, years lived with disability (YLDs), disability-adjusted life-years (DALYs), and healthy life expectancy (HALE) for 371 diseases and injuries in 204 countries and territories and 811 subnational locations, 1990–2021: a systematic analysis for the Global Burden of Disease Study 2021. Lancet 403:2133–2161
May A, Schulte LH (2016) Chronic migraine: risk factors, mechanisms and treatment. Nat Rev Neurol 12:455–464
Buse DC, Greisman JD, Baigi K et al (2019) Migraine progression: a systematic review. Headache 59:306–338
Tana C, Bentivegna E, Cho SJ, Harriott AM et al (2022) Long COVID headache. J Headache Pain 23:93
Tana C, Giamberardino MA, Martelletti P (2023) Long COVID and especially headache syndromes. Curr Opin Neurol 36:168–174
Waliszewska-Prosół M, Budrewicz S (2021) The unusual course of a migraine attack during COVID-19 infection - Case studies of three patients. J Infect Public Health 14:903–905
Moïsi L, Mino JC, Guidet B et al (2024) Frailty assessment in critically ill older adults: a narrative review. Ann Intensive Care 14:93
Tana C, Moffa L, Falasca K et al (2023) Approach to COVID-19 in older adults and indications for improving the outcomes. Ann Med 55:2265298
Tana C (2024) Editorial: Frailty in older patients during the COVID-19 era. Front Med (Lausanne) 10:1348468
Ornello R, Andreou AP, De Matteis E et al (2024) Resistant and refractory migraine: clinical presentation, pathophysiology, and management. EBioMedicine 99:104943
Sacco S, Braschinsky M, Ducros A et al (2020) European headache federation consensus on the definition of resistant and refractory migraine: developed with the endorsement of the European Migraine & Headache Alliance (EMHA). J Headache Pain. 21:76
Waliszewska-Prosół M, Nowakowska-Kotas M, Chojdak-Łukasiewicz J et al (2021) Migraine and sleep-an unexplained association? Int J Mol Sci 22:5539
Caronna E, Gallardo VJ, Egeo G, et al (2024) Redefining migraine prevention: early treatment with anti-CGRP monoclonal antibodies enhances response in the real world. J Neurol Neurosurg Psychiatry. https://doiorg.publicaciones.saludcastillayleon.es/10.1136/jnnp-2023-333295. Epub ahead of print, May 22, 2024
Steiner TJ, Stovner LJ, Jensen R et al (2020) Migraine remains second among the world’s causes of disability, and first among young women: findings from GBD2019. J Headache Pain 21:137
Stewart WF, Lipton RB, Dowson AJ et al (2001) Development and testing of the Migraine Disability Assessment (MIDAS) Questionnaire to assess headache-related disability. Neurology 56(6 Suppl 1):S20–S28
McKenna SP, Doward LC, Davey KM (1998) The Development and Psychometric Properties of the MSQOL: A Migraine-Specific Quality-of-Life Instrument. Clin Drug Investig 15:413–423
Kosinski M, Bayliss MS, Bjorner JB et al (2003) A six-item short-form survey for measuring headache impact: the HIT-6. Qual Life Res 12:963–974
Raggi A, Grazzi L, Grignani E et al (2018) The use of MIDAS in patients with chronic migraine and medication-overuse headache: should we trust it? Neurol Sci 39(Suppl 1):125–127
Leonardi M, Raggi A, Bussone G et al (2010) Health-related quality of life, disability and severity of disease in patients with migraine attending to a specialty headache center. Headache 50:1576–1586
Seo J-G, Park S-P (2015) Validation of the Patient Health Questionnaire-9 (PHQ-9) and PHQ-2 in patients with migraine. J Headache Pain 16:65
D’Amico D, Tepper SJ, Guastafierro E et al (2021) Mapping assessments instruments for headache disorders against the ICF biopsychosocial model of health and disability. Int J Environ Res Public Health 18:246
Leonardi M, Steiner TJ, Scher AT et al (2005) The global burden of migraine: measuring disability in headache disorders with WHO’s Classification of Functioning, Disability and Health (ICF). J Headache Pain 6:429–440
Raggi A, Giovannetti AM, Quintas R et al (2012) A systematic review of the psychosocial difficulties relevant to patients with migraine. J Headache Pain 13:595–606
Duan S, Ren Z, Xia H et al (2023) Associations between anxiety, depression with migraine, and migraine-related burdens. Front Neurol 14:1090878
Peres MFP, Mercante JPP, Tobo PR et al (2017) Anxiety and depression symptoms and migraine: a symptom-based approach research. J Headache Pain 18:37
Rammohan K, Mundayadan SM, Das S et al (2019) Migraine and mood disorders: prevalence, clinical correlations and disability. J Neurosci Rural Pract 10:28–33
Della Vecchia A, De Luca C, Becattini L et al (2024) Beyond pain relief: unveiling the multifaceted impact of Anti-CGRP/R mAbs on comorbid symptoms in resistant migraine patients. Biomedicines 12:677
Castro Zamparella T, Carpinella M, Peres M et al (2024) Specific cognitive and psychological alterations are more strongly linked to increased migraine disability than chronic migraine diagnosis. J Headache Pain 25:37
Torres-Ferrús M, Gallardo VJ, Alpuente A et al (2024) Improvement of migraine depressive symptoms is not related to headache frequency: exploring the impact of anti-CGRP therapies. Cephalalgia 44:03331024231222923
Stubberud A, Buse DC, Kristoffersen ES et al (2021) Is there a causal relationship between stress and migraine? Current evidence and implications for management. J Headache Pain 22:155
Perozzo P, Savi L, Castelli L et al (2005) Anger and emotional distress in patients with migraine and tension-type headache. J Headache Pain 6:392–399
Seo JG, Park SP (2018) Significance of fatigue in patients with migraine. J Clin Neurosci 50:69–73
Garcia-Monco JC, Foncea N, Bilbao A et al (2007) Impact of preventive therapy with nadolol and topiramate on the quality of life of migraine patients. Cephalalgia 27:920–928
Duan S, Ren Z, Xia H et al (2022) Association between sleep quality, migraine and migraine burden. Front Neurol 13:955298
Tiseo C, Vacca A, Felbush A et al (2020) Migraine and sleep disorders: a systematic review. J Headache Pain 21:126
Gori S, Morelli N, Maestri M et al (2005) Sleep quality, chronotypes and preferential timing of attacks in migraine without aura. J Headache Pain 6:258–260
Raggi A, Covelli V, Leonardi M et al (2014) Difficulties in work-related activities among migraineurs are scarcely collected: results from a literature review. Neurol Sci 35(Suppl 1):23–26
Shapiro RE, Martin AA, Bhardwaj S et al (2023) Relationships between headache frequency, disability, and disability-related unemployment among adults with migraine. J Manag Care Spec Pharm 29:197–209
Shapiro RE, Hochstetler HM, Dennehy EB et al (2019) Lasmiditan for acute treatment of migraine in patients with cardiovascular risk factors: post-hoc analysis of pooled results from 2 randomized, double-blind, placebo-controlled, phase 3 trials. J Headache Pain 20:90
García-Azorín D, Moya-Alarcón C, Armada B et al (2024) Societal and economic burden of migraine in Spain: results from the 2020 National Health and Wellness Survey. J Headache Pain 25:38
Lombard L, Farrar M, Ye W et al (2020) A global real-world assessment of the impact on health-related quality of life and work productivity of migraine in patients with insufficient versus good response to triptan medication. J Headache Pain 21:41
Woodland L, Brooks SK, Webster RK et al (2023) Risk factors for school-based presenteeism in children: a systematic review. BMC Psychology 11:169
Leonardi M, Guastafierro E, Toppo C et al (2023) Societal and personal impact of migraine. Handb Clin Neurol 198:23–29
Snoer AH, Høst C, Dømgaard M et al (2021) Frequent or chronic migraine negatively impacts personal, social and professional life. Dan Med J 68:A08200592
Cerami C, Crespi C, Bottiroli S et al (2021) High perceived isolation and reduced social support affect headache impact levels in migraine after the Covid-19 outbreak: a cross sectional survey on chronic and episodic patients. Cephalalgia 41:1437–1446
Takahashi TT, Ornello R, Quatrosi G et al (2021) (2021) Medication overuse and drug addiction: a narrative review from addiction perspective. J Headache Pain 22:32
Mäki K, Vahtera J, Virtanen M et al (2008) Work stress and new-onset migraine in a female employee population. Cephalalgia 28:18–25
Berardelli I, Sarubbi S, Lamis DA et al (2019) Job satisfaction mediates the association between perceived disability and work productivity in migraine headache patients. Int J Environ Res Public Health 16:3341
Kuo WY, Huang CC, Weng SF et al (2015) Higher migraine risk in healthcare professionals than in general population: a nationwide population-based cohort study in Taiwan. J Headache Pain 16:102
Wei D, Loganathan T, Wong LP (2023) Employees of the banking sector in Guizhou Province in China: prevalence of migraine, symptoms, disability and occupational risk factors. J Headache Pain 24:52
Sandoe CH, Sasikumar S, Lay C et al (2019) The impact of shift work on migraine: a case series and narrative review. Headache 59:1631–1640
Xie W, Li R, He M et al (2020) Prevalence and risk factors associated with headache amongst medical staff in South China. J Headache Pain. 21:5
Suzuki K, Okamura M, Haruyama Y et al (2022) Exploring the contributing factors to multiple chemical sensitivity in patients with migraine. J Occup Health 64:e12328
Martins R, Large S, Russell R et al (2023) The hidden economic consequences of migraine to the UK Government: burden-of-disease analysis using a fiscal framework. J Health Econ Outcomes Res 10:72–81
Begasse de Dhaem O, Sakai F (2022) Migraine in the workplace. eNeurologicalSci 27:100408
Gerth WC, Carides GW, Dasbach EJ et al (2001) The multinational impact of migraine symptoms on healthcare utilisation and work loss. Pharmacoeconomics 19:197–206
Reilly MC, Zbrozek AS, Dukes EM (1993) The validity and reproducibility of a work productivity and activity impairment instrument. Pharmacoeconomics 4:353–365
Raggi A, Covelli V, Guastafierro E et al (2018) Validation of a self-reported instrument to assess work-related difficulties in patients with migraine: the HEADWORK questionnaire. J Headache Pain 19:85
Sakai F, Igarashi H, Yokoyama M et al (2023) Diagnosis, knowledge, perception, and productivity impact of headache education and clinical evaluation program in the workplace at an information technology company of more than 70,000 employees. Cephalalgia 43:3331024231165682
Vicente-Herrero T, Burke TA, Laínez MJ (2004) The impact of a worksite migraine intervention program on work productivity, productivity costs, and non-workplace impairment among Spanish postal service employees from an employer perspective. Curr Med Res Opin 20:1805–1814
Gerth WC, Sarma S, Hu XH et al (2004) Productivity cost benefit to employers of treating migraine with rizatriptan: a specific worksite analysis and model. J Occup Environ Med 46:48–54
Riccò M, Ferraro P, Camisa V et al (2022) Managing of migraine in the workplaces: knowledge, attitudes and practices of italian occupational physicians. Medicina (Kaunas) 58:686
Gil-Gouveia R, Oliveira AG, Martins IP (2016) The impact of cognitive symptoms on migraine attack-related disability. Cephalalgia 36:422–430
Fan X, Els C, Corbet KJ et al (2016) “Decision-critical” work: a conceptual framework. J Occup Med Toxicol 11:22
DiGuiseppi CG, Johnson RL, Betz ME (2024) Migraine headaches are associated with motor vehicle crashes and driving habits among older drivers: Prospective cohort study. J Am Geriatr Soc 72:791–801
Agosti R (2018) Migraine burden of disease: from the patient’s experience to a socio-economic view. Headache 58(Suppl 1):17–32
Bloudek LM, Stokes M, Buse DC et al (2012) Cost of healthcare for patients with migraine in five European countries: results from the International Burden of Migraine Study (IBMS). J Headache Pain 13:361–378
Eltrafi A, Shrestha S, Ahmed A et al (2023) Economic burden of chronic migraine in OECD countries: a systematic review. Health Econ Rev 13:43
Husøy A, Katsarava Z, Steiner TJ (2023) The relationship between headache-attributed disability and lost productivity: 3 Attack frequency is the dominating variable. J Headache Pain 24:7
Rao GN, Kulkarni GB, Gururaj G et al (2015) The burden attributable to headache disorders in India: estimates from a community-based study in Karnataka State. J Headache Pain 16:94
Tu S, Liew D, Ademi Z et al (2020) The Health and Productivity Burden of Migraines in Australia. Headache 60:2291–2303
Steiner TJ, Jensen R, Katsarava Z (2021) Structured headache services as the solution to the ill-health burden of headache. 1. Rationale and description. J Headache Pain. 22:78
Tinelli M, Leonardi M, Paemeleire K et al (2021) Structured headache services as the solution to the ill-health burden of headache. 2. Modelling effectiveness and cost-effectiveness of implementation in Europe: methodology. J Headache Pain. 22:99
World Health Organization (2011) Atlas of headache disorders and resources in the World 2011. World Health Organization; Geneva. Available at: https://www.who.int/publications/i/item/9789241564212. Accessed 18 July 2024
Burch R, Rizzoli P, Loder E (2018) The prevalence and impact of migraine and severe headache in the United States: figures and trends from government health studies. Headache. 58(4):496–505
Friedman BW, Serrano D, Reed M et al (2009) Use of the emergency department for severe headache. A population-based study. Headache 49:21–30
Katsarava Z, Dzagnidze A, Kukava M et al (2009) Primary headache disorders in the Republic of Georgia: prevalence and risk factors. Neurology 73:1796–1803
Ayzenberg I, Katsarava Z, Sborowski A et al (2012) The prevalence of primary headache disorders in Russia: a countrywide survey. Cephalalgia 32:373–381
Lisicki M, Souza MNP, de Oliveira AB et al (2022) Bridging the gaps of headache care for underserved populations: current status of the headache field in Latin America. Cephalalgia 42:1086–1090
Mbewe E, Zairemthiama P, Yeh HH et al (2015) The epidemiology of primary headache disorders in Zambia: a population-based door-to-door survey. J Headache Pain 16:30
Zebenigus M, Tekle-Haimanot R, Worku DK (2016) The prevalence of primary headache disorders in Ethiopia. J Headache Pain 17:110
Mauser ED, Rosen NL (2014) So many migraines, so few subspecialists: analysis of the geographic location of United Council for Neurologic Subspecialties (UCNS) certified headache subspecialists compared to United States headache demographics. Headache 54:1347–1357
Najib U, Moore M, Watson D (2019) Unique considerations for special populations in episodic migraine: the underserved. Curr Pain Headache Rep 23:9
Charleston L, Royce J, Monteith TS et al (2018) Migraine care challenges and strategies in US uninsured and underinsured adults: a narrative review, part 1. Headache 58:506–511
Charleston L, Royce J, Monteith TS et al (2018) Migraine care challenges and strategies in US uninsured and underinsured adults: a narrative review, part 2. Headache 58:633–647
Ashina M, Katsarava Z, Do TP et al (2021) Migraine: epidemiology and systems of care. Lancet 397:1485–1495
Liu R, Yu S, He M et al (2013) Health-care utilization for primary headache disorders in China: a population-based door-to-door survey. J Headache Pain 14:47
Ravishankar K (2004) Barriers to headache care in India and efforts to improve the situation. Lancet Neurol 3:564–567
Armand CE, Borrero-Mejias C, Pace A, et al (2023) Update on Underrepresented Populations in Headache Medicine: What Is Known and Care Considerations. Pract Neurol. Available at: https://practicalneurology.com/articles/2023-may-june/update-on-underrepresented-populations-in-headache-medicine-what-is-known-and-care-considerations. Accessed 18 July 2024
Olesen J, Lekander I, Andlin-Sobocki P et al (2007) Funding of headache research in Europe. Cephalalgia 27:995–999
Shapiro R, Goadsby P (2007) The long drought: the dearth of public funding for headache research. Cephalalgia 27(9):991–994
Befus DR, Irby MB, Coeytaux RR et al (2018) A critical exploration of migraine as a health disparity: the imperative of an equity-oriented, intersectional approach. Curr Pain Headache Rep 22:79
Voloshin AG, Moiseeva IV (2021) Combined Interventional treatment of refractory chronic migraine. SN Compr Clin Med 3:1320–1326
Sacco S, Lampl C, Amin FM et al (2022) European Headache Federation (EHF) consensus on the definition of effective treatment of a migraine attack and of triptan failure. J Headache Pain 23:133
Meise R, Schwarz A, Luedtke K (2022) Effectiveness of patient education and cognitive behavioural treatment as a non-pharmacological intervention for migraine in adults – a systematic review. SN Compr Clin Med 4:197
Bassetti CLA, Endres M, Sander A et al (2022) The European Academy of Neurology Brain Health Strategy: One brain, one life, one approach. Eur J Neurol 29:2559–2566
Steiner TJ, Husøy A, Stovner LJ (2024) GBD2021: headache disorders and global lost health – a focus on children, and a view forward. J Headache Pain 25:91
Choi BC, Pak AW (2006) Multidisciplinarity, interdisciplinarity and transdisciplinarity in health research, services, education and policy: 1. Definitions, objectives, and evidence of effectiveness. Clin Invest Med 29:351–364
Choi BC, Pak AW (2007) Multidisciplinarity, interdisciplinarity, and transdisciplinarity in health research, services, education and policy: 2. Promotors, barriers, and strategies of enhancement. Clin Invest Med 30:E224–E232
Choi BC, Pak AW (2008) Multidisciplinarity, interdisciplinarity, and transdisciplinarity in health research, services, education and policy: 3. Discipline, inter-discipline distance, and selection of discipline. Clin Invest Med 31:E41–E48
Sun Y, Livan G, Ma A et al (2021) Interdisciplinary researchers attain better long-term funding performance. Commun Phys 4:263
Acknowledgements
Alberto Raggi is supported by the Italian Ministry of Health (RRC). Fu-Jung Hsiao is supported by the National Science and Technology. Kristin Sophie Lange and Bianca Raffaelli are participants in the BIH Charité Clinician Scientist Program funded by the Charité – Universitätsmedizin Berlin and the Berlin Institute of Health at Charité (BIH). Igor Petrušić is supported by the Ministry of Science, Technological Development and Innovation, Republic of Serbia (contract number: 451-03-66/2024-03/200146). Tsubasa Takizawa has received JSPS KAKENHI (grant number 22K15693).
Funding
Not applicable.
Author information
Authors and Affiliations
Contributions
Alberto Raggi planned the study, drafted the introduction and discussion sections of the manuscript, and collected the entire body of the manuscript. Paolo Martelletti planned the study and supervised the whole manuscript. The remaining authors drafted sections of the manuscript, in detail: Lou Grangeon and Kristin Sophie Lange, genetic basis; Adriana Della Pietra and Eloisa Rubio-Beltran, molecular pathways; Wei Wang and David Garcia-Azorin, central nervous system (CNS) and peripheral nervous system (PNS) implications; Igor Petrusic and Yonggang Wang, neuroimaging in migraine; Gianluca Coppola and Fu-Jung Hsiao, neurophysiological aspects of migraine; Valeria Caponnetto and Andreas Straube, cardiovascular, cerebrovascular and psychiatric comorbidities of migraine; Marco Arruda and Danilo Antonio Montisano, the migraine cycle: prodromes, ictal phase, and postdromes; Bianca Raffaelli and Alejandro Labastida-Ramirez, pharmacological targets for acute treatment: serotonin and CGRP; Lanfranco Pellesi and Doga Vuralli, pharmacological targets for prophylaxis: CGRP and PACAP; Licia Grazzi and Simone Vigneri, non-pharmacological targets: neuromodulation; Agnese Onofri and Alessia Marcassoli, non-pharmacological targets: cognitive behavioral therapy, relaxation and mindfulness; Parisa Gazerani and Massimiliano Valeriani, non-pharmacological targets: diet and nutraceuticals; Dilara Onan and Matteo Castaldo, non-pharmacological targets: exercise and physical therapy; Keiko Ihara and Tsubasa Takizawa, pharmacovigilance; Marta Waliszewska-Prosół and Claudio Tana, migraine prognosis; William Wells-Gatnik and Matilde Leonardi, psychosocial impact of migraine; Sebastian Straube and Xiangning Fan, occupational health consideration in migraine; Michela Tinelli and Tissa Wijeratne, economic impact of migraine; Marco Lisicki and Mario Peres, underserved populations.
Corresponding author
Ethics declarations
Ethics approval and consent to participate
Not applicable.
Consent for publication
Not applicable.
Competing interests
Alberto Raggi: Associate Editor of the Journal of Headache and Pain, Head of Public Health Section of SN Comprehensive Clinical Medicine. Matilde Leonardi: Associate Editor of the Journal of Headache and Pain. Valeria Caponnetto, Personal fees and honoraria from AbbVie and Teva. Gianluca Coppola: Associate Editor of the Journal of Headache and Pain. Adriana Della Pietra: Junior Editorial Board member of the Journal of Headache and Pain. David Garcia-Azorin: Junior Editorial Board member of the Journal of Headache and Pain, and Editorial Board member of Neurological Sciences; he has received personal compensation for consulting/advising from the World Health Organization; non-profit board membership in the Spanish Society of Neurology, and the European Union of Medical Specialist section of Neurology (UEMS); Research funding from the Carlos III Health Research Institute; and the Regional Health Administration (Gerencia Regional de Salud SACYL) in Castilla y Leon, Spain; speaker/travel grants/clinical trials from Teva, Allergan, Amgen, Eli Lilly, Lundbeck, Novartis, and Biohaven. Alejandro Labastida-Ramirez: Junior Editorial Board member of the Journal of Headache and Pain. Dilara Onan: Junior Editorial Board member of The Journal of Headache and Pain. Lanfranco Pellesi: Junior Editorial Board member of the Journal of Headache and Pain; has been employed by Lundbeck in the past two years. Mario Peres: Associate Editor of the Journal of Headache and Pain. Igor Petrušić: Junior Editorial Board member of the Journal of Headache and Pain. Bianca Raffaelli: Junior Editorial Board member of the Journal of Headache and Pain, Member of the Editorial Board of Frontiers in Neurology and BMC Neurology; Personal fees and honoraria from AbbVie, Eli Lilly, Lundbeck, Novartis, Teva; Research grants from Lundbeck, Novartis, German Research Foundation, German Migraine and Headache Society. Eloisa Rubio-Beltran: Junior Editorial Board member of the Journal of Headache and Pain; Review Editor for Frontiers in Neurology – Headache and Neurogenic Pain. Andreas Straube: Associate Editor of the Journal of Headache and Pain. Sebastian Straube: grants from the Workers’ Compensation Board of Alberta, The Government of Alberta, and the Alberta Medical Association; honoraria from the Occupational Medicine Specialist of Canada, the M.S.I. Foundation, and the Canadian Centre of Recovery Excellence. Tsubasa Takizawa: consultant/advisor and/or serves on an advisory board for Eli Lilly, Otsuka, Amgen, Pfizer, and Teijin; he has received speaker honoraria from Eli Lilly, Daiichi Sankyo, Otsuka, Amgen, Kowa, Kyowa Kirin, Eisai, UCB Japan, Takeda, and Santen Pharmaceutical and grant/funding from Eli Lilly, Pfizer and Tsumura outside the submitted work. Claudio Tana: Junior Editorial Board member of the Journal of Headache and Pain. Doga Vuralli: Junior Editorial Board member of the Journal of Headache and Pain. Marta Waliszewska-Prosół: Junior Editorial Board member of the Journal of Headache and Pain. Wei Wang: Junior Editorial Board member of the Journal of Headache and Pain, Section Editor of SN Comprehensive Clinical Medicine. Yonggang Wang: Associate Editor of the Journal of Headache and Pain and Chair of Chinese headache society of CSA. William Wells-Gatnik: Junior Editorial Board member of the Journal of Headache and Pain. Tissa Wijeratne: Associate Editor of the Journal of Headache and Pain. Paolo Martelletti: Editor-in-Chief of the Journal of Headache and Pain and of SN Comprehensive Clinical Medicine; EU Expert, European Medicines Agency. Marco Arruda, Matteo Castaldo, Xiangning Fan, Parisa Gazerani, Lou Grangeon, Licia Grazzi, Fu-Jung Hsiao, Keiko Ihara, Kristin Sophie Lange, Marco Lisicki, Alessia Marcassoli, Danilo Antonio Montisano, Agnese Onofri, Michela Tinelli, Massimiliano Valeriani, Simone Vigneri, and Tissa Wijeratne report no competing interests.
Additional information
Publisher’s Note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Rights and permissions
Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by-nc-nd/4.0/.
About this article
Cite this article
Raggi, A., Leonardi, M., Arruda, M. et al. Hallmarks of primary headache: part 1 – migraine. J Headache Pain 25, 189 (2024). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s10194-024-01889-x
Received:
Accepted:
Published:
DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s10194-024-01889-x