Skip to main content

Comparison of gepant effects at therapeutic plasma concentrations: connecting pharmacodynamics and pharmacokinetics

Abstract

Background

Orally administered second-generation gepants are effective for the treatment of migraine. The intranasal administration of the third-generation gepant zavegepant might have additional benefits including a rapid onset of action, but it is not clear yet to which extent this has clinical relevance.

Methods

We examined the effect of zavegepant on the relaxations induced by calcitonin gene-related peptide (CGRP) in human isolated middle meningeal arteries. Furthermore, we connected the pharmacodynamics and pharmacokinetics of gepants by combining data from clinical and basic research.

Results

We showed that 10 nM zavegepant potently antagonized the functional response to CGRP. We also showed that all gepants are effective at inhibiting functional responses to CGRP at their therapeutic plasma concentrations.

Conclusions

The relatively low predicted potency of zavegepant to inhibit CGRP-induced relaxation at therapeutic systemic plasma concentrations may point to the relevance of local delivery to the trigeminovascular system through intranasal administration. This approach may have additional benefits for various groups of patients, including overweight patients.

Peer Review reports

Background

Calcitonin gene-related peptide (CGRP) plays a crucial role in the pathophysiology of migraine. During a migraine attack, the levels of the potent vasodilatory neuropeptide CGRP are elevated, and infusion of CGRP may induce migraine-like attacks in patients with migraine [25, 26, 30, 37]. Upon stimulation of the trigeminal ganglion, CGRP is released from trigeminal neurons, thereby activating nociceptive pain pathways and inducing intracranial vasodilation [25].

This knowledge on the role of CGRP in the pathogenesis of migraine has led to the development of drugs directed against CGRP and its receptor, i.e., CGRP(-receptor) targeting monoclonal antibodies and small-molecule CGRP receptor antagonists called gepants. In the early twenty-first century, several small-molecule CGRP receptor antagonists were developed, the majority of which had moderate affinity for the CGRP receptor [42]. These discoveries paved the way for the first generation of gepants that were advanced into clinical trials, namely olcegepant and telcagepant. Although the first-generation gepants were effective for the acute treatment of migraine attacks, their development was ceased. Telcagepant induced liver toxicity [33], whereas the low bioavailability of olcegepant and, therefore, its intravenous administration were considered a clinical limitation [45, 49]. The second-generation gepants ubrogepant, rimegepant, and atogepant are administered orally and were developed for either the acute treatment of attacks (ubrogepant), preventive treatment (atogepant), or both (rimegepant). These gepants are effective and do not exhibit any liver toxicity [50]. All three second-generation gepants have received approval from the FDA and EMA.

The third-generation gepant zavegepant (formerly named vazegepant) was developed for the acute treatment of migraine as an intranasal formulation. Intranasal administration can induce rapid effects as a drug can move across a single layer of epithelial cells into the bloodstream, leading to several benefits over oral formulations including a shorter time to reach the maximum plasma concentration and, thus, a faster onset of treatment effects and rapid pain relief [16, 46]. Additionally, intranasal delivery could potentially provide advantages for patients with severe nausea or vomiting [38]. Finally, it cannot be excluded that intranasal delivery would exert a direct trigeminovascular effect [23, 35, 41], although we are not aware of any data that could substantiate or refute this hypothesis. Zavegepant is currently in late-stage development and appears both efficacious and safe, also in terms of hepatotoxicity [16, 38]. Whether an intranasal formulation is more efficacious than oral administration and whether all gepants are equally efficacious and safe remain to be determined, as head-to-head trials for gepants have not been performed. Moreover, information on which group of patients would benefit from which gepant or administration method is highly relevant as gepants enter the market.

Our group has previously studied and compared the vascular effects of the then available gepants in coronary and intracranial arteries in a preclinical setting [18, 21, 27, 28, 43, 47]. In the current study, we aimed to characterize the functional response to CGRP in the absence or presence of the newest gepant zavegepant in human middle meningeal arteries (HMMA). With the middle meningeal artery being innervated by trigeminal nerve endings, this vascular bed is a relevant proxy for functional responses of the trigeminovascular system. Our second aim was to compare the blocking effect of gepants at therapeutic concentrations on CGRP-induced relaxation calculated from clinical gepant plasma levels. If the systemic concentrations of gepants are related to efficacy, which we assume to be similar across all gepants [19, 29, 40], we would expect a similar blocking effect towards CGRP for all gepants. The method utilized here enables the connection of in vitro pharmacodynamics to clinical pharmacokinetics.

Methods

Characterisation of zavegepant in human isolated middle meningeal arteries

HMMAs were obtained from 6 patients (2 females, 4 males; 52 ± 19 years of age) undergoing neurosurgery at Erasmus MC University Medical Center, Rotterdam, the Netherlands. All patients that were planned for pterional or temporal approaches indicated for tumour surgery, aneurysm surgery, or trauma surgery were eligible for HMMA harvesting. HMMAs were harvested by excision from the dura only if they did not experience any trauma by the craniotomy. Immediately upon dissection, the HMMAs were stored at 4 °C in Medium 199 (Westburg, the Netherlands) and rapidly transported to the laboratory. The surrounding dura and connective tissue were gently removed and the HMMA was stored overnight at 4 °C in oxygenated (95% O2, 5% CO2) high-glucose Krebs buffer (119 mM NaCl, 4.7 mM KCl, 1.25 mM CaCl2, 1.2 mM MgSO4, 1.2 mM KH2PO4, 25 mM NaHCO3, and 11.1 mM glucose; pH = 7.4).

For functional measurements the arteries were cut into 2-mm segments and mounted using Ø 40 μm stainless-steel wires in Mulvany myograph organ baths (Danish Myo Technology, Aarhus, Denmark) filled with high-glucose oxygenated Krebs buffer at 37 °C. Tension data were recorded using LabChart data acquisition (AD Instruments, Oxford, UK). After 30 min of equilibration, the tension of the arteries was normalized to 90% of the estimated diameter at 100 mmHg transmural pressure [44]. To assess viability and to obtain an internal standard for the contractile capacity per segment, the segments were contracted with 30 mM KCl, washed, and subsequently contracted with 100 mM KCl. Next, the segments were incubated with or without 10 nM zavegepant (zavegepant hydrochloride HY-132131, Bio-Connect, the Netherlands) dissolved in dimethyl sulfoxide and diluted in milliQ for 30 min prior to constructing a concentration–response curve for human αCGRP (Polypeptide Group, Baar, Switzerland; 0.01 nM – 1 µM, half logarithmic steps). After thorough washing, endothelial function was assessed with substance P (MCE, HY-P0201; 10 – 100 nM) after precontraction with U46619 (Sigma-Aldrich, D8174; 10 – 100 nM). CGRP-induced relaxation was expressed as a percentage of the precontraction to 30 mM KCl. Using nonlinear regression in Prism 8 (GraphPad Software, San Diego, CA, USA), pEC50 values and the pKB were calculated for each experiment to assess potency. In case that the concentration–response curve with zavegepant did not reach a plateau, the pEC50 was interpolated from a nonlinear regression that was restricted to the Emax obtained with the corresponding control segment. A paired t-test was performed for pEC50 values and KCl responses.

Analysis of gepant potency

Data collection

We collected data from the literature for the following gepants: rimegepant, ubrogepant, atogepant, and zavegepant, all of which were previously or currently in clinical development. Data on clinical pharmacokinetics, plasma-protein binding, and the (proposed) clinical dose were collected. Pharmacodynamic data on the potency of the aforementioned gepants, excluding zavegepant, were sourced from prior studies conducted within the same laboratory at the Erasmus MC University Medical Center in Rotterdam, the Netherlands using the same methodology [27, 28, 43, 47]. The original data on the pharmacological characterization of zavegepant are presented in the current paper.

Analysis of pharmacokinetic data

First, we transformed the data on therapeutic plasma concentrations of gepants and calculated total and plasma protein-corrected values. For a schematic overview of the analysis, see Fig. 1. Pharmacokinetic data (i.e., Cmax) from the highest approved dose were used, because these data were accessible for all gepants and this approach would thus enhance comparability. Cmax values were first converted to nanomolar concentrations to facilitate comparing pharmacokinetic data between the gepants. Next, Cmax values were corrected for plasma-protein binding to obtain the free fraction Cmax. For subsequent calculations, we used both the values of the total Cmax (bound and unbound) and the free fraction Cmax (corrected for plasma-protein binding), as a small deviation in the amount of plasma-protein binding affects the free fraction to a major extent, and we considered therefore the comparison between data more balanced when presenting both total and plasma protein-corrected values.

Fig. 1
figure 1

Schematic overview of calculations of the potency of the gepants at their therapeutic plasma concentrations

Connecting pharmacodynamics and pharmacokinetics

We then calculated the potency of the gepants at these plasma concentrations to block CGRP-induced relaxation (see Fig. 1). To this end, we combined the abovementioned pharmacokinetic data with pharmacodynamic data on the potency of gepants (i.e., pEC50, pKB, or pA2 values, whichever were available). We used potency data obtained with 10 nM of each gepant. The pharmacodynamic data were used to calculate the pEC50 values that CGRP would have in the presence of gepant levels corresponding to the Cmax. The pEC50 of CGRP in the absence of an antagonist served as a control. Subsequently, we interpolated relaxation that would occur in response to 100 nM CGRP in the presence of the Cmax. To achieve this, the shift to the right was calculated by subtracting the calculated pEC50 from the control pEC50. The concentration of 100 nM CGRP was used in the analysis because this concentration induces almost the maximum relaxation in the absence of a gepant and was included in all pharmacological characterizations. To enhance the accuracy of the interpolation, our analysis focused solely on gepants with Cmax values and pharmacological characterizations within the nanomolar range, thereby excluding telcagepant and olcegepant. This approach reduced the need for assumptions, particularly considering that Schild plot data in HMMA were not available for all gepants.

Results

Functional responses of the human middle meningeal artery to CGRP in the absence or presence of zavegepant

We aimed to characterise the functional response to human αCGRP in the absence or presence of 10 nM zavegepant in HMMA. Prior to the construction of the concentration–response curve to CGRP, the HMMA segments were challenged with 30 mM KCl and 100 mM KCl, which did not differ between the groups (30 mM KCl: control 6.92 ± 5.11 mN, zavegepant 7.47 ± 3.44 mN; KCl 100 mM: control 5.49 ± 4.99 mN, zavegepant 5.82 ± 3.17 mN). All HMMA segments had functional endothelium (substance P-induced relaxation: control 72.3 ± 6.2% of precontraction obtained with U46619, zavegepant 62.8 ± 32.9%). CGRP induced a functional vasorelaxant response with an Emax of 75.1 ± 11.6%. Zavegepant at 10 nM significantly shifted the concentration–response curve to CGRP to the right (control pEC50: 8.40 ± 0.09; 10 nM zavegepant pEC50: 6.38 ± 0.07; p = 0.0001; mean ± SEM; Fig. 2). Zavegepant did not induce vasocontraction. The pKB for 10 nM zavegepant was 10.02 ± 0.07. The pA2 value could not be calculated because we used only one concentration of zavegepant.

Fig. 2
figure 2

The effect of 10 nM zavegepant on the αCGRP-induced relaxation of human middle meningeal arteries (n = 6). The results are expressed as the mean ± SEM

Comparison of the potency of gepants at therapeutic concentrations to block CGRP-induced relaxation

In the second part of our analysis, we calculated the potency of gepants at their therapeutic plasma concentrations to block relaxation induced by 100 nM CGRP. To this end, we performed calculations based on both clinical pharmacokinetics and in vitro pharmacodynamics (see Supplemental Table 1). Concerning the clinical pharmacokinetics, the median total Cmax ranged from 24 nM for zavegepant to 1,420 nM for rimegepant and the median free fraction Cmax ranged from a median of 2.4 nM (zavegepant) to 57 nM (rimegepant) (Table 1; Fig. 3A-B).

Table 1 Calculated pEC50 of CGRP and clinical pharmacokinetics used for calculations
Fig. 3
figure 3

Gepant plasma concentrations, pEC50 of CGRP, and interpolated CGRP-induced relaxation in the absence or presence of therapeutic gepant plasma concentrations, based on values of total Cmax (bound and unbound; A, C, E) and free fraction Cmax (corrected for plasma-protein binding; B, D, F). A-B) Maximum plasma concentration (Cmax) measured in clinical trials. C-D) pEC50 of CGRP in the absence or presence of gepants at their therapeutic plasma concentrations (Cmax). E–F) Relaxation induced by 100 nM CGRP in the presence of a therapeutic concentration of the gepants, as a percentage of the relaxation induced by 100 nM CGRP in the absence of a gepant. All data are presented as median with range

Next, we used the Cmax to calculate the potency of each gepant to block CGRP-induced relaxation at their therapeutic concentrations by means of the pEC50. While the median pEC50 of CGRP in the absence of a gepant was 8.4 (range 8.2–8.7), the median pEC50 of CGRP in the presence of a gepant ranged from 3.4 (atogepant) to 6.0 (zavegepant) for the total Cmax and from 5.2 (atogepant) to 7.0 (zavegepant) for the free fraction Cmax (Table 1; Fig. 3C-D), demonstrating a clear shift to the right for all gepants at therapeutic concentrations.

Then, we interpolated CGRP-induced relaxation when a gepant at a therapeutic concentration (i.e., Cmax) would be present. With the response to 100 nM CGRP in the absence of a gepant set to 100%, all gepants were able to block CGRP-induced vasorelaxation (Fig. 3E-F). Atogepant was most potent for both the calculations based on total Cmax and free fraction Cmax: 100 nM CGRP would induce 0% (total Cmax) or 4.5% (free fraction Cmax) relaxation in the presence of atogepant, followed by rimegepant (0.9% and 12.4%), ubrogepant (3.2% and 18.6%), and zavegepant (13.2% and 48.2%).

Demographics and clinical characteristics of the trial populations

Clinical pharmacokinetic data were mainly obtained from small samples of healthy volunteers, with the exception of rimegepant, which was measured both in healthy volunteers and lactating women (Table 2). The age of the participants, sex distribution, and body mass index were not reported for any of the studies.

Table 2 Demographics and clinical characteristics of the trial populations included in our analysis

Discussion

Our first objective was to characterize the response to CGRP in the absence or presence of zavegepant, the only intranasally administered gepant, in HMMAs. Our results demonstrated that 10 nM zavegepant potently antagonizes the functional response to CGRP. Our second aim was to compare the blocking effect of therapeutic gepant concentrations on CGRP-induced relaxation. Our comparison revealed that all approved gepants effectively block the response to CGRP at their respective therapeutic plasma concentrations, with atogepant exhibiting the most potent inhibitory effect and zavegepant demonstrating the least inhibitory effect.

Although no head-to-head trials have been performed, several meta-analyses of gepants have been published recently that corroborate the assumption of similar efficacy across gepants. For the preventive gepants atogepant and rimegepant, the primary endpoint of reduction in mean monthly migraine days compared with placebo varied between -0.8 [-1.56; -0.04] (OR [95% CI]) for rimegepant 75 mg and -1.35 [-1.85; -0.85] for atogepant 60 mg [29]. For the acutely acting gepants rimegepant, ubrogepant, and zavegepant, two-hour pain relief versus placebo was between 1.16 [1.06–1.24] (RR [95% CI]) for zavegepant 10 mg and 1.37 [1.27–1.46] for rimegepant 75 mg in a meta-analysis [19]. In a preliminary meta-analysis of rimegepant and zavegepant, the percentage of patients experiencing two-hour pain relief was very similar for rimegepant 75 mg and zavegepant 10 mg (58.0% [56.0–60.2] versus 59.5% [56.4–62.5]) [40]. No comparison was available with data on two-hour pain freedom of the acute gepants. Intranasal delivery facilitates rapid absorption through the nasal mucosa, which leads to a fast onset of action, and might also make it suitable for patients experiencing nausea and vomiting. However, based on the similar efficacy observed among gepants, it is still debated whether the intranasal delivery of zavegepant offers additional therapeutic advantages compared to the orally administered gepants [19, 29, 38, 40, 46]. Notably, comparing clinical efficacy does not take into account that zavegepant exhibits a lower systemic concentration compared to the other gepants. These lower concentrations are confirmed by a lower blocking effect of CGRP in our analyses. If systemic concentrations were linked to efficacy, the therapeutic efficacy of zavegepant should have been inferior to that of the orally administered gepants. Given the fact that the efficacy is similar across gepants, we propose that the intranasal formulation of zavegepant could offer several benefits when compared to orally administered gepants.

The similar efficacy of zavegepant may indicate that its intranasal administration has not only a systemic effect, but rather a local effect. Indeed, the findings from the current study corroborate this hypothesis. Trigeminal nerve endings innervate the HMMA, which renders it a good proxy for the trigeminovascular system [35]. The nasal cavity is also largely lined with trigeminal nerve endings [35]. In HMMA, we show here that zavegepant has a relatively low potency to block CGRP-induced relaxation at its therapeutic plasma concentration (i.e., Cmax). Therefore, these findings may indicate the relevance of local delivery directly to the trigeminovascular system through intranasal administration (Fig. 4).

Fig. 4
figure 4

Local delivery of zavegepant directly to the trigeminovascular system through intranasal administration may improve pharmacokinetics, in addition to systemic absorption via the nasal mucosa. The nasal cavity is largely lined with trigeminal nerve endings. Created with BioRender.com

The trigeminal nerve innervates both the HMMA and nasal cavity, but not via the same afferents. The HMMA is mainly, but not exclusively, innervated by the ophthalmic branch (V1), whereas the trigeminal nerve endings lining the nasal cavity are from the maxillary branch (V2) [20, 35]. Previous research has shown that activation of different branches of the trigeminal nerve is relevant in migraine, which may demonstrate general trigeminal activation during a migraine attack. CGRP measured in saliva (mandibular branch/V3) is related to the different migraine phases [3] and has shown promise as a predictor for therapeutic responses to rizatriptan [15] and erenumab [2]. In the forehead model (V1) CGRP-mediated trigeminovascular reactivity is related to the treatment response of erenumab [17]. During a migraine attack, accompanying symptoms such as nasal congestion and rhinorrhoea are present in some migraine patients [5], which may hint at activation of V2. In addition, V1 is not the only branch supplying the dura mater; collaterals from V2 and V3 also provide dural innervation to small regions [20]. Future research should investigate the exact role of each part of the trigeminal nerve during a migraine attack.

If zavegepant is indeed directly delivered to the trigeminovascular system, this might have additional advantages. As it is expected that gepants distribute to adipose tissues given their lipophilicity, their plasma concentrations would be lower in overweight patients with migraine, possibly leading to reduced efficacy [14]. However, if zavegepant is indeed delivered directly to the trigeminal nerve endings, this could be beneficial for this group of patients [1]. One of the current questions concerning gepants, is which gepant will benefit which group of patients. Although evidence on the effect of sex, body mass index, or genetic background on the efficacy or side effects of gepants is lacking, it is known that results from the trials cannot be generalized to all patient populations [1]. Whether local delivery directly to the trigeminovascular system may circumvent pharmacokinetic differences between these patient populations has yet to be revealed.

In theory, intranasal delivery of zavegepant could not only be beneficial, but affect the nasal mucosa. CGRP induces vasodilation of the nasal mucosa, but the exact role of this functional response in sickness and health is not known [7, 39]. The zavegepant clinical trials have shown side effects including taste disorder, nasal discomfort, and throat irritation, but no nasal congestion or mucosal disorder have been reported thus far [36]. Real-world studies are needed to assess whether intranasal delivery of zavegepant induces long-term nasal side effects.

Despite ongoing debate on whether the main effect of gepants is peripheral or central, our current analysis underscores the importance of their peripheral action. Although some research suggests that intranasal delivery allows medications to bypass the blood–brain barrier via the olfactory nerve [23, 31, 34, 41], this has not been demonstrated for the gepants. Localization of gepants has been observed at the trigeminovascular system and dura, which fall outside the protection of the blood–brain barrier [22]. A minor extent of blood–brain barrier crossing (1.4%) has been revealed for oral telcagepant in rhesus monkeys [48]. Although evidence is lacking, it is likely that these results are generalizable for all gepants. As estimated from our current analysis, free fraction concentrations of gepants in the central nervous system are likely in the picomolar range, which is insufficient to potently block CGRP-induced functional responses. Considering the abundant expression of CGRP in the central nervous system and the absence of reported central side effects with any gepant, this further suggests that the medication class primarily exerts its action at a peripheral site [13, 32].

This study has several limitations. First, even though our calculations are based on large amounts of data, all calculations can only approximate in vitro results. These calculations are rather a way to extract more value from available data without using large amounts of scarce material. Our comparison is meant to make a general comparison and to be hypothesis-generating. The clinical data being reported in many different statistical formats and being based on studies with a low number of participants also reduces the accuracy of the comparison between gepants. Second, data from the clinical studies on pharmacokinetics were mainly obtained from healthy volunteers participating in phase 1 trials, and the sex and body mass index of participants were not mentioned in every publication. Responses to CGRP might be different in patients with migraine due to a potentially altered balance and expression of CGRP and its receptors in the trigeminovascular system as compared with controls.

Conclusion

In conclusion, the present study applied a novel method to connect pharmacodynamics and pharmacokinetics of gepants by combining data from clinical and basic research. Using this hypothesis-generating approach, we show that all gepants are effective at inhibiting functional responses to CGRP at their therapeutic plasma levels. The relatively low predicted potency of zavegepant to inhibit CGRP-induced relaxation at its therapeutic systemic plasma concentration may point to the relevance of local delivery to the trigeminovascular system through intranasal delivery, which should be further investigated in future studies.

Availability of data and materials

The datasets used and/or analysed during the current study are available from the corresponding author on reasonable request.

Abbreviations

CGRP:

Calcitonin gene-related peptide

Cmax :

Maximum plasma concentration

HMMA:

Human middle meningeal artery

pA2 :

The negative logarithm of the molar concentration that produces a twofold shift to the right in the agonist dose–response curve

pEC50 :

Negative logarithm of the molar concentration of an agonist that would induce half of the maximum response

pKB :

The negative logarithm of the molar concentration that would occupy 50% of the receptors at equilibrium

References

  1. Al-Hassany L, Goadsby PJ, Danser AHJ, Maassenvandenbrink A (2022) Calcitonin gene-related peptide-targeting drugs for migraine: how pharmacology might inform treatment decisions. The Lancet Neurology 21(3):284–294. https://doi.org/10.1016/s1474-4422(21)00409-9

    Article  CAS  PubMed  Google Scholar 

  2. Alpuente A, Gallardo VJ, Asskour L, Caronna E, Torres-Ferrus M, Pozo-Rosich P (2022) Salivary CGRP and Erenumab Treatment Response: Towards Precision Medicine in Migraine. Ann Neurol 92(5):846–859

    Article  CAS  PubMed  Google Scholar 

  3. Alpuente A, Gallardo VJ, Asskour L, Caronna E, Torres-Ferrus M, Pozo-Rosich P (2022) Salivary CGRP can monitor the different migraine phases: CGRP (in)dependent attacks. Cephalalgia 42(3):186–196

    Article  PubMed  Google Scholar 

  4. Ankrom W, Bondiskey P, Li C-C, Palcza J, Liu W, Dockendorf MF, Matthews C, Panebianco D, Reynders T, Wagner JA, Jakate A, Mesens S, Kraft WK, Marcantonio EE (2020) Ubrogepant Is Not Associated With Clinically Meaningful Elevations of Alanine Aminotransferase in Healthy Adult Males. Clin Transl Sci 13(3):462–472. https://doi.org/10.1111/cts.12728

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Arslan HH, Tokgöz E, Yildizoğlu Ü, Durmaz A, Bek S, Gerek M (2014) Evaluation of the changes in the nasal cavity during the migraine attack. J Craniofac Surg 25(5):e446–449

    Article  PubMed  PubMed Central  Google Scholar 

  6. Baker TE, Croop R, Kamen L, Price P, Stock DA, Ivans A, Bhardwaj R, Anderson MS, Madonia J, Stringfellow J, Bertz R, Coric V, Hale TW (2022) Human Milk and Plasma Pharmacokinetics of Single-Dose Rimegepant 75 mg in Healthy Lactating Women. Breastfeed Med 17(3):277–282

    Article  PubMed  PubMed Central  Google Scholar 

  7. Baraniuk JN, Lundgren JD, Goff J, Mullol J, Castellino S, Merida M, Shelhamer JH, Kaliner MA (1990) Calcitonin gene-related peptide in human nasal mucosa. American Journal of Physiology-Lung Cellular and Molecular Physiology 258(2):L81–L88. https://doi.org/10.1152/ajplung.1990.258.2.L81

    Article  CAS  Google Scholar 

  8. Bertz R, Bhardwaj R, Morris BA, Ashbrenner E, Coric V, Croop R (2023) A placebo-controlled, randomized, single and multiple dose study to evaluate the safety, tolerability, and pharmacokinetics of rimegepant in healthy participants. Cephalalgia 43(6):03331024231179131. https://doi.org/10.1177/03331024231179131

    Article  Google Scholar 

  9. Bertz, R., Donohue, M. K., Madonia, J., Bhardwaj, R., Stringfellow, J., Anderson, M., Stock, D. A., Morris, B. A., Coric, V., & Croop, R. (2022). P-142. Safety, tolerability, and pharmacokinetics of single and multiple ascending doses of zavegepant nasal spray in healthy adults. American Headache Society 64th Annual Scientific Meeting June 9–12, 2022 Denver, Colorado. https://www.headachejournal.onlinelibrary.wiley.com, https://doi.org/10.1111/head.14317

  10. Biohaven Pharmaceuticals Inc. (2020). Nurtec ODT. Package insert. Biohaven Pharmaceuticals Inc. Retrieved 26–01 from https://biohaven-nurtec-consumer-assets.s3.amazonaws.com/nurtec-prescribing-information.pdf

  11. Boinpally R, Jakate A, Butler M, Borbridge L, Periclou A (2021) Single-Dose Pharmacokinetics and Safety of Atogepant in Adults With Hepatic Impairment: Results From an Open-Label, Phase 1 Trial. Clin Pharmacol Drug Dev 10(7):726–733. https://doi.org/10.1002/cpdd.916

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Boinpally R, Lu K (2022) Single-Dose Pharmacokinetics and Safety of Ubrogepant in Adults With Hepatic Impairment: Results From an Open-Label, Phase 1 Trial. Clin Pharmacol Drug Dev 11(7):857–864. https://doi.org/10.1002/cpdd.1066

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Borkum JM (2019) CGRP and Brain Functioning: Cautions for Migraine Treatment. Headache 59(8):1339–1357

    Article  PubMed  Google Scholar 

  14. Bruijn N, van Lohuizen R, Boron M, Fitzek M, Gabriele F, Giuliani G, Melgarejo L, Řehulka P, Sebastianelli G, Triller P, Vigneri S, Özcan B, van den Brink AM, the European Headache Federation School of Advanced, S. (2024) Influence of metabolic state and body composition on the action of pharmacological treatment of migraine. J Headache Pain 25(1):20. https://doi.org/10.1186/s10194-024-01724-3

    Article  PubMed  PubMed Central  Google Scholar 

  15. Cady RK, Vause CV, Ho TW, Bigal ME, Durham PL (2009) Elevated saliva calcitonin gene-related peptide levels during acute migraine predict therapeutic response to rizatriptan. Headache 49(9):1258–1266

    Article  PubMed  Google Scholar 

  16. Croop R, Madonia J, Stock DA, Thiry A, Forshaw M, Murphy A, Coric V, Lipton RB (2022) Zavegepant nasal spray for the acute treatment of migraine: A Phase 2/3 double-blind, randomized, placebo-controlled, dose-ranging trial. Headache: J Head Face Pain 62(9):1153–1163. https://doi.org/10.1111/head.14389

    Article  Google Scholar 

  17. de Vries Lentsch S, Al-Hassany L, Ferrari MD, Terwindt GM, MaassenVanDenBrink A (2022) CGRP-mediated trigeminovascular reactivity in migraine patients treated with erenumab. J Neurol Neurosurg Psychiatry 93(8):911–912

    Article  PubMed  Google Scholar 

  18. de Vries T, Boucherie DM, van den Bogaerdt A, Danser AHJ, MaassenVanDenBrink A (2023) Blocking the CGRP Receptor: Differences across Human Vascular Beds. Pharmaceuticals 16(8):1075. https://www.mdpi.com/1424-8247/16/8/1075

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Deng X, Zhou L, Liang C, Shang X, Hui X, Liu W, Liang S, Wang Y, Xu M, Guo K, Yang K, Li X (2024) Comparison of effectiveness and safety of lasmiditan and CGRP-antagonists for the acute treatment of migraine in adults: systematic review and network meta-analysis of randomised trials. J Headache Pain 25(1):16. https://doi.org/10.1186/s10194-024-01723-4

    Article  PubMed  PubMed Central  Google Scholar 

  20. Edvinsson JCA, Viganò A, Alekseeva A, Alieva E, Arruda R, De Luca C, D’Ettore N, Frattale I, Kurnukhina M, Macerola N, Malenkova E, Maiorova M, Novikova A, Řehulka P, Rapaccini V, Roshchina O, Vanderschueren G, Zvaune L, Andreou AP, School European Headache Federation, of Advanced, S. (2020) The fifth cranial nerve in headaches. J Headache Pain 21(1):65

    Article  CAS  Google Scholar 

  21. Edvinsson L, Chan KY, Eftekhari S, Nilsson E, de Vries R, Säveland H, Dirven CMF, Danser AHJ, MaassenVanDenBrink A (2010) Effect of the calcitonin gene-related peptide (CGRP) receptor antagonist telcagepant in human cranial arteries. Cephalalgia 30(10):1233–1240. https://doi.org/10.1177/0333102410362122

    Article  PubMed  Google Scholar 

  22. Eftekhari S, Salvatore CA, Johansson S, Chen T-B, Zeng Z, Edvinsson L (2015) Localization of CGRP, CGRP receptor, PACAP and glutamate in trigeminal ganglion. Relation to the blood–brain barrier. Brain Res 1600:93–109. https://doi.org/10.1016/j.brainres.2014.11.031

    Article  CAS  PubMed  Google Scholar 

  23. Esim O, Savaser A, Ozkan CK, Oztuna A, Goksel BA, Ozler M, Tas C, Ozkan Y (2020) Nose to brain delivery of eletriptan hydrobromide nanoparticles: Preparation, in vitro/in vivo evaluation and effect on trigeminal activation. J Drug Deliv Sci Technol 59:101919. https://doi.org/10.1016/j.jddst.2020.101919

    Article  CAS  Google Scholar 

  24. FDA. (2023). ZAVZPRET™ (zavegepant) nasal spray - HIGHLIGHTS OF PRESCRIBING INFORMATION. Retrieved 14–04 from https://www.accessdata.fda.gov/drugsatfda_docs/label/2023/216386s000lbl.pdf

  25. Goadsby PJ, Edvinsson L (1993) The trigeminovascular system and migraine: studies characterizing cerebrovascular and neuropeptide changes seen in humans and cats. Ann Neurol 33(1):48–56

    Article  CAS  PubMed  Google Scholar 

  26. Goadsby PJ, Edvinsson L, Ekman R (1990) Vasoactive peptide release in the extracerebral circulation of humans during migraine headache. Ann Neurol 28(2):183–187. https://doi.org/10.1002/ana.410280213

    Article  CAS  PubMed  Google Scholar 

  27. Gupta S, Mehrotra S, Avezaat CJJ, Villalón CM, Saxena PR, Maassenvandenbrink A (2006) Characterisation of CGRP receptors in the human isolated middle meningeal artery. Life Sci 79(3):265–271. https://doi.org/10.1016/j.lfs.2006.01.003

    Article  CAS  PubMed  Google Scholar 

  28. Gupta S, Mehrotra S, Villalón CM, Garrelds IM, de Vries R, van Kats JP, Sharma HS, Saxena PR, MaassenVanDenBrink A (2006b) Characterisation of CGRP receptors in human and porcine isolated coronary arteries: Evidence for CGRP receptor heterogeneity. Eur J Pharmacol 530(1):107–116. https://doi.org/10.1016/j.ejphar.2005.11.020

    Article  CAS  PubMed  Google Scholar 

  29. Haghdoost F, Puledda F, Garcia-Azorin D, Huessler EM, Messina R, Pozo-Rosich P (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(4):3331024231159366

    Article  PubMed  Google Scholar 

  30. Hansen JM, Hauge AW, Olesen J, Ashina M (2010) Calcitonin gene-related peptide triggers migraine-like attacks in patients with migraine with aura. Cephalalgia 30(10):1179–1186

    Article  PubMed  Google Scholar 

  31. Hanson LR, Frey WH, 2nd. (2008) Intranasal delivery bypasses the blood-brain barrier to target therapeutic agents to the central nervous system and treat neurodegenerative disease. BMC Neurosci 9(Suppl 3):S5

    Article  PubMed  PubMed Central  Google Scholar 

  32. Hendrikse ER, Bower RL, Hay DL, Walker CS (2019) Molecular studies of CGRP and the CGRP family of peptides in the central nervous system. Cephalalgia 39(3):403–419

    Article  PubMed  Google Scholar 

  33. Ho TW, Ho AP, Ge YJ, Assaid C, Gottwald R, MacGregor EA, Mannix LK, van Oosterhout WP, Koppenhaver J, Lines C, Ferrari MD, Michelson D (2016) Randomized controlled trial of the CGRP receptor antagonist telcagepant for prevention of headache in women with perimenstrual migraine. Cephalalgia 36(2):148–161

    Article  PubMed  Google Scholar 

  34. Illum L (2010) Is nose-to-brain transport of drugs in man a reality? J Pharm Pharmacol 56(1):3–17. https://doi.org/10.1211/0022357022539

    Article  CAS  Google Scholar 

  35. Joo W, Yoshioka F, Funaki T, Mizokami K, Rhoton AL Jr (2014) Microsurgical anatomy of the trigeminal nerve. Clin Anat 27(1):61–88. https://doi.org/10.1002/ca.22330

    Article  PubMed  Google Scholar 

  36. Khan ZZ, Ahmed U, Shahzad F, Ali M, Tousif K, Ahmed U, Muhammad Safwan Q, Naufil SI, Murtaza S, Saeed S, Basit J, Haider T, Shabbir H (2023) Safety and Efficacy of Zavegepant in Treating Migraine: A Systematic Review. Cureus 15(7):e41991

    PubMed  PubMed Central  Google Scholar 

  37. Lassen L, Haderslev P, Jacobsen V, Iversen H, Sperling B, Olesen J (2002) Cgrp May Play A Causative Role in Migraine. Cephalalgia 22(1):54–61. https://doi.org/10.1046/j.1468-2982.2002.00310.x

    Article  CAS  PubMed  Google Scholar 

  38. Lipton RB, Croop R, Stock DA, Madonia J, Forshaw M, Lovegren M, Mosher L, Coric V, Goadsby PJ (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(3):209–217. https://doi.org/10.1016/s1474-4422(22)00517-8

    Article  CAS  PubMed  Google Scholar 

  39. Malis D-D, Rist B, Nicoucar K, Beck-Sickinger AG, Morel DR, Lacroix J-S (2001) Modulatory effect of two novel CGRP receptor antagonists on nasal vasodilatatory responses to exogenous CGRP, capsaicin, bradykinin and histamine in anaesthetised pigs. Regul Pept 101(1):101–108. https://doi.org/10.1016/S0167-0115(01)00275-0

    Article  CAS  PubMed  Google Scholar 

  40. Matcheswalla S, Yadav KN, Jena R, Velugoti LSDR, Perkyi-Boateng PK, Budnik A, Mahuwala Z (2024) A Preliminary Systematic Review and Meta-analysis of Randomized Control Trials of Rimegepant and Zavegepant for Acute Pain Relief from Migraine. Neurol 102:6374. https://doi.org/10.1212/WNL.0000000000206425

    Article  Google Scholar 

  41. Merkus FWHM, van den Berg MP (2007) Can Nasal Drug Delivery Bypass the Blood-Brain Barrier? Drugs in R & D 8(3):133–144. https://doi.org/10.2165/00126839-200708030-00001

    Article  CAS  Google Scholar 

  42. Moore EL, Salvatore CA (2012) Targeting a family B GPCR/RAMP receptor complex: CGRP receptor antagonists and migraine. Br J Pharmacol 166(1):66–78

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Mulder IA, Li M, de Vries T, Qin T, Yanagisawa T, Sugimoto K, van den Bogaerdt A, Danser AHJ, Wermer MJH, van den Maagdenberg AMJM, MaassenVanDenBrink A, Ferrari MD, Ayata C (2020) Anti-migraine Calcitonin Gene-Related Peptide Receptor Antagonists Worsen Cerebral Ischemic Outcome in Mice. Ann Neurol 88(4):771–784. https://doi.org/10.1002/ana.25831

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Mulvany MJ, Halpern W (1977) Contractile properties of small arterial resistance vessels in spontaneously hypertensive and normotensive rats. Circ Res 41(1):19–26

    Article  CAS  PubMed  Google Scholar 

  45. Olesen J, Diener H-C, Husstedt IW, Goadsby PJ, Hall D, Meier U, Pollentier S, Lesko LM (2004) Calcitonin Gene-Related Peptide Receptor Antagonist BIBN 4096 BS for the Acute Treatment of Migraine. N Engl J Med 350(11):1104–1110. https://doi.org/10.1056/NEJMoa030505

    Article  CAS  PubMed  Google Scholar 

  46. Reuter U (2023) A nasal CGRP receptor antagonist for acute migraine therapy. Lancet Neurol 22(3):190–191. https://doi.org/10.1016/s1474-4422(23)00037-6

    Article  CAS  PubMed  Google Scholar 

  47. Rubio-Beltran E, Chan KY, Danser AJ, Maassenvandenbrink A, Edvinsson L (2020) Characterisation of the calcitonin gene-related peptide receptor antagonists ubrogepant and atogepant in human isolated coronary, cerebral and middle meningeal arteries. Cephalalgia 40(4):357–366. https://doi.org/10.1177/0333102419884943

    Article  PubMed  Google Scholar 

  48. Sur C, Hargreaves R, Bell I, Dancho M, Graham S, Hostetler E, &, et al (2009) CSF levels and binding pattern of novel CGRP receptor antagonists in rhesus monkey and human central nervous system: toward the development of a PET tracer. Cephalalgia 29:136–137

    Google Scholar 

  49. Walker CS, Hay DL (2013) CGRP in the trigeminovascular system: a role for CGRP, adrenomedullin and amylin receptors? Br J Pharmacol 170(7):1293–1307. https://doi.org/10.1111/bph.12129

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Woodhead JL, Siler SQ, Howell BA, Watkins PB, Conway C (2022) Comparing the Liver Safety Profiles of 4 Next-Generation CGRP Receptor Antagonists to the Hepatotoxic CGRP Inhibitor Telcagepant Using Quantitative Systems Toxicology Modeling. Toxicol Sci 188(1):108–116

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

Figure 4 was created using BioRender.com.

Author information

Authors and Affiliations

Authors

Contributions

DMB and AMvdB designed the study. Human tissues were provided by RD and AV. DMB performed experiments and analyses, designed the figures, and drafted the manuscript. DMB, RD, AV, AHJD, and AMvdB critically reviewed the manuscript. All authors have read and agreed to the published version of the manuscript.

Corresponding author

Correspondence to Antoinette MaassenVanDenBrink.

Ethics declarations

Competing interests

DMB, RD, AV, and AHJD declare no conflict of interest. AMvdB received personal fees (fees as advisor or speaker, consultancy, any other) from Allergan-Abbvie, Lilly, Lundbeck, Novartis and Teva. She received research support from Novartis, Satsuma and Tonix, as well as independent research support from the Dutch Research Council and the Netherlands Organisation for Health Research and Development.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary Information

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, 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 changes were made. 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/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Boucherie, D.M., Dammers, R., Vincent, A. et al. Comparison of gepant effects at therapeutic plasma concentrations: connecting pharmacodynamics and pharmacokinetics. J Headache Pain 25, 141 (2024). https://doi.org/10.1186/s10194-024-01846-8

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s10194-024-01846-8

Keywords