In vitro and in vivo evaluation of a novel testosterone transdermal delivery system (TTDS) using palm oil base

Document Type : Original Article

Authors

1 Shimadzu-UMMC Center for Xenobiotics Studies (SUCXeS), University of Malaya, 50603 Kuala Lumpur, Malaysia

2 University of Malaya Bioequivalence and Testing Center (UBAT), Department of Pharmacology, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia

3 Department of Pharmacy, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia

Abstract

Objective (s): Transdermal preparations for testosterone are becoming popular because of their unique advantages such as avoidance of first-pass effect, convenience, improved bioavailability, and reduction of systemic side effects. A novel testosterone transdermal delivery system (TDDS) was developed using a palm oil base called HAMINTM (a commercial product) and tested using in vitro and in vivo skin permeability test methods.
Materials and Methods: The physical characteristics of the formulation such as particle size and viscosity were determined by using Franz diffusion cell and Brookfield viscometer, respectively. In vivo skin permeability test was performed on healthy rabbits through the skin. Testosterone in serum was analyzed using the validated Liquid Chromatography-Tandem Mass Spectrometry (LC-MS/MS) technique.
Results:  In vitro study showed that the cumulative amount of testosterone diffused was between 40 to 1400 ngcm-² over a period of five hr after application of TDDS through the artificial Strat-M™ membrane. In the in vivo rabbit skin permeability test, the results indicated that testosterone was well absorbed with a mean Cmax and Tmax of 60.94 ngml-1 and 2.29 hr after application of TDDS while no increase was observed in placebo treatment. Particle size analysis ranged from 79.4 nm to 630.0 nm for placebo and 97 to 774.0 nm for TDDS.
Conclusion: The formulation was successfully prepared using HAMINTM, which has demonstrated great potential for topical delivery of testosterone.

Keywords


1.  Mazur A, Booth A. Testosterone and dominance in men. Behav Brain Sci 1998; 21:353-397.
2.  Nieschlag E. Testosterone treatment comes of age: new options for hypogonadal men. Clin Endocrinol 2006; 65:275–281.
3. Nieschlag E, Behre HM, Bouchard P, Corrales JJ, Jones TH, Stalla GK, et al. Testosterone replacement therapy: current trends and future directions. Hum Reprod  2004; 10:409–419.
4. Bhasin S, Cunningham GR, Hayes FJ, Matsumoto AM, Snyder PJ, Swerdloff RS et al. Testosterone therapy in adult men with androgen deficiency syndromes: an endocrine society clinical practice guideline. J Clin Endocrinol Metab  2010; 95:2536–2559.
5. Katznelson L, Finkelstein J, Schoenfeld D, Rosenthal K, Klibanski A. Increase in bone density and lean body mass during testosterone administration in men with acquired hypogonadism. J Clin Endocrinol Metab 1996; 81: 4358–4365.
6. Behre H, Kliesch S, Leifke R, Link R, Nieschlag E. Long-term effect of testosterone therapy on bone mineral density in hypogonadal men. J Clin Endocrinol Metab 1997; 82:2386-2390.
7. Wang C, Alexander G, Berman N, Salehian B, Davidson T, McDonald V, et al. Testosterone replacement therapy improves mood in hypogonadal men-a clinical research center study. J Clin Endocrinol Metab 1996; 81:3578-3583.
8. Handelsman DJ, Conway AJ, Boylan LM. Pharmacokinetics and pharmacodynamics of testosterone pellets In man. J Clin Endocrinol Metab 1990; 71;216-222.
9. Korenman SG, Viosca S, Garza D, Guralnik M,                Place V, Campbell P, et al. Androgen therapy of hypogonadal men with transscrotal testosterone systems. Am J Med 1987; 83:8.
10. Fossa S, Opjordsmoen S, Haug E. Androgen replacement and quality of life in patients treated for bilateral testicular cancer. Eur J Cancer 1999; 35:1220-1225.
11. Naik A, Kalia YN, Guy HR. Transdermal Drug Delivery: Overcoming The Skin’s Barrier Function. Pharm Sci Technol Today  2000;3:318–326.
12. Thomas BJ, Finnin BC.The transdermal revolution. Drug Discov Today 2004; 9:697-703.
13. Ware RS. Transdermal delivery of steroids. Contraception 1989; 39:1-20.
14. Zeven AC. The partial and complete domestication of the oil palm (Elaeis guineensis). Economy Botany, 1972; 26:274-279.
15. Malaysian Palm Oil Board, M.P.O.B., Economics & Industry Development Division, 2012.
16. Abdulkarim MF, Abdullah GZ, chitneni M, salman IM, Ameer OZ, Yam MF, et al. Topical piroxicam in vitro release and in vivo anti-inflammatory and analgesic effects from palm oil esters-based nanocream. Int J Nanomedcine 2010; 5:915-924.
17. Zainol S, Basri M, Basri HB, Shamsuddin AF, Abdul-Gani SS, Karjiban RA, et al. Formulation optimization of a palm-based nanoemulsion system containing levodopa. Int J Mol Sci 2012; 13:13049-13064.
18. Sakeena M, Muthanna F, Ghassan Z, Kanakal M, Elrashid S, Munavvar A, et al. Formulation and in vitro evaluation of ketoprofen in palm oil esters nanoemulsion for topical delivery. J Oleo Sci 2010; 59:223-228.
19. Guide for Industry B.M.V., Guide for Industry, Bioanalytical Method Validation, U.S Department of Health and Human Services Food and Drug Administration (FDA). 2001. 5-15
20. Turner AS. Animal models of osteoporosis - necessity and limitations. Eur Cells Materials 2001; 1:66-81.
21. Hye Jin Chung, Lim S, Kim IS, Bu Y, Kim H, Kim DH, Yoo HH. Simultaneous determination of baicalein, baicalin, wogonin, and wogonoside in rat plasma by LC-MS/MS for studying the pharmacokinetics of the standardized extract of scutellariae radix. Bull Korean Chem Soc  2012; 33:177.
22. Bartek MJ, Labudde JA, Maibach HI. Skin permeability in vivo: Comparison in rat, rabbit,pig and man. J Invest Dermatol 1972; 58:114-123.
23. Godin B, Touitou E. Transdermal skin delivery: Predictions for humans from in vivo ex vivo and animal models. Adv Drug Deliv Rev 2007; 59:1152-1161.
24.  Dick IP, Scott RC. Pig ear skin as an in-vitro model for human skin permeability. J Pharm Pharm 1992; 44: 640-645.
25.  Lu W, Luo H, Wu Y, Zhu Z, Wangn H. Preparation and characterization of a metered dose transdermal spray for testosterone. Acta Pharm Sin B 2013; 3:392-399.
26. Kalustian P. Pharmaceutical and cosmetic uses of palm and lauric products. J Am Oil Chem Soc 1985; 62:431-432.
27. Duffield J, Shapouri H, Graboski M, McCormick R, Wilson R. New Markets for Conventional and Genetically Modified Agricultural Products. In: E.R.S. Department of Agriculture.editor. Washington, DC: US, 1998.
28. Mahat SA.The Palm Oil Industry From The Perspective of Sustainable Development: A Case Study of Malaysian Palm Oil Industry. In: Graduate School of Asia Pacific Studies, Ritsumeikan Asia Pacific University of Japan, 2012.
29. Salama, Malaysia: Palm oil base for phramaceutical and cosmetic products, in, 2011.
30. Rolland A, Wagner N, Shroot B, Schaefer H. Pharmaceutical particulate carriers: Therapeutic Applications, J Controlled Release, 1993; 34: 274-275.
31. El-Kamel AH, Al-Fagih IM, Alsarra IA. Testosterone solid lipid microparticles for transdermal drug delivery. Formulation and physicochemical characterization. J Microencapsul 2007; 24:457-475.
32.  Akomeah FK, Martin GP, Brown MB. Variability in human skin permeability in vitro: comparing penetrants with different physicochemical properties. J Pharm Sci  2007; 96:824-834.
33. Franz TJ. Percutaneous absorption. On the relevance of In vitro data. J Invest Dermatol 1975; 64:190-195.
34.  Joshi V, Brewster D, Colonero P. In vitro diffusion studies in transdermal research: a synthetic membrane model in place of human skin drug development and Delivery, 2012.
35. Imran SA, Anand U, Agu R. Human Skin Substitute (Strat-M®) As an Alternative for Testing Transdermal Delivery of Levothyroxine (T4), Endocrine Society. 2014.
36. Kim MK, Zhao H, Lee CH, Kim DD. Formulation of
a reservoir-type testosterone transdermal delivery system. Int J Pharm 2001; 219:51-59.