Skip to main content
Log in

Probucol Release from Novel Multicompartmental Microcapsules for the Oral Targeted Delivery in Type 2 Diabetes

  • Research Article
  • Published:
AAPS PharmSciTech Aims and scope Submit manuscript

Abstract

In previous studies, we developed and characterised multicompartmental microcapsules as a platform for the targeted oral delivery of lipophilic drugs in type 2 diabetes (T2D). We also designed a new microencapsulated formulation of probucol-sodium alginate (PB-SA), with good structural properties and excipient compatibility. The aim of this study was to examine the stability and pH-dependent targeted release of the microcapsules at various pH values and different temperatures. Microencapsulation was carried out using a Büchi-based microencapsulating system developed in our laboratory. Using SA polymer, two formulations were prepared: empty SA microcapsules (SA, control) and loaded SA microcapsules (PB-SA, test), at a constant ratio (1:30), respectively. Microcapsules were examined for drug content, zeta potential, size, morphology and swelling characteristics and PB release characteristics at pH 1.5, 3, 6 and 7.8. The production yield and microencapsulation efficiency were also determined. PB-SA microcapsules had 2.6 ± 0.25% PB content, and zeta potential of −66 ± 1.6%, suggesting good stability. They showed spherical and uniform morphology and significantly higher swelling at pH 7.8 at both 25 and 37°C (p < 0.05). The microcapsules showed multiphasic release properties at pH 7.8. The production yield and microencapsulation efficiency were high (85 ± 5 and 92 ± 2%, respectively). The PB-SA microcapsules exhibited distal gastrointestinal tract targeted delivery with a multiphasic release pattern and with good stability and uniformity. However, the release of PB from the microcapsules was not controlled, suggesting uneven distribution of the drug within the microcapsules.

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.

Institutional subscriptions

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5

Similar content being viewed by others

References

  1. Barbeau WE, Bassaganya-Riera J, Hontecillas R. Putting the pieces of the puzzle together—a series of hypotheses on the etiology and pathogenesis of type 1 diabetes. Med Hypotheses. 2007;68(3):607–19.

    Article  CAS  PubMed  Google Scholar 

  2. Moore PA, Zgibor JC, Dasanayake AP. Diabetes: a growing epidemic of all ages. J Am Dent Assoc. 2003;134(Spec No):11S–5S.

    Article  PubMed  Google Scholar 

  3. Negrulj R, Mooranian A, Al-Salami H. Potentials and limitations of bile acids in type 2 diabetes: applications of microencapsulation as a novel oral delivery system. J Endocrinol Diabetes Mellitus. 2013;1:1–11.

    Google Scholar 

  4. Cani PD et al. Selective increases of bifidobacteria in gut microflora improve high-fat-diet-induced diabetes in mice through a mechanism associated with endotoxaemia. Diabetologia. 2007;50(11):2374–83.

    Article  CAS  PubMed  Google Scholar 

  5. Goldfine AB, Fonseca V, Shoelson SE. Therapeutic approaches to target inflammation in type 2 diabetes. Clin Chem. 2011;57(2):162–7.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  6. Wu R et al. Probucol ameliorates the development of nonalcoholic steatohepatitis in rats fed high-fat diets. Dig Dis Sci. 2013;58(1):163–71.

    Article  CAS  PubMed  Google Scholar 

  7. Yamashita S, Matsuzawa Y. Where are we with probucol: a new life for an old drug? Atherosclerosis. 2009;207(1):16–23.

    Article  CAS  PubMed  Google Scholar 

  8. Shimizu H et al. Probucol attenuated hyperglycemia in multiple low-dose streptozotocin-induced diabetic mice. Life Sci. 1991;49(18):1331–8.

    Article  CAS  PubMed  Google Scholar 

  9. Russell JC et al. Cardioprotective effect of probucol in the atherosclerosis-prone JCR:LA-cp rat. Eur J Pharmacol. 1998;350(2–3):203–10.

    Article  CAS  PubMed  Google Scholar 

  10. Zimetbaum P, Eder H, Frishman W. Probucol: pharmacology and clinical application. J Clin Pharmacol. 1990;30(1):3–9.

    Article  CAS  PubMed  Google Scholar 

  11. Vedantham K et al. Development of a probucol-releasing antithrombogenic drug eluting stent. J Biomed Mater Res B Appl Biomater. 2012;100(4):1068–77.

    Article  PubMed  Google Scholar 

  12. Nourooz-Zadeh J et al. Measurement of plasma probucol levels by high-performance liquid chromatography. J Chromatogr B Biomed Appl. 1994;654(1):55–60.

    Article  CAS  PubMed  Google Scholar 

  13. Ajun W et al. Preparation of aspirin and probucol in combination loaded chitosan nanoparticles and in vitro release study. Carbohydr Polym. 2009;75(4):566–74.

    Article  Google Scholar 

  14. Mooranian A, Negrulj R, Martinez J, Mathavan S, Sciarretta J, Chen-Tan N, et al. Stability and release kinetics of an advanced gliclazide-cholic acid formulation: the use of artificial-cell microencapsulation in slow release targeted oral delivery of antidiabetics. J Pharm Innov, 2014 (in press).

  15. Mooranian A, Negrulj R, Martinez J, Mathavan S, Sciarretta J, Chen-Tan N, Mukkur TK, Mikov M, Lalic-Popovic M, Stojančević M, Golocorbin-Kon S, Al-Salami H, A complex microencapsulated system: a platform for optimised oral delivery of antidiabetic drug-bile acid formulations. Pharmaceutical Development and Technology, 2014 (in press).

  16. Pal D, Nayak AK. Novel tamarind seed polysaccharide-alginate mucoadhesive microspheres for oral gliclazide delivery: in vitro-in vivo evaluation. Drug Deliv. 2012;19(3):123–31.

    Article  CAS  PubMed  Google Scholar 

  17. Awasthi R, Kulkarni GT. Development of novel gastroretentive drug delivery system of gliclazide: hollow beads. Drug Dev Ind Pharm, 2013(0):1–11.

  18. Mooranian A, et al. Stability and release kinetics of an advanced gliclazide-cholic acid formulation: the use of artificial-cell microencapsulation in slow release targeted oral delivery of antidiabetics. J Pharm Innov, 2014:1–8.

  19. Mooranian A, et al. Novel artificial cell microencapsulation of a complex gliclazide-deoxycholic bile acid formulation: a characterisation study. Drug Des Dev Ther, 2014 (in press).

  20. Mladenovska K et al. 5-ASA loaded chitosan–Ca–alginate microparticles: preparation and physicochemical characterization. Int J Pharm. 2007;345(1):59–69.

    Article  CAS  PubMed  Google Scholar 

  21. Abdelbary A, El-Gendy NA, Hosny A. Microencapsulation approach for orally extended delivery of glipizide: in vitro and in vivo evaluation. Indian J Pharm Sci. 2012;74(4):319–30.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  22. Duro R et al. The adsorption of cellulose ethers in aqueous suspensions of pyrantel pamoate: effects on zeta potential and stability. Eur J Pharm Biopharm. 1998;45(2):181–8.

    Article  CAS  PubMed  Google Scholar 

  23. Xie HG et al. Effect of surface wettability and charge on protein adsorption onto implantable alginate chitosan alginate microcapsule surfaces. J Biomed Mater Res Part A. 2010;92(4):1357–65.

    Google Scholar 

  24. George M, Abraham TE. Polyionic hydrocolloids for the intestinal delivery of protein drugs: alginate and chitosan—a review. J Control Release. 2006;114(1):1–14.

    Article  CAS  PubMed  Google Scholar 

  25. Yang L et al. Physicochemical and biological characterization of monoketocholic acid, a novel permeability enhancer. Mol Pharm. 2009;6(2):448–56.

    Article  CAS  PubMed  Google Scholar 

  26. Mikov M et al. Pharmacology of bile acids and their derivatives: absorption promoters and therapeutic agents. Eur J Drug Metab Pharmacokinet. 2006;31(3):237–51.

    Article  CAS  PubMed  Google Scholar 

  27. Mikov M et al. Pharmacokinetics and hypoglycaemic effect of 3 alpha, 7 alpha-dihydroxy-12-oxo-5beta-cholanate (MKC) in diabetic rat. FEBS J. 2006;273:210.

    Google Scholar 

  28. Al-Salami H et al. Probiotic treatment proceeded by a single dose of bile acid and gliclazide exert the most hypoglycemic effect in type 1 diabetic rats. Med Hypothesis Res. 2008;4(2):93–101.

    Google Scholar 

  29. Mikov M et al. The influence of 3alpha, 7alpha-dihydroxy-12-keto-5beta-cholanate on gliclazide pharmacokinetics and glucose levels in a rat model of diabetes. Eur J Drug Metab Pharmacokinet. 2008;33(3):137–42.

    Article  CAS  PubMed  Google Scholar 

  30. Al-Salami H et al. Influence of the semisynthetic bile acid (MKC) on the ileal permeation of gliclazide in healthy and diabetic rats. Pharmacol Rep. 2008;60(4):532–41.

    CAS  PubMed  Google Scholar 

  31. Al-Salami H et al. Probiotics decreased the bioavailability of the bile acid analog, monoketocholic acid, when coadministered with gliclazide, in healthy but not diabetic rats. Eur J Drug Metab Pharmacokinet. 2012;37(2):99–108.

    Article  CAS  PubMed  Google Scholar 

  32. Al-Salami H et al. Gliclazide reduces MKC intestinal transport in healthy but not diabetic rats. Eur J Drug Metab Pharmacokinet. 2009;34(1):43–50.

    Article  CAS  PubMed  Google Scholar 

  33. Al-Salami H et al. Probiotic treatment reduces blood glucose levels and increases systemic absorption of gliclazide in diabetic rats. Eur J Drug Metab Pharmacokinet. 2008;33(2):101–6.

    Article  CAS  PubMed  Google Scholar 

  34. Al-Kassas RS, Al-Gohary OM, Al-Faadhel MM. Controlling of systemic absorption of gliclazide through incorporation into alginate beads. Int J Pharm. 2007;341(1–2):230–7.

    Article  CAS  PubMed  Google Scholar 

  35. Efentakis M, Buckton G. The effect of erosion and swelling on the dissolution of theophylline from low and high viscosity sodium alginate matrices. Pharm Dev Technol. 2002;7(1):69–77.

    Article  CAS  PubMed  Google Scholar 

  36. Silva CM et al. Insulin encapsulation in reinforced alginate microspheres prepared by internal gelation. Eur J Pharm Sci. 2006;29(2):148–59.

    Article  CAS  PubMed  Google Scholar 

  37. Andersen E, Karlaganis G, Sjovall J. Altered bile acid profiles in duodenal bile and urine in diabetic subjects. Eur J Clin Investig. 1988;18(2):166–72.

    Article  CAS  Google Scholar 

  38. Cook MT et al. Microencapsulation of probiotics for gastrointestinal delivery. J Control Release. 2012;162(1):56–67.

    Article  CAS  PubMed  Google Scholar 

  39. Duboc H et al. Connecting dysbiosis, bile-acid dysmetabolism and gut inflammation in inflammatory bowel diseases. Gut. 2013;62(4):531–9.

    Article  CAS  PubMed  Google Scholar 

  40. Caesar R, Fak F, Backhed F. Effects of gut microbiota on obesity and atherosclerosis via modulation of inflammation and lipid metabolism. J Intern Med. 2010;268(4):320–8.

    Article  CAS  PubMed  Google Scholar 

  41. Cani PD et al. Changes in gut microbiota control inflammation in obese mice through a mechanism involving GLP-2-driven improvement of gut permeability. Gut. 2009;58(8):1091–103.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  42. Huang X, Brazel CS. On the importance and mechanisms of burst release in matrix-controlled drug delivery systems. J Control Release. 2001;73(2):121–36.

    Article  CAS  PubMed  Google Scholar 

  43. Narasimhan B, Langer R. Zero-order release of micro-and macromolecules from polymeric devices: the role of the burst effect. J Control Release. 1997;47(1):13–20.

    Article  CAS  Google Scholar 

Download references

Acknowledgments

The authors acknowledge the CHIRI at Curtin University and the Curtin-seeding grant for the support and also acknowledge the use of equipment, scientific and technical assistance of the Curtin University Electron Microscope Facility, which has been partially funded by the University, State and Commonwealth Governments.

Conflict of Interest

The authors declare no conflict of interest.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Hani Al-Salami.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Mooranian, A., Negrulj, R., Al-Sallami, H.S. et al. Probucol Release from Novel Multicompartmental Microcapsules for the Oral Targeted Delivery in Type 2 Diabetes. AAPS PharmSciTech 16, 45–52 (2015). https://doi.org/10.1208/s12249-014-0205-9

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1208/s12249-014-0205-9

KEY WORDS

Navigation