Next Article in Journal
How Do Gepotidacin and Zoliflodacin Stabilize DNA Cleavage Complexes with Bacterial Type IIA Topoisomerases? 1. Experimental Definition of Metal Binding Sites
Previous Article in Journal
Amino Acid Compound 2 (AAC2) Treatment Counteracts Insulin-Induced Synaptic Gene Expression and Seizure-Related Mortality in a Mouse Model of Alzheimer’s Disease
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Novel Calcitonin Gene-Related Peptide (CGRP) Interfering Migraine Therapies and Stroke—A Review

1
Department of Neurology, Medical University of Innsbruck, 6020 Innsbruck, Austria
2
VASCage—Centre on Clinical Stroke Research, 6020 Innsbruck, Austria
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2024, 25(21), 11685; https://doi.org/10.3390/ijms252111685
Submission received: 7 October 2024 / Revised: 29 October 2024 / Accepted: 29 October 2024 / Published: 30 October 2024
(This article belongs to the Special Issue Molecular Insights into Migraines)

Abstract

:
Migraine and stroke are neurological disorders with significant global prevalence and impact. Recent advances in migraine therapy have focused on the calcitonin gene-related peptide (CGRP) pathway. This review examines the shared pathomechanisms between migraine and stroke, with emphasis on the role of CGRP. We analyze the current literature on CGRP’s functions in cerebrovascular regulation, edema formation, neuroinflammation, and neuroprotection. CGRP acts as a potent vasodilator and plays a crucial role in trigeminovascular activation during migraine attacks. In stroke, CGRP has demonstrated neuroprotective effects by improving collateral circulation and reducing ischemia-reperfusion injury. Concerns have been raised about the potential impact of CGRP inhibitors on stroke risk and outcomes. Studies in animals suggest that CGRP receptor antagonists may worsen cerebral ischemia by impairing collateral flow. We discuss the implications of these findings for the use of CGRP-targeting therapies in migraine patients, especially those at increased risk of stroke. Additionally, we explore the complex interplay between CGRP, endothelial function, and platelet activity in both conditions. This review highlights the need for further research to elucidate the long-term cerebrovascular safety of CGRP pathway inhibitors and to identify potential subgroups of migraine patients who may be at higher risk of adverse cerebrovascular events with these novel therapies.

1. Physiological Effects of Calcitonin Gene-Related Peptide (CGRP)

The 37 amino acids that comprise CGRP, along with its two subtypes, alpha- and beta-CGRP, are mainly expressed in neurons of the central and peripheral nervous system. A high density of CGRP receptors has been reported in the trigeminal nerve axis, particularly in the trigeminal ganglion (TG). CGRP has also been suggested to occur in non-neuronal cells such as lymphocytes, mononuclear cells, adipocytes, and endothelial and epithelial cells [1,2,3]. Ischemia-induced TG activation leads to CGRP release in the meningeal vasculature. CGRP binds to CLR:RAMP1 (calcitonin receptor-like receptor, receptor activity modifying protein 1), a member of the CLR:RAMP family. The G protein-coupled CLR must heterodimerize with one of the three RAMPs 1–3 to become biologically active. Subsequent receptor downstream results in cAMP accumulation, calcium mobilization, and nitric oxide (NO) release, resulting in a following vasodilation [2,4]. Besides CGRP’s vasoactive potential, it sensitizes the NMDA (N-Methyl-D-Aspartate) receptor and activates direct and indirect anti-apoptotic and growth-factor signaling [5]. These findings propose CGRP as a crucial player in collateral blood flow during the acute phase of ischemic stroke and as a potential neuroprotectant in subarachnoid and intracerebral hemorrhage [6,7]. Additionally, CGRP has been found to upregulate Bcl2/Bax (B-cell lymphoma 2/Bcl-2-associated X protein), indicating mitochondrial stabilization and the prevention of apoptosis [8]. Recently, CGRP has been suggested to promote edema formation, tumor necrosis factor alpha elevation, and accumulation of neutrophils in mouse skin [9,10].
In the nociceptive system, CGRP represents one of the most involved neuropeptides. The nociceptive potential is mediated by the activation of the CLR:RAMP1 complex, which subsequently sensitizes cells [3,11]. It is distributed widely across the nociceptive axis, playing a significant role, particularly in those who experience migraine. With a lower affinity, CGRP also binds to CTR:RAMP1 (calcitonin receptor), a receptor complex in which amylin primarily acts as a ligand. CGRP is not the sole ligand for the CLR:RAMP family. Adrenomedullin (AM) has been observed to exhibit a preferential binding affinity for the CLR:RAMP2 (AM1), followed by the CLR:RAMP3 (AM2). Furthermore, evidence indicates that CLR:RAMP2/3 receptors possess substantial vasodilative, cardioprotective, and anti-apoptotic effects, whereas CTR:RAMP1 has not been demonstrated to exhibit such functions. Amylin (and CGRP) predominantly mediate an increase in gastrointestinal (GI) motility via CLR:RAMP1, which could potentially lead to a CGRP-mediated GI hyperactivity, as observed in some migraine patients [2,12].
Migraine is a very common neurovascular disorder that affects about 15% of the general population, predominantly women, and is the third leading cause of global years lived with disability [13]. CGRP levels are elevated in the jugular vein during an acute migraine attack [14]. Moreover, CGRP elevation has been found in tears and saliva during and between acute migraine attacks [15]. In chronic migraine patients, an elevation of plasma CGRP levels has been reported compared to healthy control groups. An intravenous (i.v.) infusion of CGRP in migraine patients compared to non-migraine patients induced a migraine-like headache in the migraine patients’ group, whereas in the control group, CGRP infusion induced only a mild headache without migraine-like symptoms, suggesting that migraine patients are more sensitive to CGRP [16,17,18]. The final and possibly most convincing evidence of CGRP’s profound involvement in the pathogenesis of migraine is the effectiveness of CGRP targeting therapies in migraine prevention and acute treatment.

2. Migraine Treatment with CGRP-Pathway Targeting Therapeutics

CGRP has now become the focus of numerous studies demonstrating a significant impact in the prevention and abortion of migraine attacks via monoclonal antibodies or gepants. Four monoclonal antibodies (Erenumab, Galcanezumab, Fremanezumab, and Eptinezumab) are currently on the market and have been approved for episodic migraine and chronic migraine. They are applied subcutaneously (Erenumab, Galcanezumab, and Fremanezumab) or intravenously (Eptinezumab). Due to their long half-life of 21–50 days, the frequency of administration varies between monthly for Erenumab, Galcanezumab, and Fremanezumab and every three months for Eptinezumab and high-dose Fremanezumab [19]. Erenumab binds to the canonical CGRP receptor CLR:RAMP1 at its extracellular domain. With lower affinity, it interacts with CLR:RAMP2, CLR:RAMP3, and CTR:RAMP1, physiologically activated by adrenomedullin (CLR:RAMP1 and CLR:RAMP2) and amylin (CTR:RAMP1). By binding at CTR:RAMP1, Erenumab could lead to decreased GI activity and the commonly reported side-effect of constipation [12]. Eptinezumab, Fremanezumab, and Galcanezumab bind to the receptor-binding site of endogenous alpha- and beta-CGRP, with no evidence of additional interactions (see Figure 1) [20]. Due to their high molecular weight of around 150,000 Da, mAbs are unable to cross the intact blood-brain barrier (BBB) in significant amounts. However, one study has raised data that mAbs may act within the vessel wall or indirectly influence cerebral vasomotor potential [21]. The primary site of action for mAbs is considered to be the trigeminal ganglion and the dura mater, characterized by a high density of CGRP receptors and a location outside of the BBB. Fluorescently labeled Fremanezumab has not been observed to cross the intact BBB in rats. However, high amounts were detected in dural vessels, the dura mater, the trigeminal ganglion, the C2 dorsal nerve ganglion, the sphenopalatine ganglion, and the superior sympathetic cervical ganglion. No signals of fluorescently labeled Fremanezumab were detected in the cortex, thalamus, hypothalamus, or spinal trigeminal nucleus [1,22,23].
With a mean molecular weight of 600 Da, gepants are of significantly smaller molecular weight than mAbs. This might implicate a simplified penetration of the BBB for gepants. However, only small amounts of MK-3207, a studied CGRP receptor antagonist, have been observed to cross the BBB. For Olcegepant, no central nervous point of action could be shown [24,25].
Gepants are the newest treatments approved for migraine headaches in the United States and some European countries. Ubrogepant is approved for acute migraine treatment, while Atogepant is approved for prophylactic migraine treatment in episodic migraine patients. Rimegepant is approved for both acute and prophylactic use in episodic migraine patients [19,26]. Gepants are small molecules that exhibit a high affinity for the RAMP1 subunit of the canonical CGRP receptor, with an almost insignificant affinity for the AM receptors CLR:RAMP2 and CLR:RAMP3. Due to their selectivity for the RAMP1 subunit, gepants can also interact with the amylin receptor CTR:RAMP1. In contrast to Erenumab, constipation is not a frequently reported adverse effect in patients undergoing therapy with gepants [20]. Unlike subcutaneously applied mAbs, gepants are available as oral treatments. Ubrogepant is a medication for acute migraine that should be taken in a manner like triptans for treating acute migraine headaches. Atogepant and Rimegepant are both used for the prevention of migraines. The frequency of use for Atogepant is daily, while for Rimegepant, every other day, and Rimegepant may also be used as acute treatment on days when no prophylactic therapy tablet is taken. Zavegepant is another FDA-approved gepant for acute migraine treatment administered by using a nasal spray [27]. Figure 1 illustrates the targets of all currently approved CGRP pathways targeting migraine drugs.

3. The Complex Relationship Between Migraine and Stroke: Insights into Risk Factors and Vascular Mechanisms

The relationship between migraine and stroke is well covered in other reviews [28,29,30,31]; therefore, we will focus on the most important points. In a large epidemiological study, migraine with aura (MA) was among the four most important risk factors for stroke and is more strongly associated with stroke and other cardiovascular diseases (CVD) than obesity or unfavorable lipid levels. However, the absolute risk of stroke in individuals aged < 45 years is modest [30]. However, the stroke risk within migraineurs with aura is higher for women, women who smoke, and women who take combined oral contraceptives. Therefore, it is recommended that women with MA avoid taking combined oral contraceptives, which may limit their contraceptive options [31]. Observational studies have suggested a possible association between patent foramen ovale (PFO) and migraines, as well as an increased incidence of PFO in patients with MA, resulting in a higher risk of paradoxical embolism [32].
In a Danish population-based, nationwide study, a connection between migraine with aura and cardioembolic stroke due to atrial fibrillation (AF) has been reported [33]. However, no correlation between MA and AF has been found in an Italian cohort study, suggesting a complex condition of stroke etiology in migraineurs [34]. Particularly in the male population, an increased risk of carotid artery dissection has been observed in migraine without aura (MO). Additional data also suggests an elevated risk for hemorrhagic stroke in migraineurs [30,35].
Increasing evidence suggests that migraine is a predictor of the risk of non-stroke vascular events, such as myocardial infarction, as well as a potent risk factor for any vascular event per se [30].
In support of these findings, studies have identified endothelial dysfunction in the migraine population. They have described elevated numbers of endothelial progenitor cells and endothelial microparticles in migraineurs, indicating a disrupted endothelium [36,37,38]. Moreover, migraineurs exhibited increased platelet adhesion and activation, especially female migraineurs with aura, showed elevated levels of fibrinogen and factor II, suggesting that these patients may be in a hypercoagulable state with altered platelet activity [39,40,41,42,43]. Although these findings are limited by inconsistent study designs and heterogeneous inclusion and exclusion criteria, one can assume that some endothelial alterations are present in migraineurs. However, it is difficult to conclude whether these endothelial alterations cause migraines or if they are a result of this complex neurological disorder. C-reactive protein (CRP), a marker of inflammation and vascular disease, is elevated in individuals who suffer from migraine. CRP levels were found to be elevated in patients with both MA and MO, especially in women between the ages of 19 and 34 with MO, compared to healthy women of the same age [44,45,46,47]. Other pro-inflammatory cytokines, IL-1 and IL-6, are also elevated in migraineurs, supporting the theory of a (pro-)inflammatory state in this population [39,48]. In support of these findings, studies have identified endothelial dysfunction in the migraine population and increased platelet adhesion and activation [37,38,39].

4. The Role of Attack Medication and Cortical Spreading Depression (CSD)

Triptans and ergot alkaloids are two drugs commonly used to treat acute migraines. Triptans selectively bind to the 5-HT1B, 5-HT1D, and 5-HT1F receptors, while ergot alkaloids have a wider range of receptor targets, resulting in a less favorable adverse effect profile compared to triptans. By binding to the 5-HT1B/1D receptors, triptans and ergots have a vasoconstrictive potential. It has been suggested that the excessive use of ergotamine may be associated with an increased risk of cerebrovascular and cardiovascular events. For triptans, this association has not been found. Nevertheless, it is advisable to exercise caution when prescribing triptans to patients with a history of cardiovascular or cerebrovascular disease [49,50].
Cortical spreading depression is a phenomenon characterized by a wave of depolarization that starts in the occipital lobe and migrates to the frontal lobe at a rate of approximately 1–2 mm/s. There is considerable evidence that CGRP plays a complex role in the development of CSD, primarily through its effects on cerebral blood vessels and the trigeminovascular system. However, CGRP-pathway mAbs have not been shown to reduce migraine aura in patients with MA [51,52]. Cortical spreading depression is associated with the clinical manifestation of a migraine aura, whereas CSD has also been observed in the penumbral tissue of ischemic stroke patients. CSD causes an ion translocation between neuronal cell bodies and the interstitium and an increase in energy consumption. In healthy brain tissue, increased energy turnover can be balanced by subsequent vasodilation. However, in energy-compromised tissue, such as the penumbra in ischemic stroke, this CSD could result in cell death. CSD has also been observed to result in the release of excitotoxic glutamate, which could lead to an additional risk of cell death in the penumbra [53]. Figure 2 identifies the most common and significant risk factors for stroke in migraineurs.

5. Molecular Implications of CGRP’s Role in Stroke

In acute ischemic stroke, the BBB becomes compromised, leading to increased permeability for substances normally restricted from entering the brain. A compromised BBB could also result in the formation of cerebral edema. CGRP has been implicated in promoting the development of edema, although conflicting findings suggest it may also play a protective role by preventing BBB injury and reducing infarct size [10,54,55]. Even if these results are conflicting, the beneficial effects of CGRP in ischemic events are far more prominent. CGRP and AM exhibit significant vasodilatory potential in the microvasculature [56]. During sudden drops in systemic blood pressure, CGRP helps maintain cerebral blood flow (CBF) [57]. However, reintroducing blood flow after ischemic conditions can lead to reperfusion injury due to oxidative stress, and CGRP offers both vasodilatory and antioxidative properties [58]. Additionally, pharmacologic preconditioning with morphine has been shown to stimulate systemic CGRP release, which reduces reperfusion injuries, indicating a potential interaction with the opioid signaling pathway [59]. This protective effect of CGRP can be diminished by triptans, which reduce CGRP release. CGRP demonstrates significant anti-inflammatory effects, particularly relevant in neuroprotection during stroke and other inflammatory conditions. CGRP inhibits the release of pro-inflammatory cytokines such as TNF-alpha, IL-1β, and IL-6 from various immune cells, effectively reducing the inflammatory response that can exacerbate tissue damage during and after a stroke [9,60].
CGRP’s neuroprotective anti-apoptotic effects were found in rodents mimicking ischemic events. It turned out that these effects are primarily mediated through the CREB (cyclic AMP response element-binding transcription factor) pathway, which upregulates Bcl-2 and subsequently reduces caspase-3 levels [61,62]. Caspase-3 is a critical pro-apoptotic molecule, while Bcl-2 stabilizes the mitochondrial membrane through the Bcl-2/BAX pathway, also influenced by CGRP. Thus, CGRP can enhance neuronal function and reduce apoptosis in brain tissues following ischemic injury by modulating MAPK (mitogen-activated protein kinase) pathways [8,63].
In the context of hemorrhagic stroke, CGRP has been found to decrease neuronal apoptosis, reduce the risk of vasospasm, and support nerve regeneration, respectively [64]. Not only has CGRP shown beneficial effects in stroke, but AM has demonstrated anti-inflammatory effects, including a reduction in oxidant production in the cerebellum [63,64]. Table 1 lists CGRP’s potential roles and the related effects in stroke.
In brief, CGRP plays a complex and predominantly beneficial role in stroke, exhibiting vasodilatory, neuroprotective, and anti-inflammatory effects. While its impact on cerebral edema remains controversial, CGRP helps maintain cerebral blood flow, reduces reperfusion injury, inhibits pro-inflammatory cytokine release, and demonstrates anti-apoptotic properties through various molecular pathways, suggesting its potential importance in stroke management and therapy.

6. Cardiovascular Risk Signals in Clinical Trials Using Monoclonal Antibodies and Gepants

In a trial involving 60 patients with MO who received 70 mg of Erenumab subcutaneously every four weeks, vasomotor reactivity (VMR) and brachial flow-mediated dilation (FMD) were monitored during follow-up. The results showed no differences in cerebral or systemic vasomotor activity compared to the healthy control group [65]. However, there was little effect on cerebral hemodynamics for CGRP-targeted mAbs, especially in migraineurs responding to mAb preventive treatment. The decrease in cerebral blood flow velocity (v) in this subgroup should be critically interpreted, as the number of participants was small [6]. A meta-analysis of 3300 participants of five placebo-controlled RCTs of Erenumab raised no concerns regarding the CV risk profile [66]. However, all analyzed studies recruited subjects aged ≤ 65 years. Of all participants, 143 were aged ≥ 60 years, and 63 were enrolled in the treatment arm, representing less than 10% of all participants. It is therefore of utmost importance to acknowledge that all studies finally leading to market authorization used strict inclusion and exclusion criteria, mostly enrolling patients lacking CVD or other relevant preconditions. A post-hoc pooled analysis of long-term clinical trial data on Erenumab examined adverse events in relation to participants’ estimated 10-year CV risk. Across all risk categories (no risk, low, moderate, and high), Erenumab exposure did not lead to any significant AE signals [67].
For Eptinezumab, Fremanezumab, and Galcanezumab, RCTs included patients up to 70–75 years old with non-concerning CV safety data [19,68]. However, all the abovementioned RCTs excluded patients with pre-existing CVD, and the number of participants ≥ 60 years old was low. This highlights once more the limited evidence of RCTs regarding efficacy and safety in elderly patients, especially in those with preexisting CVDs.
A Spanish multicenter observational study enrolled patients aged 65 to 87 years, evaluating the efficacy and safety of Erenumab, Fremanezumab, and Galcanezumab [69]. Participants had a high prevalence of comorbidities, including dyslipidemia, hypertension, anxiety, depression, and ischemic cardiovascular disease. In this real-world observational study, elderly migraineurs showed a similar benefit in migraine prevention compared to younger migraineurs. No signals for CVD adverse events were raised in this elderly cohort with pre-existing comorbidities.
CVD safety studies for mAbs are not only limited by their preselected patient cohort but also by a lack of long-term safety data. However, post-marketing pharmacovigilance is common in many countries, and so far, no CVD signals have been documented. Additionally, a 5-year open-label follow-up of Erenumab has shown no CV safety concerns [70].
As gepants are the most recently approved CGRP receptor inhibitors in the treatment of migraine, no long-term CV safety data are available. Nevertheless, all phase 3 clinical trials for Atogepant, Rimegepant, and Ubrogepant showed no increase in CVD adverse events in treated patients [19,27,68]. A recent network meta-analysis found no CV concerns for Atogepant and Rimegepant in phase 2/3 studies [19]. These studies, however, excluded or underrepresented subjects > 65 years of age and patients with a history of CVD. A recently conducted controlled, open-label industry-sponsored study included 735 participants with CV risk factors to evaluate the safety of Rimegepant for acute migraine treatment with doses up to once daily [71]. The follow-up was one year, and no CV concerns were raised. However, patients with former CV events and uncontrolled CVD were excluded

7. Cardiovascular Risk Signals in Non-Clinical Studies

A study by Inge A. Mulder and colleagues demonstrated a significant effect of gepants on cerebral infarct volume and functional outcome after transient ischemic attack (TIA) or stroke-mimicking experiments [72]. Two gepants, Olcegepant and Rimegepant, were administered as single or repeated doses. Mice were randomized into two arms: the treatment arm or a matching placebo control arm.
To simulate a TIA, the MCA was occluded for 12 min. Prior to MCA occlusion, a single dose of Olcegepant (1 mg/kg, n = 19) or placebo (n = 18) was administered. In the Olcegepant-pretreated group, 14 out of 19 mice developed infarction, whereas 6 out of 18 mice in the naive group developed infarction.
In the second experiment, Olcegepant (1 mg/kg) or vehicle was administered before MCA occlusion for 60 min. After 60 min of occlusion, all mice developed infarction. In Olcegepant-pretreated mice, the infarct volume was more than twice as large as in Olcegepant-naive mice.
Rimegepant was further evaluated. Prior to 60 min of MCA occlusion, 100 mg/kg of Rimegepant or placebo was administered. In the Rimegepant group, 6/8 mice died compared to 0/8 in the vehicle group.
In the above-mentioned TIA model, at a dose of 10 mg/kg Rimegepant versus placebo, all mice developed infarction. In the Rimegepant-pretreated group, the infarct size was 60% larger than in the vehicle group. It should be noted that the doses of Rimegepant studied, 10 mg/kg and 100 mg/kg, are 10–100 times higher than the approved human dosage.
In the fourth set of experiments, mice were treated with 0.1 mg or 1 mg/kg of Olcegepant for two weeks. Both dose regimens increased infarct volume and neurological deficits comparable to those of a single dose of Olcegepant.
Nevertheless, other animal models investigating gepants have demonstrated that short-term inhibition of CGRP signaling does not significantly alter hemodynamic parameters, including heart rate, blood pressure, cardiac output, or coronary flow. Furthermore, no exacerbation in ischemic severity has been observed. These findings suggest that the acute blockade of CGRP-mediated pathways exerts minimal influence on systemic cardiovascular dynamics, which is in accordance with the results of the major clinical RCTs [73].
In a study by L. Ohlsson et al., CGRP receptors were blocked with Erenumab in isolated human cranial arteries, demonstrating no significant effect on vasoconstriction or vasodilation [74].

8. CGRP Levels in Acute Stroke Patients

Bhar-Hosseini et al. [75] evaluated CGRP levels in acute stroke patients. Blood samples were drawn from a peripheral vein at admission and again the following morning. The study cohort had a mean age of 78 (70–86) years, with 37% women and a mean NIHSS of 13 (7–18) on the day of admission. They observed normally distributed CGRP levels with no changes from admission to the morning after admission. A multimodal CT was performed at baseline to evaluate leptomeningeal collaterals, penumbral size, and the ischemic core. Final infarct volumes were measured by CT or MRI within 24 h after admission. The study found no correlation between admission or next-day CGRP levels and collateral flow, penumbral size, or infarct size. The 90-day modified Rankin Scale (mRS) was evaluated to determine any correlation with admission or next-day CGRP levels, but no such correlation was found. It is worth noting that data on CGRP in acute stroke patients are scarce, and this study is the only prospective study to measure CGRP levels in such patients.

9. CGRP and Other Vascular Disease

CGRP is one of the most potent vasodilators in humans, primarily acting on the microvascular bed. CGRP does not appear to play a key role in physiological blood pressure regulation, as evidenced by studies in which CGRP receptors were blocked in healthy volunteers or CGRP knockout mice [76]. However, under pathophysiologic conditions, CGRP has been found to influence arterial hypertension [77]. Some studies have reported elevated systolic and diastolic blood pressure in migraine patients treated with mAbs, while other phase 2/3 and post-marketing studies have not found such an association [78,79].
In patients with stable angina pectoris, intra-coronary injection of CGRP has been shown to delay the onset of myocardial ischemia [80]. In mice with acute myocardial infarction (MI), CGRP has been reported to significantly reduce ischemia/reperfusion injury [81]. In congestive heart failure, i.v. α-CGRP infusion improved functional cardiac parameters [82].
In mice, the administration of exogenous CGRP has been demonstrated to have antiplatelet effects by inhibiting platelet-derived tissue factor [83].

10. Discussion

CGRP plays a crucial role in (cranial) vasodilation. Blocking CGRP receptors with gepants can worsen ischemic stroke in mice. In contrast, none of the large, placebo-controlled, phase 2/3 trials of mAbs or gepants have raised concerns about vascular safety. These findings suggest that there may be compensatory mechanisms in migraineurs without cardiovascular diseases when the CGRP pathway is blocked. Whether this compensation is triggered by other peptides, such as adrenomedullin, remains unknown. It is important to note that these RCTs, with some exceptions, lasted no longer than 12 weeks to 6 months and mainly excluded patients with a history of CVD.
The use of mAbs and gepants as preventive treatments for migraine has been increasing in recent years, with the possibility that they may become the first-line preventive treatment for migraine in the future.
Migraine is a neurological disorder that imposes a significant burden on patients. Therefore, prophylactic treatment with targeted therapies is highly justified. Traditional oral preventive treatments have been associated with a higher number of adverse events, and some are contraindicated in women of childbearing potential, who represent the majority of migraineurs. CGRP-interfering drugs have demonstrated fewer adverse events with higher efficacy, making them an excellent option for many patients. Women < 45 with MA have a higher risk of ischemic stroke than men with MA, a risk that further increases if they smoke or use oral contraceptives. Although the role of CGRP in migraine aura pathogenesis is not yet fully understood, there is no evidence indicating that women with MA on CGRP-targeted therapies face additional stroke risk. However, patients with cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL) should avoid CGRP-targeted treatment, as CGRP primarily acts on small blood vessels [84]. Yet, the predominant age of patients receiving these targeted migraine prophylactic treatments is ≤65 years. As the population of migraineurs continues to age, more patients on CGRP-interfering therapies may face stroke, MI, and other cardiovascular diseases.
A multicenter observational study was conducted on patients aged ≥ 65 years with cardiovascular comorbidities, which found no safety issues for preventive migraine treatment with mAbs [69]. However, the impact of a (long-term) blocked CGRP pathway in combination with coincident acute ischemic stroke and other CVDs remains unclear.

10.1. Concerns in Acute Vascular Events

Monoclonal antibodies and gepants cannot cross the BBB in significant amounts in individuals with an intact BBB. In contrast, one study has shown an alteration of cerebrovascular vasodilation by mAbs in rats [21]. In cases of acute ischemic stroke or subarachnoid hemorrhage (SAH), where the BBB is compromised, mAbs/gepants may diffuse into areas that are normally protected by the BBB. In ischemic stroke, vasodilation plays a crucial role in maintaining sufficient blood supply to the vulnerable penumbra [53]. Conversely, the CGRP pathway may be a potential target for preventing post-ischemic edema. However, the evidence supporting this hypothesis is inconclusive, with conflicting results reported in the literature. Additionally, the only study to investigate the role of CGRP in edema utilized a murine model of skin injury [10]. In acute stroke patients, CGRP levels at admission and the morning after admission did not correlate with infarct size or functional outcome after 90 days following ischemic stroke [75]. Notably, since plasma was collected from a peripheral vein, no conclusions can be drawn regarding CGRP levels in brain-supplying vessels. Obtaining comparable CGRP levels is not a trivial task, given the existing data on the half-life of CGRP of approximately 7 min [85]. A precise description of pre-analytic processing is crucial, calling for future studies investigating the role of CGRP in various vascular diseases.

10.2. Dual CGRP-Targeted Treatment by mAbs and Gepants

An increasing number of patients are receiving preventive migraine treatment with mAbs and are using gepants as acute attack therapy. However, there are currently only very limited safety or efficacy studies available investigating this combination. It is, therefore, essential to determine whether these patients face an increased risk of stroke per se or a worse prognosis in the event of an acute stroke.
As mentioned above, Erenumab binds with a high affinity to CLR:RAMP1 and a lower affinity to CLR:RAMP2-3. Eptinezumab, Fremanezumab, and Galcanezumab target the CGRP ligand. Gepants are selective for the RAMP1 subunit of CLR:RAMP1 but may also act at CTR:RAMP1 (amylin receptor). A randomized, open-label study evaluated the safety profile of dual CGRP-targeting therapy with the preventive use of Erenumab or Galcanezumab and Ubrogepant for acute treatment [86]. No safety issues or pharmacokinetic interactions were reported after 45 days of follow-up. Real-world data also suggest a favorable risk profile of dual CGRP-targeted therapy [87].
However, in human arteries, the combination of Erenumab with Rimegepant or Olcegepant increases the threshold at which CGRP induces vasodilation. In contrast, isolated higher doses of Erenumab alone do not show such an effect [88]. This could potentially endanger penumbral tissue in acute stroke patients on dual therapy with a compromised BBB, as both monoclonal mAbs and gepants may directly affect the microvasculature or brain tissue. The vasodilative and anti-apoptotic effects of CGRP could be inhibited by these drugs [53]. Unlike CGRP, other peptides that bind to these receptors, such as AM and amylin, are not blocked by gepants, which could allow for compensatory mechanisms in acute stroke settings [88]. Since gepants for acute treatment are infrequently used and have a shorter half-life compared to monoclonal mAbs, this may not pose a significant concern in clinical practice when treating acute migraines in migraineurs with absent CVD.

10.3. Molecular Findings Leading to Clinical Implications

Based on the abovementioned involvement of CGRP in stroke and other acute CV events, as well as limited data on CGRP antagonism and acute stroke, some suggestions can be made for clinical practice:
In patients > 65 years of age and relevant CVD, the individual benefits and harms of a newly initiated migraine treatment specifically acting within the CGRP pathway should be critically reconsidered. In these cases, beta-blockers or candesartan, which offer more favorable cardiovascular risk profiles, should be considered as alternative treatments. If CGRP-targeted therapy is necessary, gepants, which have a shorter half-life compared to mAbs, may be a better choice for managing potential side effects.
In patients with coincident acute stroke currently under CGRP-interfering migraine preventive treatment, these migraine treatments should be stopped immediately. Due to the long half-life of mAbs, short-term recovery could be jeopardized. Motor recovery after a stroke generally occurs within the first three months, while other remodeling processes can continue for up to one year [89,90].
Thus, reinitiating CGRP-targeted therapy after a stroke should be carefully considered on an individual basis. It is important to take stroke etiology into account, as CGRP plays a significant role in the microvasculature. Therefore, patients with microvascular disease should avoid these treatments [76]. For other post-stroke patients, gepants may be a safer alternative compared to mAbs, given their shorter half-life.
These recommendations emphasize a personalized approach to managing migraine for patients with elevated cardiovascular risk or a history of stroke, aiming to achieve a balance between treatment efficacy and patient safety.

11. Conclusions

In summary, while CGRP plays a crucial role in cranial vasodilation, studies have shown conflicting effects of CGRP-targeting therapies on ischemic stroke development in mice versus humans. Large clinical trials have not raised concerns about vascular safety, suggesting potential compensatory mechanisms in migraine sufferers without cardiovascular disease. However, the impact of CGRP pathway blockage on the outcome of coincidental acute stroke or other vascular events remains unclear.
Current data suggest that a cautious approach to CGRP-targeted therapies is necessary, particularly in patients with existing cardiovascular comorbidities. Further research is imperative to guide clinical decision-making and ensure patient safety. This research must include real-world studies, especially in older populations with cardiovascular comorbidities.

Author Contributions

Conceptualization, M.T.E. and G.B.; writing—original draft preparation, M.T.E. and G.B.; writing—review and editing, M.T.E., F.F., K.K., A.K., M.K. and G.B; visualization, F.F.; supervision, M.K. and G.B. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Acknowledgments

Figures were created with Adobe Photoshop (Adobe Inc., San José, CA, USA, Version 26.0), Adobe In Design (Adobe Inc., San José, CA, USA, Version 2024), Adobe Illustrator (Adobe Inc., San José, CA, USA, Version 29.0) and BioRender (BioRender, Toronto, ON, Canada).

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Eftekhari, S.; Salvatore, C.A.; Johansson, S.; Chen, T.-B.; Zeng, Z.; Edvinsson, L. Localization of CGRP, CGRP Receptor, PACAP and Glutamate in Trigeminal Ganglion. Relation to the Blood-Brain Barrier. Brain Res. 2015, 1600, 93–109. [Google Scholar] [CrossRef] [PubMed]
  2. Balint, L.; Nelson-Maney, N.P.; Tian, Y.; Serafin, S.D.; Caron, K.M. Clinical Potential of Adrenomedullin Signaling in the Cardiovascular System. Circ. Res. 2023, 132, 1185–1202. [Google Scholar] [CrossRef] [PubMed]
  3. Bernstein, C.; Burstein, R. Sensitization of the Trigeminovascular Pathway: Perspective and Implications to Migraine Pathophysiology. J. Clin. Neurol. 2012, 8, 89–99. [Google Scholar] [CrossRef]
  4. Altamura, C.; Vernieri, F. Commentary: Enhanced Hemodynamic and Clinical Response to ACGRP in Migraine Patients—A TCD Study. Front. Neurol. 2021, 12, 663818. [Google Scholar] [CrossRef]
  5. Okutsu, Y.; Takahashi, Y.; Nagase, M.; Shinohara, K.; Ikeda, R.; Kato, F. Potentiation of NMDA Receptor-Mediated Synaptic Transmission at the Parabrachial-Central Amygdala Synapses by CGRP in Mice. Mol. Pain 2017, 13. [Google Scholar] [CrossRef]
  6. Carter, S.C.; Cucchiara, B.; Reehal, N.; Hamilton, K.; Kaiser, E.A.; Favilla, C.G. Effect of CGRP Inhibitors on Interictal Cerebral Hemodynamics in Individuals with Migraine. Front. Neurol. 2024, 15, 1399792. [Google Scholar] [CrossRef]
  7. Zhai, L.; Sakurai, T.; Kamiyoshi, A.; Ichikawa-Shindo, Y.; Kawate, H.; Tanaka, M.; Xian, X.; Hirabayashi, K.; Dai, K.; Cui, N.; et al. Endogenous Calcitonin Gene-Related Peptide Suppresses Ischemic Brain Injuries and Progression of Cognitive Decline. J. Hypertens. 2018, 36, 876–891. [Google Scholar] [CrossRef]
  8. Guo, Z.; Liu, N.; Chen, L.; Zhao, X.; Li, M.R. Independent Roles of CGRP in Cardioprotection and Hemodynamic Regulation in Ischemic Postconditioning. Eur. J. Pharmacol. 2018, 828, 18–25. [Google Scholar] [CrossRef]
  9. Sorby-Adams, A.J.; Marcoionni, A.M.; Dempsey, E.R.; Woenig, J.A.; Turner, R.J. The Role of Neurogenic Inflammation in Blood-Brain Barrier Disruption and Development of Cerebral Oedema Following Acute Central Nervous System (CNS) Injury. Int. J. Mol. Sci. 2017, 18, 1788. [Google Scholar] [CrossRef]
  10. Zarban, A.A.; Chaudhry, H.; de Sousa Valente, J.; Argunhan, F.; Ghanim, H.; Brain, S.D. Elucidating the Ability of CGRP to Modulate Microvascular Events in Mouse Skin. Int. J. Mol. Sci. 2022, 23, 12246. [Google Scholar] [CrossRef]
  11. Eftekhari, S.; Salvatore, C.A.; Calamari, A.; Kane, S.A.; Tajti, J.; Edvinsson, L. Differential Distribution of Calcitonin Gene-Related Peptide and Its Receptor Components in the Human Trigeminal Ganglion. Neuroscience 2010, 169, 683–696. [Google Scholar] [CrossRef] [PubMed]
  12. Hage La Cour, S.; Juhler, K.; Kogelman, L.J.A.; Olesen, J.; Klærke, D.A.; Kristensen, D.M.; Jansen-Olesen, I. Characterization of Erenumab and Rimegepant on Calcitonin Gene-Related Peptide Induced Responses in Xenopus Laevis Oocytes Expressing the Calcitonin Gene-Related Peptide Receptor and the Amylin-1 Receptor. J. Headache Pain 2022, 23, 59. [Google Scholar] [CrossRef] [PubMed]
  13. Steinmetz, J.D.; Seeher, K.M.; Schiess, N.; Nichols, E.; Cao, B.; Servili, C.; Cavallera, V.; Cousin, E.; Hagins, H.; Moberg, M.E.; et al. 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. 2024, 23, 344–381. [Google Scholar] [CrossRef] [PubMed]
  14. Sarchielli, P.; Alberti, A.; Codini, M.; Floridi, A.; Gallai, V. Nitric Oxide Metabolites, Prostaglandins and Trigeminal Vasoactive Peptides in Internal Jugular Vein Blood during Spontaneous Migraine Attacks. Cephalalgia 2000, 20, 907–918. [Google Scholar] [CrossRef]
  15. Kamm, K.; Straube, A.; Ruscheweyh, R. Calcitonin Gene-Related Peptide Levels in Tear Fluid Are Elevated in Migraine Patients Compared to Healthy Controls. Cephalalgia 2019, 39, 1535–1543. [Google Scholar] [CrossRef]
  16. Cernuda-Morollon, E.; Larrosa, D.; Ramon, C.; Vega, J.; Martinez-Camblor, P.; Pascual, J.; Cernuda-Morollón, E.; Larrosa, D.; Ramón, C.; Vega, J.; et al. Interictal Increase of CGRP Levels in Peripheral Blood as a Biomarker for Chronic Migraine. Neurology 2013, 81, 1191–1196. [Google Scholar] [CrossRef]
  17. Hansen, J.M.; Hauge, A.W.; Olesen, J.; Ashina, M. Calcitonin Gene-Related Peptide Triggers Migraine-like Attacks in Patients with Migraine with Aura. Cephalalgia 2010, 30, 1179–1186. [Google Scholar] [CrossRef]
  18. Guo, S.; Vollesen, A.L.H.; Olesen, J.; Ashina, M. Premonitory and Nonheadache Symptoms Induced by CGRP and PACAP38 in Patients with Migraine. Pain 2016, 157, 2773–2781. [Google Scholar] [CrossRef]
  19. Messina, R.; Huessler, E.-M.; Puledda, F.; Haghdoost, F.; Lebedeva, E.R.; Diener, H.-C. Safety and Tolerability of Monoclonal Antibodies Targeting the CGRP Pathway and Gepants in Migraine Prevention: A Systematic Review and Network Meta-Analysis. Cephalalgia 2023, 43, 3331024231152169. [Google Scholar] [CrossRef]
  20. Bhakta, M.; Vuong, T.; Taura, T.; Wilson, D.S.; Stratton, J.R.; Mackenzie, K.D. Migraine Therapeutics Differentially Modulate the CGRP Pathway. Cephalalgia 2021, 41, 499–514. [Google Scholar] [CrossRef]
  21. Edvinsson, L.; Nilsson, E.; Jansen-Olesen, I. Inhibitory Effect of BIBN4096BS, CGRP 8-37, a CGRP Antibody and an RNA-Spiegelmer on CGRP Induced Vasodilatation in the Perfused and Non-Perfused Rat Middle Cerebral Artery. Br. J. Pharmacol. 2007, 150, 633–640. [Google Scholar] [CrossRef] [PubMed]
  22. Lundblad, C.; Haanes, K.A.; Grände, G.; Edvinsson, L. Experimental Inflammation Following Dural Application of Complete Freund’s Adjuvant or Inflammatory Soup Does Not Alter Brain and Trigeminal Microvascular Passage. J. Headache Pain 2015, 16, 91. [Google Scholar] [CrossRef]
  23. Noseda, R.; Schain, A.J.; Melo-Carrillo, A.; Tien, J.; Stratton, J.; Mai, F.; Strassman, A.M.; Burstein, R. Fluorescently-Labeled Fremanezumab Is Distributed to Sensory and Autonomic Ganglia and the Dura but Not to the Brain of Rats with Uncompromised Blood Brain Barrier. Cephalalgia 2020, 40, 229–240. [Google Scholar] [CrossRef] [PubMed]
  24. Salvatore, C.A.; Moore, E.L.; Calamari, A.; Cook, J.J.; Michener, M.S.; O’Malley, S.; Miller, P.J.; Sur, C.; Williams, D.L.J.; Zeng, Z.; et al. Pharmacological Properties of MK-3207, a Potent and Orally Active Calcitonin Gene-Related Peptide Receptor Antagonist. J. Pharmacol. Exp. Ther. 2010, 333, 152–160. [Google Scholar] [CrossRef] [PubMed]
  25. Christensen, S.L.; Ernstsen, C.; Olesen, J.; Kristensen, D.M. No Central Action of CGRP Antagonising Drugs in the GTN Mouse Model of Migraine. Cephalalgia 2020, 40, 924–934. [Google Scholar] [CrossRef] [PubMed]
  26. Dodick, D.W.; Lipton, R.B.; Ailani, J.; Lu, K.; Finnegan, M.; Trugman, J.M.; Szegedi, A. Ubrogepant for the Treatment of Migraine. N. Engl. J. Med. 2019, 381, 2230–2241. [Google Scholar] [CrossRef]
  27. Lipton, R.B.; Croop, R.; Stock, D.A.; Madonia, J.; Forshaw, M.; Lovegren, M.; Mosher, L.; Coric, V.; Goadsby, P.J. 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. 2023, 22, 209–217. [Google Scholar] [CrossRef]
  28. Sacco, S.; Harriott, A.M.; Ayata, C.; Ornello, R.; Bagur, R.; Jimenez-Ruiz, A.; Sposato, L.A. Microembolism and Other Links Between Migraine and Stroke: Clinical and Pathophysiologic Update. Neurology 2023, 100, 716–726. [Google Scholar] [CrossRef]
  29. Øie, L.R.; Kurth, T.; Gulati, S.; Dodick, D.W. Migraine and Risk of Stroke. J. Neurol. Neurosurg. Psychiatry 2020, 91, 593–604. [Google Scholar] [CrossRef]
  30. Kurth, T.; Rist, P.M.; Ridker, P.M.; Kotler, G.; Bubes, V.; Buring, J.E. Association of Migraine With Aura and Other Risk Factors with Incident Cardiovascular Disease in Women. JAMA 2020, 323, 2281–2289. [Google Scholar] [CrossRef]
  31. Ibrahimi, K.; Rist, P.M.; Carpenet, C.; Rohmann, J.L.; Buring, J.E.; Maassen van den Brink, A.; Kurth, T. Vascular Risk Score and Associations with Past, Current, or Future Migraine in Women: Cohort Study. Neurology 2022, 99, e1694–e1701. [Google Scholar] [CrossRef] [PubMed]
  32. West, B.H.; Noureddin, N.; Mamzhi, Y.; Low, C.G.; Coluzzi, A.C.; Shih, E.J.; Gevorgyan Fleming, R.; Saver, J.L.; Liebeskind, D.S.; Charles, A.; et al. Frequency of Patent Foramen Ovale and Migraine in Patients with Cryptogenic Stroke. Stroke 2018, 49, 1123–1128. [Google Scholar] [CrossRef] [PubMed]
  33. Adelborg, K.; Szépligeti, S.K.; Holland-Bill, L.; Ehrenstein, V.; Horváth-Puhó, E.; Henderson, V.W.; Sørensen, H.T. Migraine and Risk of Cardiovascular Diseases: Danish Population Based Matched Cohort Study. BMJ 2018, 360, k96. [Google Scholar] [CrossRef] [PubMed]
  34. de Giuli, V.; Grassi, M.; Locatelli, M.; Gamba, M.; Morotti, A.; Bonacina, S.; Mazzoleni, V.; Pezzini, D.; Magoni, M.; Monastero, R.; et al. Cardiac Sources of Cerebral Embolism in People with Migraine. Eur. J. Neurol. 2021, 28, 516–524. [Google Scholar] [CrossRef]
  35. de Giuli, V.; Grassi, M.; Lodigiani, C.; Patella, R.; Zedde, M.; Gandolfo, C.; Zini, A.; DeLodovici, M.L.; Paciaroni, M.; Del Sette, M.; et al. Association Between Migraine and Cervical Artery Dissection: The Italian Project on Stroke in Young Adults. JAMA Neurol. 2017, 74, 512–518. [Google Scholar] [CrossRef]
  36. Lee, S.-T.; Chu, K.; Jung, K.-H.; Kim, D.-H.; Kim, E.-H.; Choe, V.N.; Kim, J.-H.; Im, W.-S.; Kang, L.; Park, J.-E.; et al. Decreased Number and Function of Endothelial Progenitor Cells in Patients with Migraine. Neurology 2008, 70, 1510–1517. [Google Scholar] [CrossRef]
  37. Oterino, A.; Toriello, M.; Palacio, E.; Quintanilla, V.G.; Ruiz-Lavilla, N.; Montes, S.; de la Vega, M.S.; Martinez-Nieto, R.; Castillo, J.; Pascual, J. Analysis of Endothelial Precursor Cells in Chronic Migraine: A Case-Control Study. Cephalalgia 2013, 33, 236–244. [Google Scholar] [CrossRef]
  38. Liman, T.G.; Bachelier-Walenta, K.; Neeb, L.; Rosinski, J.; Reuter, U.; Böhm, M.; Endres, M. Circulating Endothelial Microparticles in Female Migraineurs with Aura. Cephalalgia 2015, 35, 88–94. [Google Scholar] [CrossRef]
  39. Tietjen, G.E.; Khubchandani, J.; Herial, N.; Palm-Meinders, I.H.; Koppen, H.; Terwindt, G.M.; van Buchem, M.A.; Launer, L.J.; Ferrari, M.D.; Kruit, M.C. Migraine and Vascular Disease Biomarkers: A Population-Based Case-Control Study. Cephalalgia 2018, 38, 511–518. [Google Scholar] [CrossRef]
  40. Fischer, M.; Gaul, C.; Shanib, H.; Holle, D.; Loacker, L.; Griesmacher, A.; Lackner, P.; Broessner, G. Markers of Endothelial Function in Migraine Patients: Results from a Bi-Center Prospective Study. Cephalalgia 2015, 35, 877–885. [Google Scholar] [CrossRef]
  41. Tietjen, G.E.; Al-Qasmi, M.M.; Athanas, K.; Dafer, R.M.; Khuder, S.A. Increased von Willebrand Factor in Migraine. Neurology 2001, 57, 334–336. [Google Scholar] [CrossRef] [PubMed]
  42. Tietjen, G.E.; Al-Qasmi, M.M.; Athanas, K.; Utley, C.; Herial, N.A. Altered Hemostasis in Migraineurs Studied with a Dynamic Flow System. Thromb. Res. 2007, 119, 217–222. [Google Scholar] [CrossRef] [PubMed]
  43. Tietjen, G.E.; Herial, N.A.; Utley, C.; White, L.; Yerga-Woolwine, S.; Joe, B. Association of von Willebrand Factor Activity with ACE I/D and MTHFR C677T Polymorphisms in Migraine. Cephalalgia 2009, 29, 960–968. [Google Scholar] [CrossRef]
  44. Vanmolkot, F.H.; de Hoon, J.N. Increased C-Reactive Protein in Young Adult Patients with Migraine. Cephalalgia 2007, 27, 843–846. [Google Scholar] [CrossRef]
  45. Tietjen, G.E.; Herial, N.A.; White, L.; Utley, C.; Kosmyna, J.M.; Khuder, S.A. Migraine and Biomarkers of Endothelial Activation in Young Women. Stroke 2009, 40, 2977–2982. [Google Scholar] [CrossRef]
  46. Hamed, S.A.; Hamed, E.A.; Ezz Eldin, A.M.; Mahmoud, N.M. Vascular Risk Factors, Endothelial Function, and Carotid Thickness in Patients with Migraine: Relationship to Atherosclerosis. J. Stroke Cerebrovasc. Dis. 2010, 19, 92–103. [Google Scholar] [CrossRef]
  47. Güzel, I.; Taşdemir, N.; Celik, Y. Evaluation of Serum Transforming Growth Factor Β1 and C-Reactive Protein Levels in Migraine Patients. Neurol. Neurochir. Pol. 2013, 47, 357–362. [Google Scholar] [CrossRef]
  48. Domínguez, C.; López, A.; Ramos-Cabrer, P.; Vieites-Prado, A.; Pérez-Mato, M.; Villalba, C.; Sobrino, T.; Rodriguez-Osorio, X.; Campos, F.; Castillo, J.; et al. Iron Deposition in Periaqueductal Gray Matter as a Potential Biomarker for Chronic Migraine. Neurology 2019, 92, e1076–e1085. [Google Scholar] [CrossRef]
  49. Roberto, G.; Raschi, E.; Piccinni, C.; Conti, V.; Vignatelli, L.; D’Alessandro, R.; De Ponti, F.; Poluzzi, E. Adverse Cardiovascular Events Associated with Triptans and Ergotamines for Treatment of Migraine: Systematic Review of Observational Studies. Cephalalgia 2015, 35, 118–131. [Google Scholar] [CrossRef]
  50. de Vries, T.; Villalón, C.M.; MaassenVanDenBrink, A. Pharmacological Treatment of Migraine: CGRP and 5-HT beyond the Triptans. Pharmacol. Ther. 2020, 211, 107528. [Google Scholar] [CrossRef]
  51. Close, L.N.; Eftekhari, S.; Wang, M.; Charles, A.C.; Russo, A.F. Cortical Spreading Depression as a Site of Origin for Migraine: Role of CGRP. Cephalalgia 2019, 39, 428–434. [Google Scholar] [CrossRef] [PubMed]
  52. Cresta, E.; Bellotti, A.; Rinaldi, G.; Corbelli, I.; Sarchielli, P. Effect of Anti-CGRP-Targeted Therapy on Migraine Aura: Results of an Observational Case Series Study. CNS Neurosci. Ther. 2024, 30, e14595. [Google Scholar] [CrossRef] [PubMed]
  53. Sueiras, M.; Thonon, V.; Santamarina, E.; Sánchez-Guerrero, Á.; Poca, M.A.; Quintana, M.; Riveiro, M.; Sahuquillo, J. Cortical Spreading Depression Phenomena Are Frequent in Ischemic and Traumatic Penumbra: A Prospective Study in Patients with Traumatic Brain Injury and Large Hemispheric Ischemic Stroke. J. Clin. Neurophysiol. 2021, 38, 47–55. [Google Scholar] [CrossRef]
  54. Chang, C.L.; Cai, Z.; Hsu, S.Y.T. Sustained Activation of CLR/RAMP Receptors by Gel-Forming Agonists. Int. J. Mol. Sci. 2022, 23, 13408. [Google Scholar] [CrossRef]
  55. Liu, G.; Cai, H.; Xu, X.; Liu, Z.; Wang, Q.; Feng, G.; Li, Y.; Xu, C.; Li, Z. The Effects of Calcitonin Gene-Related Peptide on BFGF and AQP4 Expression after Focal Cerebral Ischemia Reperfusion in Rats. Pharmazie 2010, 65, 274–278. [Google Scholar] [CrossRef]
  56. Tam, C.; Brain, S.D. The Assessment of Vasoactive Properties of CGRP and Adrenomedullin in the Microvasculature: A Study Using in Vivo and in Vitro Assays in the Mouse. J. Mol. Neurosci. 2004, 221, 17–124. [Google Scholar] [CrossRef]
  57. Shin, H.K.; Hong, K.W. Importance of Calcitonin Gene-Related Peptide, Adenosine and Reactive Oxygen Species in Cerebral Autoregulation under Normal and Diseased Conditions. Clin. Exp. Pharmacol. Physiol. 2004, 31, 1–7. [Google Scholar] [CrossRef]
  58. Leticia, F.; Anita, I. Adrenomedullin and Angiotensin II Signaling Pathways Involved in the Effects on Cerebellar Antioxidant Enzymes Activity. Brain Res. Bull. 2017, 128, 83–91. [Google Scholar] [CrossRef]
  59. Rehni, A.K.; Singh, T.G.; Jaggi, A.S.; Singh, N. Pharmacological Preconditioning of the Brain: A Possible Interplay between Opioid and Calcitonin Gene Related Peptide Transduction Systems. Pharmacol. Rep. 2008, 60, 904. [Google Scholar]
  60. Cozene, B.; Sadanandan, N.; Farooq, J.; Kingsbury, C.; Park, Y.J.; Wang, Z.J.; Moscatello, A.; Saft, M.; Cho, J.; Gonzales-Portillo, B.; et al. Mesenchymal Stem Cell-Induced Anti-Neuroinflammation Against Traumatic Brain Injury. Cell Transpl. 2021, 30, 09636897211035715. [Google Scholar]
  61. Zhu, J.; Chen, Y.H.; Ji, J.J.; Lu, C.X.; Liu, Z.F. Calcitonin Gene-Related Peptide Inhibits Neuronal Apoptosis in Heatstroke Rats via PKA/p-CREB Pathway. Chin. J. Traumatol. 2024, 27, 18–26. [Google Scholar] [CrossRef] [PubMed]
  62. Xiong, J.; Wang, Z.; Bai, J.; Cheng, K.; Liu, Q.; Ni, J. Calcitonin Gene-Related Peptide: A Potential Protective Agent in Cerebral Ischemia–Reperfusion Injury. Front. Neurosci. 2023, 17, 1184766. [Google Scholar] [CrossRef] [PubMed]
  63. Yang, S.; Yuan, Y.; Jiao, S.; Luo, Q.; Yu, J. Calcitonin Gene-Related Peptide Protects Rats from Cerebral Ischemia/Reperfusion Injury via a Mechanism of Action in the MAPK Pathway. Biomed. Rep. 2016, 4, 699–703. [Google Scholar] [CrossRef] [PubMed]
  64. Kelly, P.D.; Yengo-Kahn, A.M.; Tang, A.R.; Jonathan, S.V.; Reynolds, R.A.; Ye, F.; Zhao, Z.; Froehler, M.T.; Fusco, M.R.; Morone, P.J.; et al. Conditional Vasospasm-Free Survival Following Aneurysmal Subarachnoid Hemorrhage. Neurocrit. Care 2022, 37, 81–90. [Google Scholar] [CrossRef]
  65. Altamura, C.; Viticchi, G.; Fallacara, A.; Costa, C.M.; Brunelli, N.; Fiori, C.; Silvestrini, M.; Vernieri, F. Erenumab Does Not Alter Cerebral Hemodynamics and Endothelial Function in Migraine without Aura. Cephalalgia 2021, 41, 90–98. [Google Scholar] [CrossRef]
  66. Lampl, C.; Kraus, V.; Lehner, K.; Loop, B.; Chehrenama, M.; Maczynska, Z.; Ritter, S.; Klatt, J.; Snellman, J. Safety and Tolerability of Erenumab in Individuals with Episodic or Chronic Migraine across Age Groups: A Pooled Analysis of Placebo-Controlled Trials. J. Headache Pain 2022, 23, 104. [Google Scholar] [CrossRef]
  67. Kudrow, D.; Dafer, R.; Dodick, D.W.; Starling, A.; Ailani, J.; Dougherty, C.; Kalidas, K.; Zhang, F.; Jeswani, R.; Patel, N.; et al. Evaluation of Vascular Risk in Patients with Migraine with and without Aura Treated with Erenumab: Post Hoc Analysis of Pooled Long-Term Clinical Trial Data. Headache 2023, 63, 418–428. [Google Scholar] [CrossRef]
  68. Haghdoost, F.; Puledda, F.; Garcia-Azorin, D.; Huessler, E.-M.; Messina, R.; Pozo-Rosich, P. 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 2023, 43, 3331024231159366. [Google Scholar] [CrossRef]
  69. Muñoz-Vendrell, A.; Campoy, S.; Caronna, E.; Alpuente, A.; Torres-Ferrus, M.; Nieves Castellanos, C.; Olivier, M.; Campdelacreu, J.; Prat, J.; Camiña Muñiz, J.; et al. Effectiveness and Safety of Anti-CGRP Monoclonal Antibodies in Patients over 65 Years: A Real-Life Multicentre Analysis of 162 Patients. J. Headache Pain 2023, 24, 63. [Google Scholar] [CrossRef]
  70. Ashina, M.; Goadsby, P.J.; Reuter, U.; Silberstein, S.; Dodick, D.W.; Xue, F.; Zhang, F.; Paiva da Silva Lima, G.; Cheng, S.; Mikol, D.D. Long-Term Efficacy and Safety of Erenumab in Migraine Prevention: Results from a 5-Year, Open-Label Treatment Phase of a Randomized Clinical Trial. Eur. J. Neurol. 2021, 28, 1716–1725. [Google Scholar] [CrossRef]
  71. True, D.; Mullin, K.; Croop, R. Safety of Rimegepant in Adults with Migraine and Cardiovascular Risk Factors: Analysis of a Multicenter, Long-Term, Open-Label Study. Pain Ther. 2024, 13, 1203–1218. [Google Scholar] [CrossRef] [PubMed]
  72. Mulder, I.A.; Li, M.; de Vries, T.; Qin, T.; Yanagisawa, T.; Sugimoto, K.; van den Bogaerdt, A.; Danser, A.H.J.; Wermer, M.J.H.; van den Maagdenberg, A.M.J.M.; et al. Anti-Migraine Calcitonin Gene-Related Peptide Receptor Antagonists Worsen Cerebral Ischemic Outcome in Mice. Ann. Neurol. 2020, 88, 771–784. [Google Scholar] [CrossRef] [PubMed]
  73. Favoni, V.; Giani, L.; Al-Hassany, L.; Asioli, G.M.; Butera, C.; de Boer, I.; Guglielmetti, M.; Koniari, C.; Mavridis, T.; Vaikjärv, M.; et al. CGRP and Migraine from a Cardiovascular Point of View: What Do We Expect from Blocking CGRP? J. Headache Pain 2019, 20, 27. [Google Scholar] [CrossRef]
  74. Ohlsson, L.; Haanes, K.A.; Kronvall, E.; Xu, C.; Snellman, J.; Edvinsson, L. Erenumab (AMG 334), a Monoclonal Antagonist Antibody against the Canonical CGRP Receptor, Does Not Impair Vasodilatory or Contractile Responses to Other Vasoactive Agents in Human Isolated Cranial Arteries. Cephalalgia 2019, 39, 1745–1752. [Google Scholar] [CrossRef] [PubMed]
  75. Bahr-Hosseini, M.; Meißner, N.; Reidler, P.; Saver, J.L.; Tiedt, S. Plasma CGRP Levels Are Not Associated With Collateral Flow and Outcome After Stroke. Stroke 2023, 54, e203–e204. [Google Scholar] [CrossRef] [PubMed]
  76. Sohn, I.; Sheykhzade, M.; Edvinsson, L.; Sams, A. The Effects of CGRP in Vascular Tissue—Classical Vasodilation, Shadowed Effects and Systemic Dilemmas. Eur. J. Pharmacol. 2020, 881, 173205. [Google Scholar] [CrossRef]
  77. Smillie, S.-J.; King, R.; Kodji, X.; Outzen, E.; Pozsgai, G.; Fernandes, E.; Marshall, N.; de Winter, P.; Heads, R.J.; Dessapt-Baradez, C.; et al. An Ongoing Role of α-Calcitonin Gene-Related Peptide as Part of a Protective Network against Hypertension, Vascular Hypertrophy, and Oxidative Stress. Hypertension 2014, 63, 1056–1062. [Google Scholar] [CrossRef]
  78. Saely, S.; Croteau, D.; Jawidzik, L.; Brinker, A.; Kortepeter, C. Hypertension: A New Safety Risk for Patients Treated with Erenumab. Headache 2021, 61, 202–208. [Google Scholar] [CrossRef]
  79. de Vries Lentsch, S.; van der Arend, B.W.H.; Maassen VanDenBrink, A.; Terwindt, G.M. Blood Pressure in Patients with Migraine Treated with Monoclonal Anti-CGRP (Receptor) Antibodies: A Prospective Follow-up Study. Neurology 2022, 99, e1897–e1904. [Google Scholar] [CrossRef]
  80. Mair, J.; Lechleitner, P.; Längle, T.; Wiedermann, C.; Dienstl, F.; Saria, A. Plasma CGRP in Acute Myocardial Infarction. Lancet 1990, 335, 168. [Google Scholar] [CrossRef]
  81. Strecker, T.; Reeh, P.W.; Weyand, M.; Messlinger, K. Release of Calcitonin Gene-Related Peptide from the Isolated Mouse Heart: Methodological Validation of a New Model. Neuropeptides 2006, 40, 107–113. [Google Scholar] [CrossRef] [PubMed]
  82. Gennari, C.; Nami, R.; Agnusdei, D.; Fischer, J.A. Improved Cardiac Performance with Human Calcitonin Gene Related Peptide in Patients with Congestive Heart Failure. Cardiovasc. Res. 1990, 24, 239–241. [Google Scholar] [CrossRef] [PubMed]
  83. Li, D.; Peng, J.; Xin, H.-Y.; Luo, D.; Zhang, Y.-S.; Zhou, Z.; Jiang, D.-J.; Deng, H.-W.; Li, Y.-J. Calcitonin Gene-Related Peptide-Mediated Antihypertensive and Anti-Platelet Effects by Rutaecarpine in Spontaneously Hypertensive Rats. Peptides 2008, 29, 1781–1788. [Google Scholar] [CrossRef] [PubMed]
  84. de Boer, I.; MaassenVanDenBrink, A.; Terwindt, G.M. The Potential Danger of Blocking CGRP for Treating Migraine in CADASIL Patients. Cephalalgia 2020, 40, 1676–1678. [Google Scholar] [CrossRef]
  85. Kraenzlin, M.E.; Ch’ng, J.L.; Mulderry, P.K.; Ghatei, M.A.; Bloom, S.R. Infusion of a Novel Peptide, Calcitonin Gene-Related Peptide (CGRP) in Man. Pharmacokinetics and Effects on Gastric Acid Secretion and on Gastrointestinal Hormones. Regul. Pept. 1985, 10, 189–197. [Google Scholar] [CrossRef]
  86. Jakate, A.; Blumenfeld, A.M.; Boinpally, R.; Butler, M.; Borbridge, L.; Contreras-De Lama, J.; McGeeney, D.; Periclou, A.; Lipton, R.B. Pharmacokinetics and Safety of Ubrogepant When Coadministered with Calcitonin Gene–related Peptide-Targeted Monoclonal Antibody Migraine Preventives in Participants with Migraine: A Randomized Phase 1b Drug–Drug Interaction Study. Headache 2021, 61, 642–652. [Google Scholar] [CrossRef]
  87. Alsaadi, T.; Suliman, R.; Santos, V.; Al Qaisi, I.; Carmina, P.; Aldaher, B.; Haddad, S.; Bader, Y. Safety and Tolerability of Combining CGRP Monoclonal Antibodies with Gepants in Patients with Migraine: A Retrospective Study. Neurol. Ther. 2024, 13, 465–473. [Google Scholar] [CrossRef]
  88. de Vries, T.; Rubio-Beltrán, E.; van den Bogaerdt, A.; Dammers, R.; Danser, A.H.J.; Snellman, J.; Bussiere, J.; MaassenVanDenBrink, A. Pharmacology of Erenumab in Human Isolated Coronary and Meningeal Arteries: Additional Effect of Gepants on Top of a Maximum Effect of Erenumab. Br. J. Pharmacol. 2024, 181, 1720–1733. [Google Scholar] [CrossRef]
  89. Cirillo, C.; Brihmat, N.; Castel-Lacanal, E.; Le Friec, A.; Barbieux-Guillot, M.; Raposo, N.; Pariente, J.; Viguier, A.; Simonetta-Moreau, M.; Albucher, J.F.; et al. Post-Stroke Remodeling Processes in Animal Models and Humans. J. Cereb. Blood Flow Metab. 2020, 40, 3–22. [Google Scholar] [CrossRef]
  90. van der Vliet, R.; Selles, R.W.; Andrinopoulou, E.-R.; Nijland, R.; Ribbers, G.M.; Frens, M.A.; Meskers, C.; Kwakkel, G. Predicting Upper Limb Motor Impairment Recovery After Stroke: A Mixture Model. Ann. Neurol. 2020, 87, 383–393. [Google Scholar] [CrossRef]
Figure 1. Targets of CGRP-targeting therapies at the canonical CGRP-receptor. (1) Illustrates the activation of the CGRP receptor complex by its endogenous ligand CGRP. (2a) Shows a mAb-dependent blockage of the CGRP receptor. (2b) Demonstrates a mAb-dependent binding of the CGRP ligand. (3) Shows an inactivation of the CGRP receptor complex due to small molecules (gepants). CGRP (Calcitonin-gene related peptide), CLR (Calcitonin receptor-like receptor), GαS (Gs alpha subunit), RAMP1 (Receptor activity modifying protein 1), RCP (Receptor Component Protein).
Figure 1. Targets of CGRP-targeting therapies at the canonical CGRP-receptor. (1) Illustrates the activation of the CGRP receptor complex by its endogenous ligand CGRP. (2a) Shows a mAb-dependent blockage of the CGRP receptor. (2b) Demonstrates a mAb-dependent binding of the CGRP ligand. (3) Shows an inactivation of the CGRP receptor complex due to small molecules (gepants). CGRP (Calcitonin-gene related peptide), CLR (Calcitonin receptor-like receptor), GαS (Gs alpha subunit), RAMP1 (Receptor activity modifying protein 1), RCP (Receptor Component Protein).
Ijms 25 11685 g001
Figure 2. Risk factors for stroke in migraineurs. This figure illustrates the most important studied risk factors for stroke in migraineurs. mABs (monoclonal Antibodies).
Figure 2. Risk factors for stroke in migraineurs. This figure illustrates the most important studied risk factors for stroke in migraineurs. mABs (monoclonal Antibodies).
Ijms 25 11685 g002
Table 1. Roles of CGRP in stroke. Bcl-2 (B-cell lymphoma 2), BBB (blood-brain barrier), CBF (cerebral blood flow), CREB (cyclic AMP response element-binding protein), CGRP (calcitonin gene-related peptide), MAPK (mitogen-activated protein kinase), ERK (extracellular signal-regulated kinase), JNK (c-Jun N-terminal kinase), p38 (p38 mitogen-activated protein kinase).
Table 1. Roles of CGRP in stroke. Bcl-2 (B-cell lymphoma 2), BBB (blood-brain barrier), CBF (cerebral blood flow), CREB (cyclic AMP response element-binding protein), CGRP (calcitonin gene-related peptide), MAPK (mitogen-activated protein kinase), ERK (extracellular signal-regulated kinase), JNK (c-Jun N-terminal kinase), p38 (p38 mitogen-activated protein kinase).
Role of CGRP in StrokeMolecular MechanismEffect in StrokeComments
Blood-Brain Barrier (BBB) Influence [10,54,55]CGRP influences BBB permeabilityIncreases permeability, which can lead to cerebral edema; conflicting findings on protective effects against BBB injuryIncreased permeability may be linked to both beneficial or detrimental outcomes depending on conditions
Vasodilation [56,57]CGRP and adrenomedullin act as vasodilators, especially in microvasculatureMaintains cerebral blood flow (CBF) during ischemic conditions; counteracts hypoperfusionHelps in regulating blood flow, particularly during systemic hypotension
Antioxidative Effects [58,59]CGRP reduces oxidative stress during reperfusion injuryProtects against oxidative stress induced by reintroduction of blood flow post-ischemiaBeneficial in mitigating damage from reperfusion injury
Anti-Inflammatory Effects [9,60,63,64]CGRP (and adrenomedullin) show anti-inflammatory propertiesReduced inflammationSupports neuronal recovery by attenuating inflammatory response
Neuroprotection via CREB Pathway [8,61,62]Activates CREB pathway leading to increased Bcl-2, reduction of caspase-3Anti-apoptotic effect, protecting neuronsStabilizes mitochondrial membrane via Bcl-2/BAX pathway
Modulation of MAPK Pathways [63]Increases ERK phosphorylation, reduces JNK and p38 phosphorylationImproves neuronal function, reduces apoptosisMAPK pathway modulation contributes significantly to neuroprotective role
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Eller, M.T.; Frank, F.; Kaltseis, K.; Karisik, A.; Knoflach, M.; Broessner, G. Novel Calcitonin Gene-Related Peptide (CGRP) Interfering Migraine Therapies and Stroke—A Review. Int. J. Mol. Sci. 2024, 25, 11685. https://doi.org/10.3390/ijms252111685

AMA Style

Eller MT, Frank F, Kaltseis K, Karisik A, Knoflach M, Broessner G. Novel Calcitonin Gene-Related Peptide (CGRP) Interfering Migraine Therapies and Stroke—A Review. International Journal of Molecular Sciences. 2024; 25(21):11685. https://doi.org/10.3390/ijms252111685

Chicago/Turabian Style

Eller, Michael Thomas, Florian Frank, Katharina Kaltseis, Anel Karisik, Michael Knoflach, and Gregor Broessner. 2024. "Novel Calcitonin Gene-Related Peptide (CGRP) Interfering Migraine Therapies and Stroke—A Review" International Journal of Molecular Sciences 25, no. 21: 11685. https://doi.org/10.3390/ijms252111685

APA Style

Eller, M. T., Frank, F., Kaltseis, K., Karisik, A., Knoflach, M., & Broessner, G. (2024). Novel Calcitonin Gene-Related Peptide (CGRP) Interfering Migraine Therapies and Stroke—A Review. International Journal of Molecular Sciences, 25(21), 11685. https://doi.org/10.3390/ijms252111685

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop