Skip to main content

Advertisement

Log in

Pharmacokinetic and pharmacodynamic evaluation of Solid self-nanoemulsifying delivery system (SSNEDDS) loaded with curcumin and duloxetine in attenuation of neuropathic pain in rats

  • Original Article
  • Published:
Neurological Sciences Aims and scope Submit manuscript

Abstract

The present investigation is focused on improving oral bioavailability of poorly soluble and lipophilic drugs, curcumin (CRM) and duloxetine (DXH), through the solid self-nanoemulsifying drug delivery system (S-SNEDDS) and identifying their potential against attenuation of NP in chronic constriction injury (CCI)–induced rats through the solid self-nanoemulsifying drug delivery system (S-SNEDDS). The optimized batch of S-SNEDDS reported was containing CRM and DXH (30 mg each), castor oil (20% w/w), tween-80 (40% w/w), transcutol-P (40% w/w), and syloid 244 FP (1 g). The high dose of each of naïve CRM (NCH), naïve DXH (NDH), physical mixture of DXH and CRM (C-NCM-DXH), S-SNEDDS-CRM (SCH), S-SNEDDS-DXH (SDH), and S-SNEDDS-CRM-DXH (C-SCH-SDH) was subjected for MTT assay. The developed formulations were subjected to pharmacokinetic studies and results showed about 8 to 11.06 and 2-fold improvement in oral bioavailability of CRM and DXH through S-SNEDDS. Furthermore, CCI-induced male Wistar rats were treated with SSNEDDS containing CRM and DXH, S-SNEDDS containing individual drug, individual naïve forms, and their combination from the day of surgery for 14 days and evaluated for behavioral at pre-determined time intervals. On the terminal day, animals were sacrificed to assess tissue myeloperoxidase, superoxide anion, protein, tumor necrosis factor-α, total calcium levels, and histopathological changes. Pronounced effect was observed in rats treated with S-SNEDDS containing both drugs with respect to rats receiving any of other treatments owing to enhanced oral bioavailability through S-SNEDDS. Therefore, it can be concluded that S-SNEDDS of both drugs and their coadministration can accelerate the prevention of NP.

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

References

  1. Zhang M-T, Wang B, Jia Y-N, Liu N, Ma P-S, Gong S-S, Niu Y, Sun T, Li Y-X, Yu J-Q (2017) Neuroprotective effect of liquiritin against neuropathic pain induced by chronic constriction injury of the sciatic nerve in mice. Biomed Pharmacother 95:186–198

    CAS  PubMed  Google Scholar 

  2. L. Colloca, T. Ludman, D. Bouhassira, R. Baron, A.H. Dickenson, D. Yarnitsky, R. Freeman, A. Truini, N. Attal, N.B. Finnerup, Neuropathic pain, Nature reviews Disease primers 3 (2017) 17002

  3. Dworkin RH, O’connor AB, Audette J, Baron R, Gourlay GK, Haanpää ML, Kent JL, Krane EJ, LeBel AA, Levy RM (2010) Recommendations for the pharmacological management of neuropathic pain: an overview and literature update, Mayo Clinic Proceedings, Elsevier, pp. S3-S14

  4. Gangadhar M, Kumar Mishra R, Sriram D, Yogeeswari P (2014) Future directions in the treatment of neuropathic pain: a review on various therapeutic targets. CNS & Neurological Disorders-Drug Targets (Formerly Current Drug Targets-CNS & Neurological Disorders) 13(1):63–81

    CAS  Google Scholar 

  5. Schestatsky P, Vidor L, Winckler PB, Araújo TGd, Caumo W (2014) Promising treatments for neuropathic pain. Arquivos de neuro-psiquiatria 72(11):881–888

    PubMed  Google Scholar 

  6. Mendlik MT, Uritsky TJ (2015) Treatment of neuropathic pain. Curr Treat Options Neurol 17(12):50

    PubMed  Google Scholar 

  7. Kahya MC, Nazıroğlu M, Övey İS (2017) Modulation of diabetes-induced oxidative stress, apoptosis, and Ca 2+ entry through TRPM2 and TRPV1 channels in dorsal root ganglion and hippocampus of diabetic rats by melatonin and selenium. Mol Neurobiol 54(3):2345–2360

    CAS  PubMed  Google Scholar 

  8. Halliwell B (2006) Oxidative stress and neurodegeneration: where are we now? J Neurochem 97(6):1634–1658

    CAS  PubMed  Google Scholar 

  9. Uslusoy F, Nazıroğlu M, Övey İS, Sönmez TT (2019) Hypericum perforatum L. supplementation protects sciatic nerve injury-induced apoptotic, inflammatory and oxidative damage to muscle, blood and brain in rats. Journal of Pharmacy and Pharmacology 71(1):83–92

    CAS  PubMed  Google Scholar 

  10. Díaz-Triste NE, González-García MP, Jiménez-Andrade JM, Castañeda-Hernández G, Chávez-Piña AE (2014) Pharmacological evidence for the participation of NO–c GMP–K ATP pathway in the gastric protective effect of curcumin against indomethacin-induced gastric injury in the rat. Eur J Pharmacol 730:102–106

    PubMed  Google Scholar 

  11. Jeon Y, Kim C-E, Jung D, Kwak K, Park S, Lim D, Kim S, Baek W (2013) Curcumin could prevent the development of chronic neuropathic pain in rats with peripheral nerve injury. Curr Ther Res 74:1–4

    CAS  PubMed  Google Scholar 

  12. Kaur M, Singh A, Kumar B, Singh SK, Bhatia A, Gulati M, Prakash T, Bawa P, Malik AH (2017) Protective effect of co-administration of curcumin and sildenafil in alcohol induced neuropathy in rats. Eur J Pharmacol 805:58–66

    CAS  PubMed  Google Scholar 

  13. Zhao X, Xu Y, Zhao Q, Chen C-R, Liu A-M, Huang Z-L (2012) Curcumin exerts antinociceptive effects in a mouse model of neuropathic pain: descending monoamine system and opioid receptors are differentially involved. Neuropharmacology 62(2):843–854

    CAS  PubMed  Google Scholar 

  14. Argyriou AA, Kyritsis AP, Makatsoris T, Kalofonos HP (2014) Chemotherapy-induced peripheral neuropathy in adults: a comprehensive update of the literature. Cancer Manag Res 6:135–147

    PubMed  PubMed Central  Google Scholar 

  15. Perahia D, Kajdasz D, Walker D, Raskin J, Tylee A (2006) Duloxetine 60 mg once daily in the treatment of milder major depressive disorder. Int J Clin Pract 60(5):613–620

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Sagman D, McIntosh D, Lee M, Li H, Ruschel S, Hussain N, Granger R, Lee A, Raskin J (2011) Attributes of response in depressed patients switched to treatment with duloxetine. Int J Clin Pract 65(1):73–81

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Katsuyama S, Aso H, Otowa A, Yagi T, Kishikawa Y, Komatsu T, Sakurada T, Nakamura H (2014) Antinociceptive effects of the serotonin and noradrenaline reuptake inhibitors milnacipran and duloxetine on vincristine-induced neuropathic pain model in mice, ISRN Pain 2014

  18. Murakami M, Osada K, Mizuno H, Ochiai T, Alev L, Nishioka K (2015) A randomized, double-blind, placebo-controlled phase III trial of duloxetine in Japanese fibromyalgia patients. Arthritis research & therapy 17(1):224

    Google Scholar 

  19. Patel K, Padhye S, Nagarsenker M (2012) Duloxetine HCl lipid nanoparticles: preparation, characterization, and dosage form design. AAPS PharmSciTech 13(1):125–133

    PubMed  Google Scholar 

  20. Smith EML, Pang H, Cirrincione C, Fleishman S, Paskett ED, Ahles T, Bressler LR, Fadul CE, Knox C, Le-Lindqwister N (2013) Effect of duloxetine on pain, function, and quality of life among patients with chemotherapy-induced painful peripheral neuropathy: a randomized clinical trial. Jama 309(13):1359–1367

    PubMed  PubMed Central  Google Scholar 

  21. Kumar B, Garg V, Singh S, Pandey NK, Bhatia A, Prakash T, Gulati M, Singh SK (2018) Impact of spray drying over conventional surface adsorption technique for improvement in micromeritic and biopharmaceutical characteristics of self-nanoemulsifying powder loaded with two lipophilic as well as gastrointestinal labile drugs. Powder Technol 326:425–442

    CAS  Google Scholar 

  22. Sood S, Jain K, Gowthamarajan K (2014) Optimization of curcumin nanoemulsion for intranasal delivery using design of experiment and its toxicity assessment. Colloids Surf B: Biointerfaces 113:330–337

    CAS  PubMed  Google Scholar 

  23. Rao SVR, Shao J (2008) Self-nanoemulsifying drug delivery systems (SNEDDS) for oral delivery of protein drugs: I. Formulation development, International journal of pharmaceutics 362(1):2–9

    CAS  Google Scholar 

  24. Rajesh SY, Singh SK, Pandey NK, Sharma P, Bawa P, Kumar B, Gulati M, Jain SK, Gowthamarajan K, Singh S (2018) Impact of various solid carriers and spray drying on pre/post compression properties of solid SNEDDS loaded with glimepiride: in vitro-ex vivo evaluation and cytotoxicity assessment, Drug Dev Ind Pharm 1–14

  25. Bennett GJ, Xie Y-K (1988) A peripheral mononeuropathy in rat that produces disorders of pain sensation like those seen in man. Pain 33(1):87–107

    PubMed  Google Scholar 

  26. Muthuraman A, Jaggi AS, Singh N, Singh D (2008) Ameliorative effects of amiloride and pralidoxime in chronic constriction injury and vincristine induced painful neuropathy in rats. Eur J Pharmacol 587(1–3):104–111

    CAS  PubMed  Google Scholar 

  27. Sommer C, Schäfers M (1998) Painful mononeuropathy in C57BL/Wld mice with delayed Wallerian degeneration: differential effects of cytokine production and nerve regeneration on thermal and mechanical hypersensitivity1. Brain Res 784(1–2):154–162

    CAS  PubMed  Google Scholar 

  28. Muthuraman A, Singh N (2011) Attenuating effect of Acorus calamus extract in chronic constriction injury induced neuropathic pain in rats: an evidence of anti-oxidative, anti-inflammatory, neuroprotective and calcium inhibitory effects. BMC Complement Altern Med 11(1):24

    PubMed  PubMed Central  Google Scholar 

  29. Muthuraman A, Singh N, Jaggi AS (2011) Effect of hydroalcoholic extract of Acorus calamus on tibial and sural nerve transection-induced painful neuropathy in rats. J Nat Med 65(2):282–292

    PubMed  Google Scholar 

  30. Eddy NB, Touchberry CF, Lieberman JE (1950) Synthetic analgesics: I. Methadone isomers and derivatives. Journal of Pharmacology and Experimental Therapeutics 98(2):121–137

    CAS  Google Scholar 

  31. Kaur G, Jaggi AS, Singh N (2010) Exploring the potential effect of Ocimum sanctum in vincristine-induced neuropathic pain in rats. Journal of brachial plexus and peripheral nerve injury 5(1):3

    PubMed  PubMed Central  Google Scholar 

  32. Muthuraman A, Diwan V, Jaggi AS, Singh N, Singh D (2008) Ameliorative effects of Ocimum sanctum in sciatic nerve transection-induced neuropathy in rats. J Ethnopharmacol 120(1):56–62

    CAS  PubMed  Google Scholar 

  33. Muthuraman A, Singh N (2012) Neuroprotective effect of saponin rich extract of Acorus calamus L. in rat model of chronic constriction injury (CCI) of sciatic nerve-induced neuropathic pain. Journal of Ethnopharmacology 142(3):723–731

    CAS  PubMed  Google Scholar 

  34. Di Wang H, Pagano PJ, Du Y, Cayatte AJ, Quinn MT, Brecher P, Cohen RA (1998) Superoxide anion from the adventitia of the rat thoracic aorta inactivates nitric oxide. Circ Res 82(7):810–818

    CAS  PubMed  Google Scholar 

  35. Severenghaus J, Ferrebee J (1950) Calcium determination by flame photometry; methods for serum, urine, and other fluids. J Biol Chem 187:621–630

    Google Scholar 

  36. Lowry OH, Rosebrough NJ, Farr AL, Randall RJ (1951) Protein measurement with the Folin phenol reagent. J Biol Chem 193(1):265–275

    CAS  PubMed  Google Scholar 

  37. Ellman GL (1959) Tissue sulfhydryl groups. Arch Biochem Biophys 82(1):70–77

    CAS  PubMed  Google Scholar 

  38. Kaur G, Bedi O, Sharma N, Singh S, Deshmukh R, Kumar P (2016) Anti-hyperalgesic and anti-nociceptive potentials of standardized grape seed proanthocyanidin extract against CCI-induced neuropathic pain in rats. J Basic Clin Physiol Pharmacol 27(1):9–17

    CAS  PubMed  Google Scholar 

  39. Joshi RP, Negi G, Kumar A, Pawar YB, Munjal B, Bansal AK, Sharma SS (2013) SNEDDS curcumin formulation leads to enhanced protection from pain and functional deficits associated with diabetic neuropathy: an insight into its mechanism for neuroprotection. Nanomedicine 9(6):776–785

    CAS  PubMed  Google Scholar 

  40. Leung L, Cahill CM (2010) TNF-α and neuropathic pain-a review. J Neuroinflammation 7(1):27

    PubMed  PubMed Central  Google Scholar 

  41. Kumar B, Garg V, Singh A, Pandey NK, Singh S, Panchal S, Melkani I, Raji R, Axel M, Mohanta S, Jyoti J, Som S, Gulati M, Bhatia A, T P, Singh SK (2018) Investigation and optimization of formulation parameters for selfnanoemulsifying delivery system of two lipophilic and gastrointestinal labile drugs using box-behnken design, 2018, 7

  42. Melkani I, Kumar B, Panchal S, Singh SK, Singh A, Gulati M, Gill SBS, Jyoti J, Pandey NK, Kumar S (2019) Comparison of sildenafil, fluoxetine and its co-administration against chronic constriction injury induced neuropathic pain in rats: an influential additive effect, Neurol Res 1–8

  43. Rasoulian B, Hajializadeh Z, Esmaeili-Mahani S, Rashidipour M, Fatemi I, Kaeidi A (2019) Neuroprotective and antinociceptive effects of rosemary (Rosmarinus officinalis L.) extract in rats with painful diabetic neuropathy, the. J Physiol Sci 69(1):57–64

    CAS  PubMed  Google Scholar 

  44. Birmann PT, Sousa FS, Domingues M, Brüning CA, Vieira BM, Lenardão EJ, Savegnago L (2019) 3-(4-Chlorophenylselanyl)-1-methyl-1H-indole promotes recovery of neuropathic pain and depressive-like behavior induced by partial constriction of the sciatic nerve in mice. J Trace Elem Med Biol 54:126–133

    CAS  PubMed  Google Scholar 

  45. Nakamura S, Atsuta Y (2006) The effects of experimental neurolysis on ectopic firing in a rat chronic constriction nerve injury model. The Journal of hand surgery 31(1):35–39

    PubMed  Google Scholar 

  46. Jaggi AS, Jain V, Singh N (2011) Animal models of neuropathic pain. Fundamental & clinical pharmacology 25(1):1–28

    CAS  Google Scholar 

  47. Maves TJ, Pechman PS, Gebhart G, Meller ST (1993) Possible chemical contribution from chromic gut sutures produces disorders of pain sensation like those seen in man. Pain 54(1):57–69

    PubMed  Google Scholar 

  48. Zhang W, Suo M, Yu G, Zhang M (2019) Antinociceptive and anti-inflammatory effects of cryptotanshinone through PI3K/Akt signaling pathway in a rat model of neuropathic pain. Chem Biol Interact 305:127–133

    CAS  PubMed  Google Scholar 

  49. Sasaki H, Kihara M, Zollman PJ, Nickander KK, Smithson IL, Schmelzer JD, Willner CL, Benarroch EE, Low PA (1996) Chronic constriction model of rat sciatic nerve: nerve blood flow, morphologic and biochemical alterations. Acta Neuropathol 93(1):62–70

    Google Scholar 

  50. E.P.o.F. Additives, N.S.a.t. Food (2010) Scientific opinion on the re-evaluation of curcumin (E 100) as a food additive. EFSA Journal 8(9):1679

    Google Scholar 

  51. Zhu J, Xiao L, Xue Z, Zhang Q, Yang C, Meng J (2019) Duloxetine, a balanced serotonin-norepinephrine reuptake inhibitor, improves painful chemotherapy-induced peripheral neuropathy by inhibiting the activation of p38 MAPK and NF-κB. Front Pharmacol 10:365

    PubMed  PubMed Central  Google Scholar 

  52. Izham M, Nadiah M, Hussin Y, Aziz MNM, Yeap SK, Rahman HS, Masarudin MJ, Mohamad NE, Abdullah R, Alitheen NB (2019) Preparation and characterization of self nano-emulsifying drug delivery system loaded with citraland its antiproliferative effect on colorectal cells in vitro. Nanomaterials 9(7):1028

    CAS  Google Scholar 

  53. Young W (1992) Role of calcium in central nervous system injuries. J Neurotrauma 9:S9–S25

    PubMed  Google Scholar 

  54. Thiagarajan VR, Shanmugam P, Krishnan UM, Muthuraman A, Singh N (2012) Ameliorative potential of Butea monosperma on chronic constriction injury of sciatic nerve induced neuropathic pain in rats. An Acad Bras Cienc 84(4):1091–1104

    PubMed  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to T. Prakash.

Ethics declarations

Conflict of interest

None

Ethical approval

Approval for this study was granted with protocol no: LPU/LSPS/IAEC/CPCSEA/MEETING NO.1/SEPTEMBER2015/2016 PROTOCOL NO.2. All parts of experiment were strictly carried out in accordance with the Committee for the Purpose of Control and Supervision of Experiments on Animals (CPCSEA).

Additional information

Publisher’s note

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

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Kumar, B., Singh, S.K., Prakash, T. et al. Pharmacokinetic and pharmacodynamic evaluation of Solid self-nanoemulsifying delivery system (SSNEDDS) loaded with curcumin and duloxetine in attenuation of neuropathic pain in rats. Neurol Sci 42, 1785–1797 (2021). https://doi.org/10.1007/s10072-020-04628-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s10072-020-04628-7

Keywords

Navigation