A Review on Thermosensitive Zolmitriptan Mucoadhesive NanobioGel: Intranasal Delivery for Abortive treatment of Migraine

Authors

  • Shweta Kannao
  • Jitendra Banweer

Keywords:

Thermosensitive, Mucoadhesive, Nanoparticulate, Nose-To-Brain Delivery

Abstract

In order to improve migraine treatment by circumventing the blood-brain barrier and minimizing side effects throughout the body, this study investigates a novel thermosensitive mucoadhesive nanoparticulate biogel for intranasal delivery of zolmitriptan. The limitations of oral zolmitriptan, such as its delayed onset and poor absorption, make it necessary to develop novel drug delivery techniques for migraine, a crippling neurological condition. By utilizing the olfactory mucosa to facilitate the quickest possible drug delivery to the brain, intranasal medicine administration offers a non-invasive method. Zomitriptan-loaded nanoparticles made of biocompatible polymers will be included into the created biogel to improve drug encapsulation, regulate drug release, and strengthen mucoadhesion. Because of its thermosensitivity, it can be administered as a liquid at room temperature and then gelled in situ at body temperature, which guarantees extended contact with the nasal mucosa and improved absorption. Formulation characteristics such drug release, mucoadhesion, encapsulation efficiency, and particle size will be optimized by a design of experiments method. Technetium-labeled formulations will monitor biodistribution, and in vivo trials will evaluate the effectiveness of treating migraines in comparison to oral zolmitriptan. Toxicological tests will evaluate biocompatibility in the nasal cavity and central nervous system. Both small and large molecules can pass through the blood-brain barrier when medications are administered intranasally through the olfactory and trigeminal nerves. PLGA, PEG, and chitosan-based carriers are examples of nanoparticles that provide controlled release and improved delivery specificity. Drug transport is further supported by lipid-based carriers like lipid nanoparticles and liposomes. If this biogel is developed successfully, it has the potential to transform the treatment of migraines by enhancing patient outcomes and lowering treatment costs.

Downloads

Download data is not yet available.

Metrics

Metrics Loading ...

References

Abdelbary, G. A., & Tadros, M. I. (2013). Brain targeting of olanzapine via intranasal delivery of core–shell difunctional block copolymer mixed nanomicellar carriers: In vitro characterization, ex vivo estimation of nasal toxicity and in vivo biodistribution studies. International Journal of Pharmaceutics, 452: 300.

Abram JA, Patel P. Zolmitriptan. 2023 Nov 12. In: StatPearls [Internet]. Treasure Island (FL): StatPearls Publishing; PMID: 32491581.

Agbetou M and Adoukonou T (2022) Lifestyle Modifications for Migraine Management. Front Neurol. 18(13):719467.

Agrawal M, Saraf S, Saraf S and Sophia G (2018) Antimisiaris, Mahavir Bhupal Chougule, Sunday A. Shoyele, Amit Alexander, Nose-to-brain drug delivery: An update on clinical challenges and progress towards approval of anti-Alzheimer drugs, Journal of Controlled Release, 281:139-177

Ahn, J., Ko, J., Lee, S., Yu, J., Kim, Y., & Jeon, N. L. (2018). Microfluidics in nanoparticle drug delivery; From synthesis to pre-clinical screening. Advanced Drug Delivery Reviews, 128: 29.

Akopian, A. N., Chen, C. C., Ding, Y., Cesare, P., & Wood, J. N. (2000). A new member of the acid-sensing ion channel family. Neuroreport, 11(10), 2217–2222. https://doi.org/10.1097/00001756-200007140-00031

Almawash, S., Osman, S. K., Mustafa, G., & Hamd, M. A. E. (2022). Current and Future Prospective of Injectable Hydrogels—Design Challenges and Limitations. Pharmaceuticals, 15(3):371.

Amina S. J., Guo B. (2020). A review on the synthesis and functionalization of gold nanoparticles as a drug delivery vehicle. Int. J. Nanomedicine 15, 9823–9857.

Anselmo AC and Mitragotri S (2016) Nanoparticles in the clinic. Bioeng Transl Med. 1:10–29.

Ashina, M., Terwindt, G. M., Al‐Karagholi, M. A., Boer, I. de, Lee, M. J., Hay, D. L., Schulte, L., Hadjikhani, N., Sinclair, A. J., Ashina, H., Schwedt, T. J., & Goadsby, P. J. (2021). Migraine: disease characterisation, biomarkers, and precision medicine. The Lancet, 397(10283): 1496.

Asthana, A. (2017). Technological Innovations in Muccoadhessive Gastroretentive Drug Delivery System (MGRDDS). Advanced Research in Gastroenterology & Hepatology, 5:(4).

Bai X., Sun Q., Cui H., Guerzoni L. P. B., Wuttke S., Kiessling F., et al. (2022). Controlled covalent self-assembly of a homopolymer for multiscale materials engineering. Adv. Mat. 34, 2109701.

Baratloo, A., Mirbaha, S., Kasmaei, H. D., Payandemehr, P., Elmaraezy, A., & Negida, A. (2017). Intravenous caffeine citrate vs. magnesium sulfate for reducing pain in patients with acute migraine headache; a prospective quasi-experimental study. The Korean journal of pain, 30(3):176.

Bateman DN, Kahn C, Davies DS (1980) The pharmacokinetics of metoclopramide in man with observations in the dog.Br J Clin Pharmacol 9: 371-377

Benatto, M. T., Florêncio, L. L., Bragatto, M. M., Dach, F., Fernández‐de‐las‐Peñas, C., & Bevilaqua‐Grossi, D. (2020). Neck-specific strengthening exercise compared with sham ultrasound when added to home-stretching exercise in patients with migraine: study protocol of a two-armed, parallel-groups randomized controlled trial. Chiropractic & Manual Therapies, 28(1).

Blau JN, Kell CA and Sperling JM (2004) Water-deprivation headache: a new headache with two variants. Headache J Head Face Pain. 44:79–83.

Blumenfeld, A., Durham, P. L., Feoktistov, A., Hay, D. L., Russo, A. F., & Turner, I. (2021). Hypervigilance, Allostatic Load, and Migraine Prevention: Antibodies to CGRP or Receptor. Neurology and Therapy, 10(2):469.

Boddupalli, B., Mohammed, ZulkarN. K., Nath, R., & Banji, D. (2010). Mucoadhesive drug delivery system: An overview. Journal of Advanced Pharmaceutical Technology and Research, 1(4): 381.

Borkum, J. M. (2017). The Migraine Attack as a Homeostatic, Neuroprotective Response to Brain Oxidative Stress: Preliminary Evidence for a Theory. Headache The Journal of Head and Face Pain, 58(1):118.

Borkum, J. M. (2018). Harnessing migraines for neural regeneration. Neural Regeneration Research, 13(4): 609.

Bourganis, V., Kammona, A. Alexopoulos and C. Kiparissides (2018). Recent advances in carrier mediated nose-to-brain delivery of pharmaceutics, Eur. J. Pharm. Biopharm. 128 (2018) 337–362

Boustta M., Colombo P. E., Lenglet S., Poujol S., Vert M. (2014). Versatile UCST-based thermoresponsive hydrogels for loco-regional sustained drug delivery. J. Control. Release 174, 1–6.

Boz, R. K., Aydın, D. Y., Kocak, S., Golba, B., Sanyal, R., & Sanyal, A. (2022). Redox-Responsive Hydrogels for Tunable and “On-Demand” Release of Biomacromolecules. Bioconjugate Chemistry, 33(5):839.

Brown H, Newman C, Noad R, Weatherby S. (2012) Behavioural management of migraine. Ann Indian Acad Neurol. 15(Suppl 1):S78-82.

Brown, T., Habibi, N., Wu, D., Lahann, J., & Mitragotri, S. (2020). Effect of Nanoparticle Composition, Size, Shape, and Stiffness on Penetration Across the Blood–Brain Barrier. ACS Biomaterials Science & Engineering, 6(9): 4916).

Buwalda, S., Vermonden, T., & Hennink, W. E. (2016). Hydrogels for Therapeutic Delivery: Current Developments and Future Directions. Biomacromolecules, 18(2): 316. American Chemical Society.

Calhoun AH and Ford S (2007) Behavioral sleep modification may revert transformed migraine to episodic migraine. Headache J Head Face Pain. 47:1178–83.

Cao L., Fan J., Huang J., Chen Y. (2019). A robust and stretchable cross-linked rubber network with recyclable and self-healable capabilities based on dynamic covalent bonds. J. Mat. Chem. A 7, 4922–4933. 10.1039/c8ta11587g

Carvalho, G. F., Schwarz, A., Szikszay, T. M., Adamczyk, W. M., Bevilaqua‐Grossi, D., & Luedtke, K. (2019). Physical therapy and migraine: musculoskeletal and balance dysfunctions and their relevance for clinical practice. Brazilian Journal of Physical Therapy, 24(4): 306.

Charlesworth BR, Dowson AJ, Purdy A, Becker WJ, Boes-Hansen S, Färkkilä M.CNS Drugs (2003) Speed of onset and efficacy of zolmitriptan nasal spray in the acute treatment of migraine: a randomised, double-blind, placebo-controlled, dose-ranging study versus zolmitriptan tablet. 17(9):653-67. doi: 10.2165/00023210-200317090-00005.

Choonara BF, Choonara YE, Kumar P, Bijukumar D, du Toit LC and Pillay V (2014) A review of advanced oral drug delivery technologies facilitating the protection and absorption of protein and peptide molecules. Biotechnol Adv, 32:1269-1282

Ciolacu, D., Nicu, R., & Ciolacu, F. (2020). Cellulose-Based Hydrogels as Sustained Drug-Delivery Systems. Materials, 13(22):5270.

Collado-González, M., Espinosa, G., & Goycoolea, F. M. (2019). Interaction Between Chitosan and Mucin: Fundamentals and Applications. Biomimetics, 4(2):32.

Colombo, P., Catellani, P. L., Peppas, N. A., Maggi, L., & Conte, U. (1992). Swelling characteristics of hydrophilic matrices for controlled release New dimensionless number to describe the swelling and release behavior. International Journal of Pharmaceutics, 88: 99.

Coșman, B.-P., Bucătariu, S.-M., Constantin, M., & Fundueanu, G. (2022). Temperature/pH-Sensitive Double Cross-Linked Hydrogels as Platform for Controlled Delivery of Metoclopramide. Gels, 8(12): 824.

Costa, C. P., Moreira, J.N., Sousa Lobo, J.M. and Silva, A.C. (2021). Intranasal delivery of nanostructured lipid carriers, solid lipid nanoparticles and nanoemulsions: a current overview of in vivo studies, Acta Pharm. Sin. B. 925–940

Costa, C., Moreira, J.N., Amaral, M.H., Sousa Lobo, J.M. and Silva, A.C. (2019). Nose-to-brain delivery of lipid-based nanosystems for epileptic seizures and anxiety crisis, J. Control. Release 295,187–200

Croop R, Goadsby PJ, Stock DA, Conway CM, Forshaw M, Stock EG, et al. (2019) Efficacy, safety, and tolerability of rimegepant orally disintegrating tablet for the acute treatment of migraine: a randomised, phase 3, double-blind, placebo-controlled trial. Lancet 31;394(10200):737–45.

Cubała, W. J., Landowski, J., & Wichowicz, H. (2007). Zolpidem abuse, dependence and withdrawal syndrome: sex as susceptibility factor for adverse effects. British Journal of Clinical Pharmacology, 65(3): 444.

Cunha, S.,Forbes, B., Sousa Lobo, J.M. and Silva, A.C. (2021) Improving drug delivery for

Alzheimer’s disease through nose-to-brain delivery using nanoemulsions, nanostructured lipid carriers (NLC) and in situ hydrogels, Int. J. Nanomed. 16(2021) 4373–4390

Daboul I, Taleb N, Rashed HM, Alkheshen MM, Abell TL (2000) Evaluation of autonomic and enteric measures can affect clinical care of patients with GI motility disorders. Gastroenterology. doi: 10.1016/s0016-5085(00)80126-2

Dafonte KK, Weber L, Chmielewski F, Böhmer AM, Lutz P, Hartmann G, Jaehde U and Coenen M (2023) Dose Recommendations for Common Drugs in Patients with Liver Cirrhosis: A Systematic Literature Review. Clin Drug Investig. 43(7):475-502.

Dale, O. (2010). Intranasal administration of opioids/fentanyl—Physiological and pharmacological aspects. European Journal of Pain Supplements, 4: 187.

Devillier, P., Grassin‐Delyle, S., Naline, E., Buenestado, A., Blouquit‐Laye, S., & Advenier, C. (2010). Administration intranasale des médicaments à visée systémique : une nouvelle voie pour les opioïdes. Therapies, 65(5): 475.

Dhuria, S. V., Hanson, L. R., & Frey, W. H. (2009). Intranasal delivery to the central nervous system: Mechanisms and experimental considerations. Journal of Pharmaceutical Sciences, 99(4): 1654.

Diamante, P. A. (2020). Music Medicine as a Component of Acute Migraine Attack Management in The Emergency Room: A Randomized Controlled Open-Label Trial. https://doi.org/10.30756/ahmj.2020.04.01

Ding, C., & Li, Z. (2017). A review of drug release mechanisms from nanocarrier systems. Materials Science and Engineering C, 76: 1440.

Dixon R, Gillotin C, Gibbens M, Posner J, Peck RW. (1997) The pharmacokinetics and effects on blood pressure of multiple doses of the novel antimigraine drug, zolmitriptan (311C90): in healthy volunteers. Br J Clin Pharmacol. 43:273–281.

Dodick D, Brandes J, Elkind A, et al. (2005) Speed of onset, efficacy and tolerability of zolmitriptan nasal spray in the acute treatment of migraine: a randomized, double-blind, placebo-controlled study. CNSDrugs, 19(2):125–36.

Dodick, D. W., & Silberstein, S. D. (2006). Central Sensitization Theory of Migraine: Clinical Implications. Headache The Journal of Head and Face Pain, 46.

Domínguez, A., Suárez-Merino, B., & Goñi‐de‐Cerio, F. (2014). Nanoparticles and Blood-Brain Barrier: The Key to Central Nervous System Diseases. Journal of Nanoscience and Nanotechnology, 14(1):766.

Donnelly, R. F., Shaikh, R., Singh, T. R. R., Garland, M., & Woolfson, A. (2011). Mucoadhesive drug delivery systems. Journal of Pharmacy And Bioallied Sciences, 3(1): 89.

Dowson A, Charlesworth BR, Green J, et al. (2005) Zolmitriptan Nasal SprayExhibits Good Long-Term Safety and Tolerability in Migraine: Resultsof the INDEX Trial. Headache. 45:17–24

Dowson AJ, Charlesworth B. (2002) Review of zolmitriptan and its clinical applications in migraine. Expert Opin Pharmacother. 3(7):993-1005. doi: 10.1517/14656566.3.7.993. PMID: 12083998.

Dreiss, C. A. (2020). Hydrogel design strategies for drug delivery. Current Opinion in Colloid & Interface Science, 48:1.

Dudhipala, N., & Narala, A. (2016). Recent Updates in the Formulation Strategies to Enhance the Bioavailability of Drugs Administered via Intranasal Route. Journal of Bioequivalence & Bioavailability, 8(5).

Edvinsson, L. (2017). Headache advances in 2017: a new horizon in migraine therapy. In The Lancet Neurology, 17(1): 5.

Fang J, Nakamura H, Maeda H. (2011) The EPR effect: Unique features of tumor blood vessels for drug delivery, factors involved and limitations and augmentation of the effect. Adv Drug Deliv Rev. 63:136–151.

Ferroni, P., Barbanti, P., Spila, A., Fratangeli, F., Aurilia, C., Fofi, L., Egeo, G., & Guadagni, F. (2018). Circulating Biomarkers in Migraine: New Opportunities for Precision Medicine. Current Medicinal Chemistry, 26(34):6191.

Formica ML, Real DA, Picchio ML, Catlin E, Donnelly RF and Paredes AJ (2022) On a highway to the brain: A review on nose-to-brain drug delivery using nanoparticles, Applied Materials Today, 29.

Freiberg, S., & Zhu, X. X. (2004). Polymer microspheres for controlled drug release. International Journal of Pharmaceutics, 282: 1.

Furtado, D., Björnmalm, M., Ayton, S., Bush, A. I., Kempe, K., & Caruso, F. (2018). Overcoming the Blood–Brain Barrier: The Role of Nanomaterials in Treating Neurological Diseases. Advanced Materials, 30(46).

Galmarini CM, Warren G, Kohli E, Zeman A, Mitin A, Vinogradov SV. (2008) Polymeric nanogels containing the triphosphate form of cytotoxic nucleoside analogues show antitumor activity against breast and colorectal cancer cell lines. Mol Cancer Ther. 7:3373–3380.

Gänger, S., & Schindowski, K. (2018). Tailoring Formulations for Intranasal Nose-to-Brain Delivery: A Review on Architecture, Physico-Chemical Characteristics and Mucociliary Clearance of the Nasal Olfactory Mucosa. Pharmaceutics, 10(3):116.

Gao, W., Zhang, Y., Zhang, Q., & Zhang, L. (2016). Nanoparticle-Hydrogel: A Hybrid Biomaterial System for Localized Drug Delivery. Annals of Biomedical Engineering, 44(6): 2049.

Gauthier, J., Ivers, H., & Carrier, S. (1996). Nonpharmacological approaches in the management of recurrent headache disorders and their comparison and combination with pharmacotherapy. Clinical Psychology Review, 16(6): 543.

Gazzaniga, A., Sangalli, M. E., Conte, U., Caramella, C., Colombo, P., & Manna, A. L. (1993). On the release mechanism from coated swellable minimatrices. International Journal of Pharmaceutics, 91:167.

Goadsby, P. J., Holland, P. R., Martins-Oliveira, M., Hoffmann, J., Schankin, C. J., & Akerman, S. (2017). Pathophysiology of Migraine: A Disorder of Sensory Processing. Physiological Reviews, 97(2): 553.

Goschorska, M., Gutowska, I., Baranowska‐Bosiacka, I., Barczak, K., & Chlubek, D. (2020). The Use of Antioxidants in the Treatment of Migraine. Antioxidants, 9(2):116.

Goyal, S., Agarwal, G., Agarwal, S., & Karar, P. (2017). Oral Sustained Release Tablets: An Overview with a special emphasis on Matrix Tablet. American Journal of Advanced Drug Delivery, 5(2). https://doi.org/10.21767/2321-547x.1000013

Hanson, L. R., & Frey, W. H. (2008). Intranasal delivery bypasses the blood-brain barrier to target therapeutic agents to the central nervous system and treat neurodegenerative disease. BMC Neuroscience, 9.

Hargreaves, R. (2007). New Migraine and Pain Research. Headache The Journal of Head and Face Pain, 47.

He, P. (1999). Chitosan microspheres prepared by spray drying. International Journal of Pharmaceutics, 187(1): 53.

Heckroth M, Luckett RT, Moser C, Parajuli D, Abell TL. (2021) Nausea and Vomiting in 2021: A Comprehensive Update. J Clin Gastroenterol, 55(4):279-299.

Henrich‐Noack, P., Nikitovic, D., Neagu, M., Docea, A. O., Engin, A. B., Gelperina, S., Штильман, М. И., Mitsias, P., Tzanakakis, G. N., Gozes, I., & Tsatsakis, A. (2019). The blood–brain barrier and beyond: Nano-based neuropharmacology and the role of extracellular matrix. Nanomedicine Nanotechnology Biology and Medicine, 17: 359.

Hoare, T., & Kohane, D. S. (2008). Hydrogels in drug delivery: Progress and challenges. Polymer, 49(8): 1993.

Hoare, T., Santamarı́a, J., Goya, G. F., Irusta, S., Lin, D., Lau, S., Padera, R. F., Langer, R., & Kohane, D. S. (2011). A Magnetically-Triggered Composite Membrane for On-Demand Drug Delivery. arXiv. Cornell University.

Hoffmann J, Baca SM, Akerman S. (2019) Neurovascular mechanisms of migraine and cluster headache. J Cereb Blood Flow Metab. 39(4):573-594. doi: 10.1177/0271678X17733655. Epub

Holroyd, K. A., & Penzien, D. (1994). Psychosocial Interventions in the Management of Recurrent Headache Disorders 1: Overview and Effectiveness. Behavioral Medicine, 20(2):53.

Hussain, A. (1998). Intranasal drug delivery. Advanced Drug Delivery Reviews, 29(39).

İmani, S., Lv, S., Qian, H., Cui, Y., Li, X., Babaeizad, A., & Wang, Q. (2024). Current innovations in mRNA vaccines for targeting multidrug-resistant ESKAPE pathogens. Biotechnology Advances, 108492.

Iyengar S, Johnson KW, Ossipov MH, Aurora SK. (2019) CGRP and the Trigeminal System in Migraine. Headache. 59(5):659-681.

Jackson, J. L., Cogbill, E., Santana‐Davila, R., Eldredge, C., Collier, W., Gradall, A., Sehgal, N., & Kuester, J. (2015). A Comparative Effectiveness Meta-Analysis of Drugs for the Prophylaxis of Migraine Headache. PLoS ONE, 10(7).

Jang, S. C., & Gho, Y. S. (2014). Could Bioengineered Exosome-Mimetic Nanovesicles Be An Efficient Strategy for The Delivery of Chemotherapeutics? Nanomedicine, 9(2): 177.

Jenquin, M. R., Sarabia, R. E., Liebowitz, S. M., & McGinity, J. W. (1992). Relationship of Film Properties to Drug Release from Monolithic Films Containing Adjuvants. Journal of Pharmaceutical Sciences, 81(10): 983.

Jiang, Y., Zhu, J., Xu, G., & Liu, X. (2011). Intranasal delivery of stem cells to the brain. Expert Opinion on Drug Delivery, 8(5): 623.

Kabanov, A. V., & Vinogradov, S. V. (2009). Nanogels as Pharmaceutical Carriers: Finite Networks of Infinite Capabilities. Angewandte Chemie International Edition, 48(30): 5418.

Kalanuria AA & Lee P (2009). A Review of the Pharmacokinetics, Pharmacodynamics and Efficacy of Zolmitriptan in the Acute Abortive Treatment of Migraine. Clinical Medicine : Therapeutics. 1. 10.4137/CMT.S2056.

Kannavou, M., Karali, K., Κάτσιλα, Θ., Siapi, E., Marazioti, A., Klepetsanis, P., Calogeropoulou, T., Charalampopoulos, I., & Antimisiaris, S. G. (2023). BNN27 nanoformulations for nose-to-brain delivery. https://doi.org/10.54985/peeref.2306p3373108

Karavasili, C., & Fatouros, D. G. (2015). Smart materials: in situ gel-forming systems for nasal delivery. Drug Discovery Today, 21(1): 157.

Kefalides, P. T. (1998). New Methods for Drug Delivery. Annals of Internal Medicine, 128, p. 1053.

Kellstein, D., Lipton, R., Geetha, R., Koronkiewicz, K., Ft, E., Stewart, W., Wilkes, K., Furey, S. A., Subramanian, T., & Cooper, S. (2000). Evaluation of a Novel Solubilized Formulation of Ibuprofen in the Treatment of Migraine Headache: A Randomized, Double-Blind, Placebo-Controlled, Dose-Ranging Study. Cephalalgia, 20(4):233.

Kim JO, Nukolova NV, Oberoi HS, Kabanov AV, Bronich TK. (2009) Block Ionomer Complex Micelles with Cross-Linked Cores for Drug Delivery. Polym Sci Ser A Chem Phys. 51:708–718.

Kim, D., & Park, K. (2003). Swelling and mechanical properties of superporous hydrogels of poly(acrylamide-co-acrylic acid)/polyethylenimine interpenetrating polymer networks. Polymer, 45(1): 189.

Kim, S. W., Bae, Y. H., & Okano, T. (1992). Hydrogels: swelling, drug loading, and release. Pharmaceutical Research, 9(3): 283.

Knipe, J. M., & Peppas, N. A. (2014). Multi-responsive hydrogels for drug delivery and tissue engineering applications. Regenerative Biomaterials, 1(1): 57.

Kulkarni, N., Rao, P., Jadhav, G. S., Kulkarni, B., Kanakavalli, N., Kirad, S., Salunke, S., Tanpure, V., & Sahu, B. (2023). Emerging Role of Injectable Dipeptide Hydrogels in Biomedical Applications. ACS Omega, 8(4): 3551.

Kumar A and Kadian R (2023) Migraine Prophylaxis, In: StatPearls. Treasure Island (FL): StatPearls Publishing.

Kumar, M., Misra, A., Mishra, A. K., Mishra, P., & Pathak, K. (2008). Mucoadhesive nanoemulsion-based intranasal drug delivery system of olanzapine for brain targeting. Journal of drug targeting, 16(10): 806.

Langer, R. (1990). New Methods of Drug Delivery. Science, 249(4976): 1527.

Lehr, C., Bouwstra, J. A., Schacht, E., & Junginger, H. E. (1992). In vitro evaluation of mucoadhesive properties of chitosan and some other natural polymers. International Journal of Pharmaceutics, 78: 43.

Leone M, Proietti CA (2017) Advances in the understanding of cluster headache. Expert Rev Neurother. 17:165‐172.

Lin CH, Chen CH, Lin ZC, Fang JY (2017) Recent advances in oral delivery of drugs and bioactive natural products using solid lipid nanoparticles as the carriers, Journal of Food and Drug Analysis, 25(2):219-234.

Lochhead, J. J., Wolak, D. J., Pizzo, M. E., & Thorne, R. G. (2014). Rapid Transport within Cerebral Perivascular Spaces Underlies Widespread Tracer Distribution in the Brain after Intranasal Administration. Journal of Cerebral Blood Flow & Metabolism, 35(3): 371.

Lockman, P. R., Mumper, R. J., Khan, M. A., & Allen, D. D. (2002). Nanoparticle Technology for Drug Delivery Across the Blood-Brain Barrier. Drug Development and Industrial Pharmacy, 28(1):1.

Loder EW, Dowson AJ, Spierings EL (2005) Part II: clinical efficacy and tolerability of zolmitriptan orally disintegrating tablet in the acute treatment of migraine. Curr Med Res Opin. 21 Suppl 3:S8-12. doi: 10.1185/030079905x46278.

Lonzar, G., Abuín‐Porras, V., Del-Blanco-Muñiz, J. Á., González-de-la-Flor, Á., García-Pérez-de-Sevilla, G., & Domínguez-Balmaseda, D. (2023). Efficacy of invasive techniques in physical therapy for migraine treatment and prevention: a systematic review of randomized controlled trials. Revista Da Associação Médica Brasileira, 69(2):346.

Lunde CE, Wu Z, Reinecke A and Sieberg CB (2024) The Application of Cognitive Behavioral Therapy for Adolescent Patients With Endometriosis: A Topical Review, Cognitive and Behavioral Practice, 31(3): 383-398.

Macrì, M., Rendina, F., Feragalli, B., Pegreffi, F., & Festa, F. (2023). Prevalence of Ponticulus Posticus and Migraine in 220 Orthodontic Patients: A Cross-Sectional Study. Biology, 12(3): 471.

Martínez‐Serrano, R. D., Cuétara‐Guadarrama, F., Vonlanthen, M., Illescas, J., Zhu, X. X., & Rivera, E. (2023). Facile Obtainment of Fluorescent PEG Hydrogels Bearing Pyrene Groups by Frontal Polymerization. Polymers, 15(7): 1687.

Marupuru, S., Almatruk, Z., Slack, M., & Axon, D. R. (2023). Use of Pharmacological and Non-Pharmacological Strategies by Community-Dwelling Adults to Manage Migraine: A Systematic Review. Clinics and Practice, 13(3):553.

May A, Bahra A, Buchel C, et al. (1999) Functional magnetic resonance imaging in spontaneous attacks of SUNCT: short-lasting neuralgiform headache with conjunctival injection and tearing. Ann Neurol, 46: 791–794.

McKeage K. (2016) Zolmitriptan Nasal Spray: A Review in Acute Migraine in Pediatric Patients 12 Years of Age or Older. Paediatr Drugs. 18(1):75-81.

Merkus, F. W. H. M. (1999). Cyclodextrins in nasal drug delivery. Advanced Drug Delivery Reviews, 36(1): 41.

Messlinger K. (2018). The big CGRP flood - sources, sinks and signalling sites in the trigeminovascular system. J. Headache Pain 19:22. 10.1186/s10194-018-0848-0

Minn, A., Leclerc, S., Heydel, J., Minn, A.-L., Denizot, C., Cattarelli, M., Netter, P., & Grădinaru, D. (2002). Drug Transport into the Mammalian Brain: The Nasal Pathway and its Specific Metabolic Barrier. Journal of Drug Targeting, 10(4): 285.

Misra, A., & Kher, G. (2012). Drug Delivery Systems from Nose to Brain. Current Pharmaceutical Biotechnology, 13(12): 2355.

Mistry, A., Stolnik, S., & Illum, L. (2009). Nanoparticles for direct nose-to-brain delivery of drugs. International Journal of Pharmaceutics, 379(1):146.

Mittal, D., Ali, A., Md, S., Baboota, S., Sahni, J. K., & Ali, J. (2013). Insights into direct nose to brain delivery: current status and future perspective. Drug Delivery, 21(2): 75.

Moscovici, M., Hlevca, C., Căşărică, A., & Păvăloiu, R.-D. (2017). Nanocellulose and Nanogels as Modern Drug Delivery Systems, pp 209.

Mueller, C., Verroken, A., Iqbal, J., & Bernkop‐Schnuerch, A. (2011). Thiolated chitosans: In vitro comparison of mucoadhesive properties. Journal of Applied Polymer Science, 124(6): 5046.

Naoto U, Tatsuo O, Akira M, Takeshi K, Kohji N, Koichi N & Masao T (2005). Bioequivalence and Rapid Absorption of Zolmitriptan Nasal Spray Compared with Oral Tablets in Healthy Japanese Subjects. Clinical drug investigation. 25. 199-208. 10.2165/00044011-200525030-00006.

Narayanaswamy, R., & Torchilin, V. P. (2019). Hydrogels and Their Applications in Targeted Drug Delivery. Molecules, 24(3): 603.

Nielsen A, Dusek JA, Taylor-Swanson L and Tick H (2022) Acupuncture Therapy as an Evidence-Based Nonpharmacologic Strategy for Comprehensive Acute Pain Care: The Academic Consortium Pain Task Force White Paper Update. Pain Med. 23(9):1582-1612.

Oberoi HS, Nukolova NV, Laquer FC, Poluektova LY, Huang J, Alnouti Y, et al. (2012) Cisplatin-loaded core cross-linked micelles: comparative pharmacokinetics, antitumor activity, and toxicity in mice. Int J Nanomedicine. 7:2557.

Onoue, S., Yamada, S., & Chan, H. (2014). Nanodrugs: pharmacokinetics and safety. International Journal of Nanomedicine, 1025.

Paloncýová, M., Čechová, P., Šrejber, M., Kührová, P., & Otyepka, M. (2021). Role of Ionizable Lipids in SARS-CoV-2 Vaccines As Revealed by Molecular Dynamics Simulations: From Membrane Structure to Interaction with mRNA Fragments. The Journal of Physical Chemistry Letters, 12(45): 11199.

Patra, J.K., Das, G., Fraceto, L.F. et al. (2018) Nano based drug delivery systems: recent developments and future prospects. J Nanobiotechnol 16:71.

Pleș H, Florian IA, Timis TL, Covache-Busuioc RA, Glavan LA, Dumitrascu DI, Popa AA, Bordeianu A, Ciurea AV. (2023) Migraine: Advances in the Pathogenesis and Treatment. Neurol Int. 15(3):1052-1105.

Raffaelli B and Reuter U (2018) The Biology of Monoclonal Antibodies: Focus on Calcitonin Gene-Related Peptide for Prophylactic Migraine Therapy, Neurotherapeutics, 15(2): 324-335.

Raichur, A. M., Thomas, T., Radhakrishnan, K., Gnanadhas, D. P., & Chakravortty, D. (2013). Intracellular delivery of doxorubicin encapsulated in novel pH-responsive chitosan/heparin nanocapsules. International Journal of Nanomedicine, 267.

Rizwan, M., Yahya, R., Hassan, A., Yar, M., Azzahari, A. D., Selvanathan, V., Sonsudin, F., & Abouloula, C. N. (2017). pH Sensitive Hydrogels in Drug Delivery: Brief History, Properties, Swelling, and Release Mechanism, Material Selection and Applications. Polymers, 9(4): 137.

Rocca, M. A., Harrer, J. U., & Filippi, M. (2020). Are machine learning approaches the future to study patients with migraine? Neurology, 94(7): 291.

Rosa, G. D., Salzano, G., Caraglia, M., & Abbruzzese, A. (2011). Nanotechnologies: A Strategy to Overcome Blood-Brain Barrier. Current Drug Metabolism, 13(1):61.

Ruel-Gariépy, È., & Leroux, J. (2004). In situ-forming hydrogels—review of temperature-sensitive systems. European Journal of Pharmaceutics and Biopharmaceutics, 58(2): 409.

Sagris, Μ., Apostolos, A., Theofilis, P., Ktenopoulos, N., Katsaros, O., Tsalamandris, S., Tsioufis, K., Tοutouzas, K., & Tousoulis, D. (2024). Myocardial Ischemia–Reperfusion Injury: Unraveling Pathophysiology, Clinical Manifestations, and Emerging Prevention Strategies. Biomedicines, 12(4): 802.

Sahay G, Kim JO, Kabanov AV, Bronich TK (2010) The exploitation of differential endocytic pathways in normal and tumor cells in the selective targeting of nanoparticulate chemotherapeutic agents. Biomaterials. 31:923–933.

Salehi S, Naghib SM, Garshasbi HR, Ghorbanzadeh S, Zhang W (2023) Smart stimuli-responsive injectable gels and hydrogels for drug delivery and tissue engineering applications: A review. Front Bioeng Biotechnol. 22;11:1104126.

Sandri, G., Bonferoni, M. C., Ferrari, F., Mori, M., & Caramella, C. (2012). The role of chitosan as a mucoadhesive agent in mucosal drug delivery. Journal of Drug Delivery Science and Technology, 22(4): 275.

Scuteri D, Adornetto A, Rombolà L, Naturale MD, Morrone LA, Bagetta G, Tonin P, Corasaniti MT (2019) New Trends in Migraine Pharmacology: Targeting Calcitonin Gene-Related Peptide (CGRP) With Monoclonal Antibodies. Front Pharmacol. 9;10:363.

Seaber E, On N, Phillips S, Churchus R, Posner J, Pack RW (1996) The tolerability and pharmacokinetics of the novel an-timigraine compound 311C90 in healthy male volunteers. Br JClin Pharmacol 41: 141-147.

Seaber EJ, Peck RW, Smith DA, Allanson J, Hefting NR, van Lier JJ, Sollie FA, Wemer J, Jonkman JH. (1998) The absolute bioavailability and effect of food on the pharmacokinetics of zolmitriptan in healthy volunteers. Br J Clin Pharmacol. 46(5):433-9.

Seaber, E & Ridout, G & Layton, G & Posner, John & Peck, Richard. (1997). The novel anti-migraine compound zolmitriptan (Zomig 311C90) has no clinically significant interactions with paracetamol or metoclopramide. European journal of clinical pharmacology. 53. 229-34.

Şekerdağ, E. (2017). Nasal Physiology and Drug Transport. Elsevier eBooks (p. 93).

Selvaraj, K., Gowthamarajan, K. and Karri, V.V.S.R. (2018) Nose to brain transport pathways

an overview: potential of nanostructured lipid carriers in nose to brain targeting, Artif. Cells Nanomed. Biotechnol. 46 (2018) 2088–2095

Sheikhpour, M., Barani, L., & Kasaeian, A. (2017). Biomimetics in drug delivery systems: A critical review. Journal of Controlled Release, 253: 97.

Şimşek, F., Bilge, N., & Ceylan, M. (2020). Migren Klinik Özelliklerinin Nötrofil-Lenfosit Orani ile İlişkisi. Acibadem Universitesi Saglik Bilimleri Dergisi. https://doi.org/10.31067/0.2020.292

Smirnoff, L., & Pham, K. (2024). A Role for Visual Art Therapy in the Management of Migraine. Current Pain and Headache Reports, 28(4), 189.

Smitherman TA, Burch R, Sheikh H, Loder E. (2013) The prevalence, impact, and treatment of migraine and severe headaches in the United States: a review of statistics from national surveillance studies. Headache. 53(3):427-36.

Song, F., Gong, J., Tao, Y., Cheng, Y., Lu, J., & Wang, H. (2021). A robust regenerated cellulose-based dual stimuli-responsive hydrogel as an intelligent switch for controlled drug delivery. International Journal of Biological Macromolecules, 176, 448.

Song, X., Zhu, Q., Su, L., Shi, L., Chi, H., Yan, Y., Mei, L., Xu, X., Liu, B., Liu, Z., & Yang, J. (2024). New perspectives on migraine treatment: a review of the mechanisms and effects of complementary and alternative therapies. Frontiers in Neurology, 15.

Soni KS, Desale SS, Bronich TK. (2016) Nanogels: An overview of properties, biomedical applications and obstacles to clinical translation. J Control Release, 28;240:109-126.

Sonvico, F., Clementino, A., Buttini, F., Colombo, G., Pescina, S., Guterres, S. S., Pohlmann, A. R., & Nicoli, S. (2018). Surface-Modified Nanocarriers for Nose-to-Brain Delivery: From Bioadhesion to Targeting. Pharmaceutics, 10(1): 34.

Sosnik, A., & Seremeta, K. P. (2017). Polymeric Hydrogels as Technology Platform for Drug Delivery Applications. Gels, 3(3): 25.

Sprenger T, Viana M, Tassorelli C. (2018) Current Prophylactic Medications for Migraine and Their Potential Mechanisms of Action. Neurotherapeutics, 15(2):313-323.

Stovner L., Hagen K., Jensen R., Katsarava Z., Lipton R., Scher A., Steiner T., Zwart J.-A. (2007) The Global Burden of Headache: A Documentation of Headache Prevalence and Disability Worldwide. Cephalalgia. 27:193–210.

Sturgeon JA, Ehde DM, Darnall BD, Barad MJ, Clauw DJ, Jensen MP (2023) Psychological Approaches for Migraine Management. Anesthesiol Clin.41(2):341-355.

Tamani, F., Bassand, C., Hamoudi, M. C., Siepmann, F., & Siepmann, J. (2021). Mechanistic explanation of the (up to) 3 release phases of PLGA microparticles: Monolithic dispersions studied at lower temperatures. International Journal of Pharmaceutics, 596: 120220.

Teleanu, D. M., Chircov, C., Grumezescu, A. M., Volceanov, A., & Teleanu, R. I. (2018). Blood-Brain Delivery Methods Using Nanotechnology. Pharmaceutics, 10(4):269.

Tenci, M., Rossi, S., Giannino, V., Vigani, B., Sandri, G., Bonferoni, M. C., Daglia, M., Longo, L. M., Macelloni, C., & Ferrari, F. (2019). An In Situ Gelling System for the Local Treatment of Inflammatory Bowel Disease (IBD). The Loading of Maqui (Aristotelia chilensis) Berry Extract as an Antioxidant and Anti-Inflammatory Agent. Pharmaceutics, 11(11): 611.

Thomas, C., & Ahsan, F. (2007). Nasal Delivery of Peptide and Nonpeptide Drugs (p. 591).

Tiwari, S., Atluri, V., Kaushik, A., Yndart, A., & Nair, M. (2019). Alzheimer’s disease: pathogenesis, diagnostics, and therapeutics. International Journal of Nanomedicine, 5541.

Ülker, Z., & Erkey, C. (2014). An emerging platform for drug delivery: Aerogel based systems. Journal of Controlled Release, 177: 51.

Vaghela, S., Chaudhary, S., & Chaudhary, A. (2021). A Systemic Review on the Self Micro Emulsifying Drug Delivery System. International Journal of Pharmaceutical Sciences Review and Research, 69(1).

Van Hal M, Dydyk AM and Green MS (2025) Acupuncture, In: StatPearls. Treasure Island (FL): StatPearls Publishing.

VanderPluym JH, Halker Singh RB, Urtecho M, et al. (2021) Acute Treatments for Episodic Migraine in Adults: A Systematic Review and Meta-analysis. JAMA. 325(23):2357–2369.

Veronesi, M. C., Zamora, M., Bhuiyan, M., O’Brien‐Penney, B., Chen, C. T., & Vannier, M. W. (2017). Use of a clinical PET/CT scanner for whole body biodistribution of intranasal nanoparticles. arXiv (Cornell University). https://doi.org/10.48550/arxiv.1704.00691

Viganò, A., Toscano, M., Puledda, F., & Piero, V. D. (2019). Treating Chronic Migraine With Neuromodulation: The Role of Neurophysiological Abnormalities and Maladaptive Plasticity. Frontiers in Pharmacology, 10.

Vikelis, M., Dermitzakis, E. V., Vlachos, G. S., Soldatos, P., Spingos, K. C., Litsardopoulos, P., Kararizou, E., & Argyriou, A. A. (2020). Open Label Prospective Experience of Supplementation with a Fixed Combination of Magnesium, Vitamin B2, Feverfew, Andrographis Paniculata and Coenzyme Q10 for Episodic Migraine Prophylaxis. Journal of Clinical Medicine, 10(1):67.

Vinogradov SV (2006) Colloidal microgels in drug delivery applications. Curr Pharm Des. 12:4703–4712.

Vyas, T., Shahiwala, A., Marathe, S., & Misra, A. (2005). Intranasal Drug Delivery for Brain Targeting. Current Drug Delivery, 2(2): 165.

Wattiez AS, Sowers LP, Russo AF. (2020) Calcitonin gene-related peptide (CGRP): role in migraine pathophysiology and therapeutic targeting. Expert Opin Ther Targets. 24(2):91-100.

Ways, T. M. M., Lau, W. M., & Khutoryanskiy, V. V. (2018). Chitosan and Its Derivatives for Application in Mucoadhesive Drug Delivery Systems. Polymers, 10(3):267.

Wild MJ, McKillop D, Butters CJ. (1999) Determination of the human cytochrome P450 isoforms involved in the metabolism of zolmitriptan. Xenobiotica. 29(8):847-57. doi: 10.1080/004982599238290. PMID: 10553725.

Woensel, M. V., Wauthoz, N., Rosière, R., Amighi, K., Mathieu, V., Lefranc, F., Gool, S. V., & Vleeschouwer, S. D. (2013). Formulations for Intranasal Delivery of Pharmacological Agents to Combat Brain Disease: A New Opportunity to Tackle GBM? Cancers, 5(3):1020.

Wu, D., Chen, Q., Chen, X., Han, F., Chen, Z., & Wang, Y. (2023). The blood–brain barrier: Structure, regulation and drug delivery. Signal Transduction and Targeted Therapy, 8(1).

Yuan H., Lauritsen C. G., Kaiser E. A., Silberstein S. D. (2017). CGRP monoclonal antibodies for migraine: rationale and progress. BioDrugs 31 487–501. 10.1007/s40259-017-0250-5

Zobdeh F, Kraiem AB, Attwood MM, Chubarev VN, Tarasov VV, Schiöth HB and Mwinyi J (2021) Pharmacological treatment of migraine: Drug classes, mechanisms of action, clinical trials and new treatments. 178(23): 4588-4607.

Downloads

Published

2025-07-30

How to Cite

1.
Kannao S, Banweer J. A Review on Thermosensitive Zolmitriptan Mucoadhesive NanobioGel: Intranasal Delivery for Abortive treatment of Migraine. J Neonatal Surg [Internet]. 2025Jul.30 [cited 2025Sep.21];14(2S):578-97. Available from: https://jneonatalsurg.com/index.php/jns/article/view/8632