Skip to main content

Advertisement

Log in

Nose-to-brain delivery of lamotrigine-loaded PLGA nanoparticles

  • Original Article
  • Published:
Drug Delivery and Translational Research Aims and scope Submit manuscript

Abstract

Direct nose-to-brain delivery of drugs and faster onset of action have made intra-nasal route a much sought-after alternative to conventional routes of drug delivery to the brain. Lamotrigine is used for the treatment and management of neuropathic pain, and in the present work, lamotrigine (LTG)-PLGA nanoparticles were developed for intra-nasal delivery. The LTG-PLGA nanoparticles were prepared using modified nanoprecipitation method via high-speed homogenization and ultra-sonication techniques. Entrapment efficiency (EE%) of developed LTG-PLGA-NPs was found to be 84.87 ± 1.2% with drug loading of 10.21 ± 0.89%. The particle size of developed nanoparticles was found to be 184.6 nm with PDI value of 0.082 and zeta potential of − 18.8 mV. Dissolution profiles were studied in PBS (pH 7.4), simulated nasal fluid, and simulated cerebrospinal fluid where almost complete release was observed within 5 h in CSF. In vitro, cytotoxicity was analyzed using MTT assay where dose-dependent cytotoxicity was observed for developed LTG-PLGA-NPs. In vitro cytokine analysis showed positive effects of LTG-PLGA-NPs as pro-inflammatory cytokine suppressors. Further, in vivo studies were performed for radiolabeled formulation and drug (99mTc-LTG-PLGA-NPs and 99mTc-LTG-aqueous) using Sprague Dawley rats where with the help of gamma scintigraphy studies, various routes of administration viz. oral, intra-nasal, and intra-venous were compared. Various pharmacokinetic parameters were evaluated using biodistribution studies to estimate the drug levels in blood and brain. For 99mTc-LTG-PLGA-NPs via intra-nasal route, drug targeting efficiency (DTE%) was found to be 129.81% and drug target organ transport (DTP%) to be 22.81% in brain with Cmax of 3.82%/g within Tmax 1.5 h. Thus, the developed PLGA nanoparticles for intra-nasal delivery provide a possible alternative for existing available drug formulation for neuropathic pain management.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5
Fig. 6
Fig. 7

Similar content being viewed by others

Abbreviations

AEDs:

Antiepileptic drugs

IL:

Interleukin

I.N:

Intra-nasal

I.V.:

Intra-venous

LTG:

Lamotrigine

PLGA:

(Poly(lactic-co-glycolic acid)

NPs:

Nanoparticles

DTE:

Drug target efficiency

DTP:

Drug transport percentage

TNF-α:

Tumor necrosis factor alpha

IFN-γ:

Interferon gamma

99mTc:

Technetium-99m

References

  1. Siddall PJ, McClelland JM, Rutkowski SB, et al. A longitudinal study of the prevalence and characteristics of pain in the first 5 years following spinal cord injury. Pain. 2003;103(3):249–57.

    Article  PubMed  Google Scholar 

  2. Kehlet H, Jensen TS, Woolf CJ. Persistent postsurgical pain: risk factors and prevention. Lancet. 2006;367(9522):1618–25.

    Article  PubMed  Google Scholar 

  3. Caraceni A, Portenoy RK. An international survey of cancer pain characteristics and syndromes. Pain. 1999;82(3):263–74.

    Article  CAS  PubMed  Google Scholar 

  4. Hewitt DJ, McDonald M, Portenoy RK, Rosenfeld B, Passik S, Breitbart W. Pain syndromes and etiologies in ambulatory AIDS patients. Pain. 1997;70(2–3):117–23.

    Article  CAS  PubMed  Google Scholar 

  5. McNamara J. Drugs acting on the central nervous system. Goodman and Gilman’s the pharmacological basis of therapeutics. 9th ed. New York: McGraw-Hill; 1996. p. 461–86.

    Google Scholar 

  6. Han HC, Lee DH, Chung JM. Characteristics of ectopic discharges in a rat neuropathic pain model. PAIN®. 2000;84(2–3):253–61.

    CAS  Google Scholar 

  7. Chong E, Dupuis LL. Therapeutic drug monitoring of lamotrigine. Ann Pharmacother. 2002;36(5):917–20.

    Article  CAS  PubMed  Google Scholar 

  8. Srichaiya A, Longchoopol C, Oo-Puthinan S, Sayasathid J, Sripalakit P, Viyoch J. Bioequivalence of generic lamotrigine 100-mg tablets in healthy Thai male volunteers: a randomized, single-dose, two-period, two-sequence crossover study. Clin Ther. 2008;30(10):1844–51.

    Article  CAS  PubMed  Google Scholar 

  9. Castel-Branco M, Lebre V, Falcao A, Figueiredo I, et al. Relationship between plasma and brain levels and the anticonvulsant effect of lamotrigine in rats. Eur J Pharmacol. 2003;482(1–3):163–8.

    Article  CAS  PubMed  Google Scholar 

  10. Löscher W, Ganter M, Fassbender CP. Correlation between drug and metabolite concentrations in plasma and anesthetic action of ketamine in swine. Am J Vet Res. 1990;51(3):391–8.

    PubMed  Google Scholar 

  11. Walker MC, Tong X, Perry H, Alavijeh MS, Patsalos PN. Comparison of serum, cerebrospinal fluid and brain extracellular fluid pharmacokinetics of lamotrigine. Br J Pharmacol. 2000;130(2):242–8.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

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

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  13. Chaturvedi M, Kumar M, Pathak K. A review on mucoadhesive polymer used in nasal drug delivery system. J Adv Pharm Technol Res. 2011;2(4):215–22.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  14. Gentile P, Chiono V, Carmagnola I, Hatton P. An overview of poly (lactic-co-glycolic) acid (PLGA)-based biomaterials for bone tissue engineering. Int J Mol Sci. 2014;15(3):3640–59.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  15. Bilati U, Allémann E, Doelker E. Strategic approaches for overcoming peptide and protein instability within biodegradable nano- and microparticles. Eur J Pharm Biopharm. 2005;59(3):375–88.

    Article  CAS  PubMed  Google Scholar 

  16. Sharma D, Maheshwari D, Philip G, Rana R, Bhatia S, Singh M, et al. Formulation and optimization of polymeric nanoparticles for intranasal delivery of lorazepam using Box-Behnken design: in vitro and in vivo evaluation. Biomed Res Int. 2014;2014:1–14.

    Google Scholar 

  17. Lalani J, Patil S, Kolate A, et al. Protein-functionalized PLGA nanoparticles of lamotrigine for neuropathic pain management. AapsPharmscitech. 2015;16(2):413–27.

    CAS  Google Scholar 

  18. Shinde VR, Shelake MR, Shetty SS, Chavan-Patil AB, Pore YV, Late SG. Enhanced solubility and dissolution rate of lamotrigine by inclusion complexation and solid dispersion technique. J Pharm Pharmacol. 2008;60(9):1121–9.

    Article  CAS  PubMed  Google Scholar 

  19. Emami J, Ghassami N, Ahmadi F. Development and validation of a new HPLC method for determination of lamotrigine and related compounds in tablet formulations. J Pharm Biomed Anal. 2006;40(4):999–1005.

    Article  CAS  PubMed  Google Scholar 

  20. Ranjan AP, Mukerjee A, Helson L, Vishwanatha JK. Scale up, optimization and stability analysis of curcumin C3 complex-loaded nanoparticles for cancer therapy. J Nanobiotechnol. 2012;10(1):38.

    Article  CAS  Google Scholar 

  21. Romero-Pérez A, García-García E, Zavaleta-Mancera A, Ramírez-Bribiesca JE, Revilla-Vázquez A, Hernández-Calva LM, et al. Designing and evaluation of sodium selenite nanoparticles in vitro to improve selenium absorption in ruminants. Vet Res Commun. 2010;34(1):71–9.

    Article  PubMed  Google Scholar 

  22. Martinac A, Filipović-Grčić J, Perissutti B, Voinovich D, Pavelić Ž. Spray-dried chitosan/ethylcellulose microspheres for nasal drug delivery: swelling study and evaluation of in vitro drug release properties. J Microencapsul. 2005;22(5):549–61.

    Article  CAS  PubMed  Google Scholar 

  23. Mohanraj K, Sethuraman S, Krishnan UM. Development of poly (butylene succinate) microspheres for delivery of levodopa in the treatment of Parkinson’s disease. J Biomed Mater Res B Appl Biomater. 2013;101(5):840–7.

    Article  CAS  PubMed  Google Scholar 

  24. Sharma A, Gupta S, Sarethy IP, Dang S, Gabrani R. Green tea extract: possible mechanism and antibacterial activity on skin pathogens. Food Chem. 2012;135(2):672–5.

    Article  CAS  PubMed  Google Scholar 

  25. Sharma Y, Bashir S, Khan MF, et al. Inhibition of Src homology 2 domain containing protein tyrosine phosphatase as the possible mechanism of metformin-assisted amelioration of obesity induced insulin resistance in high fat diet fed C57BL/6J mice. Biochem Biophys Res Commun. 2017;487(1):54–61.

    Article  CAS  PubMed  Google Scholar 

  26. Kaur A, Saxena Y, Bansal R, Gupta S, Tyagi A, Sharma RK, et al. Intravaginal delivery of polyphenon 60 and curcumin nanoemulsion gel. AAPS PharmSciTech. 2017;18(6):2188–202.

    Article  CAS  PubMed  Google Scholar 

  27. Sharma D, Sharma RK, Sharma N, Gabrani R, Sharma SK, Ali J, et al. Nose-to-brain delivery of PLGA-diazepam nanoparticles. AAPS PharmSciTech. 2015;16(5):1108–21.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  28. Rastogi R, Sultana Y, Aqil M, et al. Alginate microspheres of isoniazid for oral sustained drug delivery. Int J Pharm. 2007;334(1–2):71–7.

    Article  CAS  PubMed  Google Scholar 

  29. Keck JP, McElroy SL. Clinical pharmacodynamics and pharmacokinetics of antimanic and mood-stabilizing medications. J Clin Psychiatry. 2002;63:3–11.

    CAS  PubMed  Google Scholar 

  30. Kumar M, Misra A, Babbar AK, Mishra AK, Mishra P, Pathak K. Intranasal nanoemulsion based brain targeting drug delivery system of risperidone. Int J Pharm. 2008;358(1–2):285–91.

    Article  CAS  PubMed  Google Scholar 

  31. Cojocaru V, Ranetti AE, Hinescu LG, et al. Formulation and evaluation of in vitro release kinetics of Na3CaDTPA decorporation agent embedded in microemulsion-based gel formulation for topical delivery. Farmacia. 2015;63(5):656–64.

    CAS  Google Scholar 

  32. Shen S, Wu Y, Liu Y, Wu D. High drug-loading nanomedicines: progress, current status, and prospects. Int J Nanomedicine. 2017;12:4085–109.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  33. Zheng S, Xie Y, Li Y, et al. Development of high drug-loading nanomicelles targeting steroids to the brain. Int J Nanomedicine. 2014;9:55.

    PubMed  Google Scholar 

  34. Dong X. Current strategies for brain drug delivery. Theranostics. 2018;8(6):1481–93.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  35. Singhvi G, Singh M. In-vitro drug release characterization models. Int J Pharm Stud Res. 2011;2(1):77–84.

    Google Scholar 

  36. Kim DH, Martin DC. Sustained release of dexamethasone from hydrophilic matrices using PLGA nanoparticles for neural drug delivery. Biomaterials. 2006;27(15):3031–7.

    Article  CAS  PubMed  Google Scholar 

  37. Fu Y, Kao WJ. Drug release kinetics and transport mechanisms of non-degradable and degradable polymeric delivery systems. Expert Opin Drug Deliv. 2010;7(4):429–44.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  38. Mantovani A, Sica A, Sozzani S, et al. The chemokine system in diverse forms of macrophage activation and polarization. Trends Immunol. 2004;25(12):677–86.

    Article  CAS  PubMed  Google Scholar 

  39. Hamilton TA. Molecular basis of macrophage activation: from gene expression to phenotypic diversity. Macrophage. 2002;2:73–102.

    Google Scholar 

  40. Hines DJ, Choi HB, Hines RM, Phillips AG, MacVicar BA. Prevention of LPS-induced microglia activation, cytokine production and sickness behavior with TLR4 receptor interfering peptides. PLoS One. 2013;8(3):e60388.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  41. Murray CL, Skelly DT, Cunningham C. Exacerbation of CNS inflammation and neurodegeneration by systemic LPS treatment is independent of circulating IL-1β and IL-6. J Neuroinflammation. 2011;8(1):50.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  42. Dinarello CA. Proinflammatory cytokines. Chest. 2000;118(2):503–8.

    Article  CAS  PubMed  Google Scholar 

  43. Liu W, Wu YH, Zhang L, et al. Elevated serum levels of IL-6 and IL-17 may associate with the development of ankylosing spondylitis. Int J Clin Exp Med. 2015;8(10):17362.

    PubMed Central  PubMed  Google Scholar 

  44. Yan Y, Guo TM, Zhu C. Effects of non-steroidal anti-inflammatory drugs on serum proinflammatory cytokines in the treatment of ankylosing spondylitis. Biochem Cell Biol. 2018;96:450–6.

    Article  CAS  PubMed  Google Scholar 

  45. Nigam K, Kaur A, Tyagi A, Manda K, Gabrani R, Dang S. Baclofen-loaded poly (d, l-lactide-co-glycolic acid) nanoparticles for neuropathic pain management: in vitro and in vivo evaluation. Rejuvenation Res 2018. https://doi.org/10.1089/rej.2018.2119.

Download references

Acknowledgements

The authors would like to acknowledge Jaypee Institute of Information Technology, Noida, and Institute of Nuclear Medicine & Allied Sciences, Delhi, for providing basic infrastructural support to carry out the project. The authors would like to thank Dr. A. Panda, National Institute of Immunology, Delhi, and Dr. M. Kalia, Translational Health Science And Technology Institute, Faridabad, for providing required resources for completion of this work.

Author information

Authors and Affiliations

Authors

Contributions

The manuscript was written through contributions of all authors. All authors have given approval to the final version of the manuscript.

Corresponding author

Correspondence to Shweta Dang.

Ethics declarations

All institutional and national guidelines for the care and use of laboratory animals were followed.

Conflict of interest

The authors declare that there is no conflict of interest.

Additional information

Publisher’s note

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

Electronic supplementary material

Fig S1

Particle size distribution of developed LTG-PLGA-NPs. (PNG 19 kb)

High resolution image (TIFF 27 kb)

Fig S2

Zeta potential distribution of developed LTG-PLGA-NPs (PNG 15 kb)

High resolution image (TIFF 20 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Nigam, K., Kaur, A., Tyagi, A. et al. Nose-to-brain delivery of lamotrigine-loaded PLGA nanoparticles. Drug Deliv. and Transl. Res. 9, 879–890 (2019). https://doi.org/10.1007/s13346-019-00622-5

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s13346-019-00622-5

Keywords

Navigation