Int J Curr Pharm Res, Vol 17, Issue 5, 1-5Review Article

A REVIEW OF TRIPTAN FORMULATIONS FOR THE TREATMENT OF MIGRAINE

TEJAL MEDANKAR*, SHASHIKANT DHOLE, NILESH KULKARNI, AMISHA GHUGE

Department of Pharmaceutics, PES Modern College of Pharmacy (for ladies), Moshi, Pune 412105, Maharashtra, India
*Corresponding author: Tejal Medankar; *Email: tejalmedankar23@gmail.com

Received: 05 Jun 2025, Revised and Accepted: 25 Jul 2025


ABSTRACT

Migraine is a severe headache that induces throbbing and pounding pain all over the head and is accompanied by sensitivity towards light, sound and touch. Triptans is one of the major class used to treat migraine. These are mainly employed in treating acute migraine conditions. This review paper focuses on comprehensive analysis of triptan-based treatments for migraine, with an emphasis on creating new dosage forms that would improve bioavailability and therapeutic effectiveness. This paper includes pharmacokinetics, mechanism of action and ability to target trigeminocervical system of almotriptan, naratriptan, rizatriptan, sumatriptan, and zolmitriptan. Innovative methods that have potential to boost medication transport to brain with less systemic exposure and quick relief from migraine symptoms are explained. It is finally concluded that triptan class is still a viable treatment option for migraine, with new dosage forms providing consistent therapeutic effect.

Keywords: Migraine, Triptan, Dosage form, Bioavailability, Method of preparation, Headache


INTRODUCTION

Common primary headache disorders like migraine ranked sixth in terms of disability and was the top cause of neurological disability worldwide in 2015 [1]. Migraineurs endure excruciating headache pain that may last up to 72 h and are frequently accompanied by sensitivity towards light, sound, and touch [2]. There are two types of migraine treatment: Prophylactic (preventive) medication treatment and acute medication treatment for individual attacks [3]. The most prevalent neurological vascular headache condition, migraine headache, induces throbbing, pounding pain all over the head. It often involves inadequate arterial sensitivity in the brain, which sets off triggers that frequently cause abrupt changes in artery diameter this leads to the enlargement of arteries in the brain and scalp that causes severe headache pain [4]. Several medications available to target migraine have recently been developed. These include monoclonal antibodies that target CGRP receptor antagonists intended for absorptive use. These drugs mark a step forward in the management of migraine, providing target therapy based on established pathophysiological pathways [5].

The acute antimigraine medication triptan is a serotonergic agonist. With the release of sumatriptan 25 y ago, triptan drugs were initially authorized as absorptive medicines in the treatment of migraine headaches. Sumatriptan was first made available in Europe in 1991 the treatment of this cripping line, which affects millions of people worldwide, was completely transformed [6]. Six additional triptans have been introduced to the pharmacopeia since sumatriptan was prescribed in the US. The pharmacokinetics of triptans varies slightly, but they are all serotonergic agonists that act on 5-HT1b and 5-HT1d receptors. Although there is debate about a single triptan mechanism for preventing migraine, meningeal vasoconstriction, central trigeminocervical pathway regulation, and blocking the production of trigeminal vasoactive peptides seem to be involved [7].

The Gods of ancient Egypt, such as Horus said “My head, my head” and Thoth said, “The side of my side,” all experienced headaches. Incanations were also used to combat other migraine characteristics including nausea and anorexia. Hippocrates was the first to diagnose headaches as a real illness rather than a divine visitation, and he did so only in Ancient Greece when he distinguished between several kinds of headaches in his aphorisms. As a result, he was the first to describe several widely recognized characteristics of migraine, including the visual aura, the headache’s subsequent start, its generalization, and its alleviation by vomiting. Celcus (25 BC-50 AD) was the first person to describe migraine as a chronic, non-fatal illness with triggers and stress and stated that headache could be localized or widespread. Galen (AD 131-201) defined migraine as “a painful disorder affecting approximately one half of the head, either the right or left side, which extends along the length of longitudinal suture”. He was interested in the connections between the extracranial and intracranial because he believed that migraine was caused by toxic vapor that enter the brain from other parts of the body. For centuries the humoral theory identified four humors: blood, phlegm, black and yellow bile was prevalent. The yellow bile was linked to migraine vomiting [9].

Table 1: Types of treatment for migraine [10]

Acute treatment formulations Preventive treatment formulations
1) Triptans (eg: sumatriptan, rizatriptan) 1) Beta-blockers (eg: Propranolol)
2) Ergotamines (eg: dihydroergotamine) 2) Anticonvulsants (eg: valproate)
3) Nonsteroidal anti-inflammatory drugs (NSAIDs) (eg: ibuprofen) 3) Antidepressants (eg: amitriptyline)
4) Anti-nausea medications (eg: metoclopramide, ondansetron) 4) Calcium channel blockers (eg: verapamil)

Different dosage forms available for the treatment of migraine

Almotriptan

It is the first medication licensed by the FDA to treat adult migraine headaches with or without aura. It is especially helpful for teenagers who have experienced untreated migraines lasting four hours or more in the past.

Mechanism of action: Almotriptan selectively binds to the cranial artery 5-HT1B and 5-HT1D serotonin receptor. Researchers believe that the process of relieving migraine involves neurogenic inflammation reduction, which is linked to the activation of 5-HT receptor.

Pharmacokinetics: Almotriptan reduces cerebral blood flow, constricts blood vessels in the brain, and restricts the pain impulses reaching the brain. The oral bioavailability is 69.1% and the half-life is about 3 h. It has low water solubility and therefore, ultimately affects bioavailability. Almotriptan is predominantly metabolized by cytochrome CYP450 and is eliminated through urine. It has a short plasma half-life and better oral bioavailability in contrast to prior-generation triptans like sumatriptan [11].

Table 2: Literature survey of almotriptan formulation as novel dosage form

Drug (Triptan) Dosage form Excipients Method of preparation Outcome
Almotriptan Mucoadhesive microsphere Almotriptan malate, Span-80, n-octanol, calcium chloride, gellan gum, distilled water, isopropyl alcohol w/o crosslinking emulsification method The microsphere nasal formulation improved mucosal residence time and dosing frequency was reduced [12].
Mucoadhesive insitu nasal gel containing solid lipid nanoparticle Almotriptan malate, glyceryl behenate, glyceryl palmitostearate, stearic acid, tween-80, agar, saline 0.9%, formaldehyde, dichloromethane, phospholipon 90 H, polyvinylalcohol, Poloxamer 407, carbopol 974p, Lubrizol, sodium alginate, sodium carboxy methyl cellulose, mucin, methanol, hematoxylin, eosin stain, ethyl acetate, diethyl ether, glacial acetic acid, benzalkonium chloride, total protein, sterile water w/o/w double emulsion solvent evaporation method Small particles led to increased absorption and the brain was successfully targeted through in situ nasal gel [13].
Mucoadhesive buccal film Almotriptan, Proloc 15, eudragit RL 100, eudragit RS 100, propylene glycol, polyvinylpyrrolidone, polyethylene glycol 400, methanol, ethyl cellulose, acetone, isopropyl alcohol, dibutyl phthalate. Solvent casting method Drug release, permeability, and therapeutic efficacy were improved [14].
Mucoadhesive insitu nasal gel Almotriptan malate, PF127, dialysis membrane, PF68, carboxymethyl chitosan, benzalkonium chloride. Cold technique Increased nasal residence time and fast onset of action are achieved [15].

Naratriptan

Mechanism of action: It is a selective agonist that acts on serotonin (5-HT1B and 5-HT1D) receptors in the cranial arteries; it constricts blood vessels and minimizes sterile inflammation that is associated with a drop in migraine pain.

Pharmacokinetics: Naratriptan is well absorbed, the volume of distribution is about 170L metabolized by the liver via CYP and 50% of the total dose in unchanged form; 30% of the total dose in the form of the metabolite is eliminated through urine [16].

Rizatriptan

Mechanism of action: Rizatriptan exhibits high affinity for the 5 HT1A site and low affinity for the non-5HT site. It is demonstrated that isolated middle meningeal arteries have a greater degree of cerebral selectivity than coronary arteries in people [20].

Pharmacokinetics: Rizatriptan is well absorbed when administered through the oral route, but the absolute oral bioavailability is reduced to 45% due to first-pass metabolism. The mean volume of distribution for male participants is 140L, while for females it is about 110L. Monoamine oxidase-A (MAO-A) is the primary enzyme that mediates the oxidative deamination of rizatriptan, resulting in the formulation of non-pharmacologically active triazolomethyl-indole-3-acetic acid. The pharmacological activity of N-monodesmethyl-rizatriptan, a minor metabolite, is equivalent to the parent drug. 14 % of the unchanged form of rizatriptan is excreted through urine and 51% through first pass metabolism [21].

Table 3: Literature survey of naratriptan formulation as novel dosage form

Drug (Triptan) Dosage form Excipients Method of preparation Outcome
Naratriptan Fast disintegrating tablet Naratriptan hydrochloride, glycine, mannitol, gelatin, amylose, soluble starch, dextrin, distilled deionized water Lyophilization To treat episodes of migraine, quick pain relief is preferred. Fast disintegrating tablet with good mechanical strength provides a high rate and extent of absorption and onset of action [17].
Oral transmucosal delivery Naratriptan HCL, ethanol, transcutol P, oleic acid, methocel 60 HG, PEG400, dipropylene glycol, miglyol, PEG 200, propylene glycol, phosphate buffer saline tablet, acetonitrile, trifluoroacetic acid, triethanolamine, methanol, water The liquid dosage forms were prepared by dissolving a known amount of naratriptan base in the desired amount of solvent. Transcutol permeation increases the uptake of naratriptan through the buccal membrane [18].
Thermo reversible mucoadhesive in situ nasal gel Naratriptan hydrochloride, poloxamer 407, carbopol 934, cellophane membrane Cold technique The addition of carbopol helps to improve the penetration of the drug and act as a mucoadhesive agent, whereas the poloxamer as a thermo-reversible agent helps to deliver a drug in a sustained manner [19].

Table 4: Literature survey of rizatriptan formulation as novel dosage form

Drug (Triptan) Dosage form Excipients Method of preparation Outcome
Rizatriptan Mouth dissolving tablet Rizatriptan benzoate, indion 234, indion 414, carboxymethylcellulose calcium, aspartame, mannitol, magnesium stearate, crospovidone, avicel pH-102 Direct compression method A higher ratio of water absorption and released 99.60% of the drug in a 2 min 15 sec time frame [22].
Fast-dissolving buccal film Rizatriptan benzoate, maltodextrin, xanthan gum, gum karaya, cinnamon oil, mannitol, saccharin, starch, and citric acid. Emulsion evaporation technique The emulsion evaporation technique used to prepare disintegrating film was found to be more effective in treating migraine [23].
Chitosan nanoparticle Rizatriptan, chitosan, acetic acid, tripolyphosphate, mannitol. Ionic gelation method Chitosan nanoparticle resulted in an improved formulation that provides nasal drug delivery insights [24].
Orodispersible electrospun Rizatriptan benzoate, PVA, PVP (K60), PVP (K30), PVP (K90) Electrospinning and casting method Electrospun orodispersible film was found to improve bioavailability and effective delivery for treating migraine [25].
Mucoadhesive buccal film Rizatriptan benzoate, HPMC K4M, PVA, polyethylene oxide, glycerol, disodium hydrogen phosphate, sodium chloride, potassium chloride, potassium dihydrogen phosphate, magnesium chloride, sodium hydrogen carbonate, HCL, calcium chloride, phosphate buffer saline Solvent casting method The mucoadhesive buccal film was found to be effective delivery for the treatment of migraine due to uniform drug release [26].
Insitu nasal gel Rizatriptan, carbopol 934P, HPMC (various grades), PEG400 Cold technique In situ nasal gel formulation proved to improve bioavailability as compared to oral route [27].
Intranasal spray formulation Rizatriptan benzoate, rizatriptan base, trifluroacetic acid, acetonitrile, propylene glycol, PEG400, NF, edetate disodium, dehydrated alcohol, benzalkonium chloride, anhydrous citric acid, butylated hydroxyl anisole, methyl paraben, propyl paraben, HCL, NaOH This method employs the preparation of two phases: the water phase and the ethanol phase. Quick onset of action was achieved [28].

Table 5: Literature survey of sumatriptan formulation as novel dosage form

Drug (Triptan) Dosage form Excipients Method of preparation Outcome
Sumatriptan Chitosan-coated liposome containing sumatriptan Hydrogenated soya phosphatidyl-choline, acetonitrile, methanol, ethyl acetate, sodium phosphate, formic acid, sodium hydroxide, chitosan, and sumatriptan. Thin film hydration method The nanoliposome containing sumatriptan was found to be sale and improved bioavailability was noted [30].
Sumatriptan intrarectal mucoadhesive gel Sumatriptan, poloxamer 407, poloxamer 188, xyloglucan, sodium hydroxide, potassium dihydrogen phosphate, benzalkonium chloride. Thin film hydration method The nanoliposome containing sumatriptan was found to be sale and improved bioavailability was noted [31].
Transdermal sumatriptan microneedle system Sumatriptan succinate, polyvinylpyrrolidone, glycerine, polysorbate 80, nitrazine yellow Ionic complexation was used to formulate complex nasal inserts by electrostatic interaction. Microneedle administration method offers an affordable substitute with no unfavorable side effects [32].
Freeze-dried nasal inserts Sumatriptan succinate, chitosan, carrageenan, mannitol Solvent diffusion evaporation technique The inserts were found to be effective because uptake of water made them viscous thereby leading to electrostatic interaction and further release of the drug in a controlled manner [33].
Nanostructure lipid carrier loaded with sumatriptan Sumatriptan, acetone, stearic acid, Brij 35, Brij 72, triolein, cholesterol, deionized water, sodium hydroxide, ammonium acetate, glacial acetic acid, ethyl acetate, acetonitrile Freeze drying technology The drug delivery to the brain to treat migraine was found to be effective and provides fast relief [34].
Orodispersible tablet Sumatriptan succinate, gelatin, plasdone K90D, sorbitol, sucrose, potassium dihydrogen orthophosphate, sodium hydroxide, mannitol, disodium ethylene diamine tetra acetic acid, magnesium stearate, methanol, camphor, xanthan gum, glycine SR, sucralose, distilled water. AVP-825 breath-powered exhalation device The orodispersible tablet was found to be fast disintegration and dissolution and provide a fast onset of action [35].
Sustained release of Mucoadhesive buccal film Sumatriptan succinate, HPMC, PEG, xanthan gum, potassium persulphate, acrylamide, acetone Solvent casting method and emulsion solvent diffusion Mucoadhesive films are an excellent alternative for delivering medication in a sustained manner and avoid first pass metabolism [36].
Dry nasal powder of sumatriptan Sumatriptan, lactose Breath powder exhalation device It was demonstrated that formulation was found to have fast onset, absorption, and low systemic exposure [37].
Mucoadhesive buccal disc and sublingual film Sumatriptan succinate, metoclopramide hydrochloride, HPMC (E-15), ethanol, dichloromethane, potassium dihydrogen phosphate, Sodium chloride, potassium chloride, sodium sulfate, ammonium acetate, urea, lactic acid, liquid paraffin, span 80 and propylene glycol Emulsion solvent diffusion and solvent casting method Mucoadhesive film and disc were found to have improved the release of drug and enhanced mucoadhesive characteristics [38].
Sumatriptan succinate insitu nasal gel Sumatriptan succinate, polyvinyl pyrolide, poloxamer, carbomer, benzalkonium chloride Cold technique Nasal residence time was improved, led to prolonged release of drug [39].
Fast-dissolving oral dosage form as a tablet and oral film Sumatriptan succinate, hydroxyl propyl methyl cellulose (K100M), urad dal, polyvinyl alcohol, soluplus, propylene glycol, ethanol, mannitol, citric acid water The orally disintegrating tablet was prepared by wet granulation technique and the oral film was prepared by solvent casting method. The addition of mucilage in an orally disintegrating tablet and a combination of HPMC K100M and soluplus helps to improve disintegration, and the maximum amount of drug release within a short time [40].

Sumatriptan

Mechanism of action: Sumatriptan reduces neurogenic inflammation by vasoconstriction of large cerebral blood vessels, inhibits transmission through peripheral neuronal inhibition, and reduces pain from migraines and their accompanying symptoms by inhibiting the activity of second-order neurons in the trigeminal cervical complex.

Pharmacokinetics: sumatriptan has 14% of bioavailability and the volume of distribution is about 17L. The inactive indoleacetic analogue of sumatriptan is produced after metabolism and is mainly excreted through urine and feces [29].

Zolmitriptan

Mechanism of action: Through direct 5-HT1B mediated cerebral blood vessel dilation as well as 5-HT1D mediated reduction of CGRP release, triptans can improve vasoconstriction.

Pharmacokinetics: Zolmitriptan has fast absorption across nasal mucosa; the volume of distribution is about 7-8 l/kg, metabolized by the liver by cyp450 and eliminated through urine and feces [41].

Table 6: Literature survey of zolmitriptan formulation as novel dosage form

Drug (Triptan) Dosage form Excipients Method of preparation Outcome
Zolmitriptan Insitu mucoadhesive intranasal gel Zolmitriptan, ketorolac tromethamine, tamarind gum, pluronic F127, polyethylene glycol, potassium dihydrogen orthophosphate, acetonitrile, trimethylamine, orthophosphoric acid, sodium chloride. Mix polymer with a gelling agent followed by the addition of water and additives.

Improves bioavailability and nasal residence time

[42].

Zolmitriptan-loaded bilosome that are incorporated in insitu nasal gel. Zolmitriptan, brij35, brijO10, cholesterol, hydroxypropyl methylcellulose, poloxamer 407, sodium deoxycholate, span 20, span 40, span 60, span 80, tween 65, tween 80, dialysis tubing cellulose membrane, methylene blue, normal saline, acetonitrile, formic acid, torsemide, distilled de-ionized water. The thin film hydration method was used for the preparation of bilosomes and the mucoadhesive gel was prepared by cold method. A mucoadhesive gel containing bilosome with drug loaded in it was found to be effective for brain targeting and ultimately, the bioavailability was also enhanced [43].
Zolmitriptan intranasal spanlastics Zolmitriptan, span60, tween 80, ethanol Ethanol injection method Zolmitriptan was found to be effective in brain targeting [44].

 

CONCLUSION

It was concluded that the triptan class was found to be effective in treating migraine as its loading in novel dosage form led to improve bioavailability and therapeutic efficacy when administered through various routes. Novel approaches will help to enhance patient compliance and efficiency.

FUNDING

Nil

AUTHORS CONTRIBUTIONS

All authors have contributed equally

CONFLICT OF INTERESTS

Authors declare that we have no conflict of interest.

REFERENCES

  1. Ong JJ, De Felice M. Migraine treatment: current acute medications and their potential mechanisms of action. Neurotherapeutics. 2018 Apr;15(2):274-90. doi: 10.1007/s13311-017-0592-1, PMID 29235068, PMCID PMC5935632.

  2. Killoran PM, Capel V, D Aloisio V, Schofield A, Aczel T, Bolcskei K. Novel peptide calcitonin gene-related peptide antagonists for migraine therapy. J Pharm Pharmacol. 2023 Sep 23;75(12):1581-9. doi: 10.1093/jpp/rgad081, PMID 37742055, PMCID PMC10502456.

  3. Diener HC, May A. New migraine drugs: a critical appraisal of the reason why the majority of migraine patients do not receive an adequate medication. Cephalalgia. 2024 Mar;44(3):3331024241228605. doi: 10.1177/03331024241228605, PMID 38520255.

  4. Abdou EM, Kandil SM, Miniawy HM. Brain targeting efficiency of antimigrain drug-loaded mucoadhesive intranasal nanoemulsion. Int J Pharm. 2017;529(1-2):667-77. doi: 10.1016/j.ijpharm.2017.07.030, PMID 28729175.

  5. Bohm PE, Stancampiano FF, Rozen TD. Migraine headache: updates and future developments. Mayo Clin Proc. 2018 Nov;93(11):1648-53. doi: 10.1016/j.mayocp.2018.09.006, PMID 30392545.

  6. Humphrey PP. The discovery and development of the triptans a major therapeutic breakthrough. Headache. 2008 May;48(5):685-7. doi: 10.1111/j.1526-4610.2008.01097.x, PMID 18471110.

  7. Macone AE, Perloff MD. Triptans and migraine: advances in use, administration, formulation and development. Expert Opin Pharmacother. 2017 Apr;18(4):387-97. doi: 10.1080/14656566.2017.1288721, PMID 28129702.

  8. Cleveland Clinic. Migraine Headaches. Cleveland Clinic; 2023 Apr 18; Available from: https://my.clevelandclinic.org/health/diseases/5005-migraine-headaches#overview. [Last accessed on 03 May 2025].

  9. Rose FC. The history of migraine from mesopotamian to medieval times. Cephalalgia. 1995 Oct;15Suppl 15:1-3. doi: 10.1111/J.1468-2982.1995.TB00040.X, PMID 8749238.

  10. Rizzoli PB. Acute and preventive treatment of migraine. Continuum (Minneap Minn). 2012 Aug;18(4):764-82. doi: 10.1212/01.CON.0000418641.45522.3b, PMID 22868540.

  11. Sidhu G, Preuss C, Patel P. Almotriptan. In: Treasure Island, FL: StatPearls; 2024 Jan 26. Available from: https://www.ncbi.nlm.nih.gov/books/NBK541092/#article-17383.s3. [Last accessed on 03 May 2025].

  12. Abbas Z, Marihal S. Gellan gum-based mucoadhesive microspheres of almotriptan for nasal administration: formulation optimization using factorial design characterization and in vitro evaluation. J Pharm Bioallied Sci. 2014 Oct-Dec;6(4):267-77. doi: 10.4103/0975-7406.142959, PMID 25400410, PMCID PMC4231387.

  13. Youssef NA, Kassem AA, Farid RM, Ismail FA, El Massik MA, Boraie NA. A novel nasal almotriptan-loaded solid lipid nanoparticles in mucoadhesive in situ gel formulation for brain targeting: preparation, characterization and in vivo evaluation. Int J Pharm. 2018 Sep 1;548(1):609-24. doi: 10.1016/j.ijpharm.2018.07.014, PMID 30033394.

  14. Nair AB, Al Dhubiab BE, Shah J, Jacob S, Saraiya V, Attimarad M. Mucoadhesive buccal film of almotriptan improved therapeutic delivery in rabbit model. Saudi Pharm J. 2020 Feb;28(2):201-9. doi: 10.1016/j.jsps.2019.11.022, PMID 32042259, PMCID PMC6996216.

  15. Verekar RR, Gurav SS, Bolmal U. Thermosensitive mucoadhesive in situ gel for intranasal delivery of Almotriptan malate: formulation characterization and evaluation. J Drug Deliv Sci Technol. 2020 Aug;58:101778. doi: 10.1016/j.jddst.2020.101778.

  16. Medicine. Naratriptan Med Com. 2020 Feb 17. Available from: https://www.medicine.com/drug/naratriptan/hcp#pharmacology.

  17. Omar SM, Abd Alla FI, Abdelgawad NM. Preparation and optimization of fast-disintegrating tablet containing naratriptan hydrochloride using D-optimal mixture design. AAPS PharmSciTech. 2018 Aug;19(6):2472-87. doi: 10.1208/s12249-018-1061-9, PMID 29869307.

  18. Sattar M, Lane ME. Oral transmucosal delivery of naratriptan. Int J Pharm. 2016 Nov 30;514(1):263-9. doi: 10.1016/j.ijpharm.2016.06.039, PMID 27863671.

  19. Shelke S, Shahi S, Jalalpure S, Dhamecha D, Shengule S. Formulation and evaluation of thermoreversible mucoadhesive in-situ gel for intranasal delivery of naratriptan hydrochloride. J Drug Deliv Sci Technol. 2015 Oct;29:238-44. doi: 10.1016/j.jddst.2015.08.003.

  20. Kennewell PD. Antimigraine agents: triptans. In: Taylor JB, Triggle DJ, editors. Comprehensive medicinal chemistry II. Vol. 1. Amsterdam: Elsevier; 2007. p. 181-4.

  21. Drug Bank. Rizatriptan. Drug Bank Online; 2021. Available from: https://www.drugbank.com/drugs/db00953.

  22. Upadhyay J, Sonaji AV, Naaz F. Formulation & evaluation of anti Migrane mouth dissolving tablet. J Res Appl Sci Biotechnol. 2024 Apr;3(2):233-41. doi: 10.55544/jrasb.3.2.40.

  23. Vidyadhara S, Sasidhar RL, Balakrishna T, Vardhan MS. Formulation of rizatriptan benzoate fast dissolving buccal films by emulsion evaporation technique. Int J Pharm Investig. 2015 Apr-Jun;5(2):101-6. doi: 10.4103/2230-973X.153387, PMID 25838995, PMCID PMC4381386.

  24. Shirsat AE, Chitlange SS. Application of quality by design approach to optimize process and formulation parameters of rizatriptan-loaded chitosan nanoparticles. J Adv Pharm Technol Res. 2015 Jul-Sep;6(3):88-96. doi: 10.4103/2231-4040.157983, PMID 26317071, PMCID PMC4542404.

  25. Guo X, Cun D, Wan F, Bera H, Song Q, Tian X. Comparative assessment of in vitro/in vivo performances of orodispersible electrospun and casting films containing rizatriptan benzoate. Eur J Pharm Biopharm. 2020;154:283-9. doi: 10.1016/j.ejpb.2020.06.023.

  26. Salehi S, Boddohi S. New formulation and approach for mucoadhesive buccal film of rizatriptan benzoate. Prog Biomater. 2017 Dec;6(4):175-87. doi: 10.1007/s40204-017-0077-7, PMID 29110144, PMCID PMC5700911.

  27. Suhagiya K, Borkhataria CH, Gohil S, Manek RA, Patel KA, Patel NK. Development of mucoadhesive in situ nasal gel formulation for enhanced bioavailability and efficacy of rizatriptan in migraine treatment. Results in Chemistry. 2023;6:101010. doi: 10.1016/j.rechem.2023.101010.

  28. Chokshi A, Vaishya R, Inavolu R, Potta T. Intranasal spray formulation containing rizatriptan benzoate for the treatment of migraine. Int J Pharm. 2019 Nov 25;571:118702. doi: 10.1016/j.ijpharm.2019.118702, PMID 31593810.

  29. Barbanti P, Le Pera D, Cruccu G. Sumatriptan fast disintegrating/rapid release tablets in the acute treatment of migraine. Expert Rev Neurother. 2007 Aug;7(8):927-34. doi: 10.1586/14737175.7.8.927, PMID 17678486.

  30. Assadpour S, Akhtari J, Shiran MR. Pharmacokinetics study of chitosan coated liposomes containing sumatriptan in the treatment of migraine. Caspian J Intern Med. 2022;13(1):90-9. doi: 10.22088/cjim.13.1.90, PMID 35178213.

  31. Kassab HJ, Alkufi HK, Hussein LS. Use of factorial design in formulation and evaluation of intrarectal in situ gel of sumatriptan. J Adv Pharm Technol Res. 2023 Apr-Jun;14(2):119-24. doi: 10.4103/japtr.japtr_603_22, PMID 37255866, PMCID PMC10226710.

  32. Ronnander P, Simon L, Spilgies H, Koch A, Scherr S. Dissolving polyvinylpyrrolidone based microneedle systems for in vitro delivery of sumatriptan succinate. Eur J Pharm Sci. 2018 Mar 1;114:84-92. doi: 10.1016/j.ejps.2017.11.031, PMID 29203152.

  33. Alavi S, Mortazavi SA. Freeze-dried K-carrageenan/chitosan polyelectrolyte complex-based insert: a novel intranasal delivery system for sumatriptan succinate. Iran J Pharm Res. 2018;17(4):1172-81. doi: 10.22037/ijpr.2018.2316, PMID 30568677, PMCID PMC6269572.

  34. Masjedi M, Azadi A, Heidari R, Mohammadi Samani S. Nose-to-brain delivery of sumatriptan loaded nanostructured lipid carriers: preparation optimization, characterization and pharmacokinetic evaluation. J Pharm Pharmacol. 2020 Oct;72(10):1341-51. doi: 10.1111/jphp.13316, PMID 32579251, PMCID PMC7448981.

  35. Gugulothu D, Desai P, Pandharipande P, Patravale V. Freeze drying: exploring potential in development of orodispersible tablets of sumatriptan succinate. Drug Dev Ind Pharm. 2015;41(3):398-405. doi: 10.3109/03639045.2013.871551, PMID 24384027.

  36. Verma S, Tonk RK, Albratty M, Alhazmi HA, Najmi A, Kumar R. Design and evaluation of sustained release mucoadhesive film of sumatriptan succinate containing grafted co-polymer as the platform. Saudi Pharm J. 2022 Nov;30(11):1527-37. doi: 10.1016/j.jsps.2022.07.014, PMID 36465843.

  37. Tepper SJ, Johnstone MR. Breath-powered sumatriptan dry nasal powder: an intranasal medication delivery system for acute treatment of migraine. Med Devices (Auckl). 2018 May 3;11:147-56. doi: 10.2147/MDER.S130900, PMID 29760572, PMCID PMC5937501.

  38. Jelvehgari M, Valizadeh H, Ziapour S, Rahmani M, Montazam SH, Soltani S. Comparative study of different combinational mucoadhesive formulations of sumatriptan metoclopramide. Adv Pharm Bull. 2016 Mar 17;6(1):119-30. doi: 10.15171/apb.2016.018, PMID 27123427, PMCID PMC4845549.

  39. Sasidhar RL, Vidyadhara S, Nagaraju R, Sowjanya Lakshmi B, Sailaja Y. Preparation and evaluation of sumatriptan succinate in situ gels. Int J Pharm Pharm Res. 2023 Feb 28;26(3):97-107.

  40. Kulkarni NS, Yadav S, Patil S. Development of fast dissolving oral dosage form as tablet using binder as vigna mungo mucilage and oral film using solvent casting technique: comparative study. Asian J Pharm. 2023 Dec 5;17(4):754-62.

  41. Drug Bank. Zolmitriptan. Drug Bank Online. Available from: https://www.drugbank.com/drugs/DB00315.

  42. Kumar A, Garg T, Sarma GS, Rath G, Goyal AK. Optimization of combinational intranasal drug delivery system for the management of migraine by using statistical design. Eur J Pharm Sci. 2015 Apr 5;70:140-51. doi: 10.1016/j.ejps.2015.01.012, PMID 25676136.

  43. El Taweel MM, Aboul Einien MH, Kassem MA, Elkasabgy NA. Intranasal zolmitriptan loaded bilosomes with extended nasal mucociliary transit time for direct nose to brain delivery. Pharmaceutics. 2021 Nov 1;13(11):1828. doi: 10.3390/pharmaceutics13111828, PMID 34834242.

  44. Saleh A, Khalifa M, Shawky S, Bani Ali A, Eassa H. Zolmitriptan intranasal spanlastics for enhanced migraine treatment; formulation parameters optimized via quality by design approach. Sci Pharm. 2021 May 25;89(2):24. doi: 10.3390/scipharm89020024.