Planta Med 2010; 76(1): 62-69
DOI: 10.1055/s-0029-1185949
Pharmacology
Original Papers
© Georg Thieme Verlag KG Stuttgart · New York

Characterization of in Vitro Pharmacokinetic Properties of Hoodigogenin A from Hoodia gordonii

Vamsi Lakshmi Mohan Madgula1 , Bharathi Avula1 , Rahul S. Pawar1 , Yatin J. Shukla1 , Ikhlas A. Khan1 , 2 , Larry A. Walker1 , 3 , Shabana I. Khan1
  • 1National Center for Natural Products Research, School of Pharmacy, The University of Mississippi, University, MS, USA
  • 2Department of Pharmacognosy, School of Pharmacy, The University of Mississippi, University, MS, USA
  • 3Department of Pharmacology, School of Pharmacy, The University of Mississippi, University, MS, USA
Further Information

Publication History

received April 16, 2009 revised June 4, 2009

accepted June 11, 2009

Publication Date:
28 July 2009 (online)

Abstract

This study was aimed to predict the pharmacokinetic properties of hoodigogenin A, which is the aglycone of the oxypregnane steroidal glycoside P57AS3 (P57) isolated from Hoodia gordonii. A series of in vitro assays was used to predict its gastric, intestinal and metabolic stability, intestinal and blood brain barrier (BBB) transport, protein binding and interaction with major drug metabolising enzymes. In the simulated gastric fluid, hoodigogenin A was stable (2 % degradation in 60 minutes) whereas P57 was unstable (45 % degradation in 30 minutes). In simulated intestinal fluid, P57 was degraded to an extent of 8 % in 180 minutes, while hoodigogenin A was stable. Hoodigogenin A was efficiently transported by passive diffusion across Caco-2 and MDR1-MDCK monolayers with Papp values in the range of 32 × 10−6 cm/sec and 22 × 10−6 cm/sec, respectively. The compound was metabolically unstable in human liver microsomes and S9 fractions with a CL′int of 71 and 120 mL/min/kg, respectively and was bound to the plasma proteins to an extent of 92 %. The compound strongly inhibited CYP3A4 activity (IC50 3 µM), indicating a possibility of drug-herb/botanical interactions when products containing H. gordonii are used simultaneously with other botanicals/herbs/drugs.

References

  • 1 South African National Biodiversity Institute .Plants of Southern Africa. Available at www.plantzafrica.com. Accessed April 4, 2009
  • 2 Rubin D I, Cawthorne M A, Bindra J S. Extracts, compounds and pharmaceutical compositions having anti-diabetic activity and their uses. US Patent 7416744B2 2008
  • 3 Rubin D I, Cawthorne M A, Bindra J S. Extracts, compounds and pharmaceutical compositions having anti-diabetic activity and their use. Euro Patent 1166792 2002
  • 4 Pawar R S, Shukla Y J, Khan S I, Avula B, Khan I A. New oxypregnane glycosides from appetite suppressant herbal supplement Hoodia gordonii.  Steroids. 2007;  72 524-534
  • 5 van Heerden F R, Marthinus Horak R, Maharaj V J, Vleggaar R, Senabe J V, Gunning P J. An appetite suppressant from Hoodia species.  Phytochemistry. 2007;  68 2545-2553
  • 6 MacLean D B, Luo L G. Increased ATP content/production in the hypothalamus may be a signal for energy-sensing of satiety: studies of the anorectic mechanism of a plant steroidal glycoside.  Brain Res. 2004;  1020 1-11
  • 7 Madgula V L, Avula B, Pawar R S, Shukla Y J, Khan I A, Walker L A, Khan S I. In vitro metabolic stability and intestinal transport of P57AS3 (P57) from Hoodia gordonii and its interaction with drug metabolizing enzymes.  Planta Med. 2008;  74 1269-1275
  • 8 Gee J M, DuPont M S, Day A J, Plumb G W, Williamson G, Johnson I T. Intestinal transport of quercetin glycosides in rats involves both deglycosylation and interaction with the hexose transport pathway.  J Nutr. 2000;  130 2765-2771
  • 9 Day A J, Canada F J, Diaz J C, Kroon P A, McLauchlan R, Faulds C B, Plumb G W, Morgan M R, Williamson G. Dietary flavonoid and isoflavone glycosides are hydrolysed by the lactase site of lactase phlorizin hydrolase.  FEBS Lett. 2000;  468 166-170
  • 10 Day A J, DuPont M S, Ridley S, Rhodes M, Rhodes M J, Morgan M R, Williamson G. Deglycosylation of flavonoid and isoflavonoid glycosides by human small intestine and liver beta-glucosidase activity.  FEBS Lett. 1998;  436 71-75
  • 11 Andlauer W, Kolb J, Furst P. Isoflavones from tofu are absorbed and metabolized in the isolated rat small intestine.  J Nutr. 2000;  130 3021-3027
  • 12 Liu Y, Hu M. Absorption and metabolism of flavonoids in the caco-2 cell culture model and a perused rat intestinal model.  Drug Metab Dispos. 2002;  30 370-377
  • 13 Walgren R A, Karnaky Jr K J, Lindenmayer G E, Walle T. Efflux of dietary flavonoid quercetin 4′-beta-glucoside across human intestinal Caco-2 cell monolayers by apical multidrug resistance-associated protein-2.  J Pharmacol Exp Ther. 2000;  294 830-836
  • 14 Brown J E, Khodr H, Hider R C, Rice-Evans C A. Structural dependence of flavonoid interactions with Cu2+ ions: implications for their antioxidant properties.  Biochem J. 1998;  330 (Pt 3) 1173-1178
  • 15 Kay C D. Aspects of anthocyanin absorption, metabolism and pharmacokinetics in humans.  Nutr Res Rev. 2006;  19 137-146
  • 16 Gumbleton M, Audus K L. Progress and limitations in the use of in vitro cell cultures to serve as a permeability screen for the blood-brain barrier.  J Pharm Sci. 2001;  90 1681-1698
  • 17 Wang Q, Rager J D, Weinstein K, Kardos P S, Dobson G L, Li J, Hidalgo I J. Evaluation of the MDR-MDCK cell line as a permeability screen for the blood-brain barrier.  Int J Pharm. 2005;  288 349-359
  • 18 The United States Pharmacopeia XXXI/The National Formulary 26 .Solutions/Test solutions. Rockville; The United States Pharmacopeial Convention 2008: 817
  • 19 Asafu-Adjaye E B, Faustino P J, Tawakkul M A, Anderson L W, Yu L X, Kwon H, Volpe D A. Validation and application of a stability-indicating HPLC method for the in vitro determination of gastric and intestinal stability of venlafaxine.  J Pharm Biomed Anal. 2007;  43 1854-1859
  • 20 Madgula V L, Avula B, Reddy V L N, Khan I A, Khan S I. Transport of decursin and decursinol angelate across Caco-2 and MDR-MDCK cell monolayers: in vitro models for intestinal and blood-brain barrier permeability.  Planta Med. 2007;  73 330-335
  • 21 Madgula V L, Avula B, Choi Y W, Pullela S V, Khan I A, Walker L A, Khan S I. Transport of Schisandra chinensis extract and its biologically-active constituents across Caco-2 cell monolayers – an in-vitro model of intestinal transport.  J Pharm Pharmacol. 2008;  60 363-370
  • 22 Obach R S. Prediction of human clearance of twenty-nine drugs from hepatic microsomal intrinsic clearance data: an examination of in vitro half-life approach and nonspecific binding to microsomes.  Drug Metab Dispos. 1999;  27 1350-1359
  • 23 Crespi C L, Miller V P, Penman B W. Microtiter plate assays for inhibition of human, drug-metabolizing cytochromes P450.  Anal Biochem. 1997;  248 188-190
  • 24 Harvard Bioscience Inc. .Guide to equilibrium dialysis. Southborough; The Nest Group, Inc. 2008
  • 25 Food and Drug Administration .Guidance for industry, waiver of in vivo bioavailability and bioequivalence studies for immediate release solid oral dosage forms containing certain active moieties/active ingredients based on biopharmaceutical classification system. Rockville; CDER, Department of Health and Human Services 2000
  • 26 Gres M C, Julian B, Bourrie M, Meunier V, Roques C, Berger M, Boulenc X, Berger Y, Fabre G. Correlation between oral drug absorption in humans, and apparent drug permeability in TC-7 cells, a human epithelial intestinal cell line: comparison with the parental Caco-2 cell line.  Pharm Res. 1998;  15 726-733
  • 27 Stewart B H, Chan O H, Lu R H, Reyner E L, Schmid H L, Hamilton H W, Steinbaugh B A, Taylor M D. Comparison of intestinal permeabilities determined in multiple in vitro and in situ models: relationship to absorption in humans.  Pharm Res. 1995;  12 693-699
  • 28 Artursson P, Karlsson J. Correlation between oral drug absorption in humans and apparent drug permeability coefficients in human intestinal epithelial (Caco-2) cells.  Biochem Biophys Res Commun. 1991;  175 880-885
  • 29 Garberg P, Ball M, Borg N, Cecchelli R, Fenart L, Hurst R D, Lindmark T, Mabondzo A, Nilsson J E, Raub T J, Stanimirovic D, Terasaki T, Oberg J O, Osterberg T. In vitro models for the blood-brain barrier.  Toxicol In Vitro. 2005;  19 299-334
  • 30 Camenisch G, Alsenz J, van de Waterbeemd H, Folkers G. Estimation of permeability by passive diffusion through Caco-2 cell monolayers using the drugs' lipophilicity and molecular weight.  Eur J Pharm Sci. 1998;  6 317-324
  • 31 Obach R S, Baxter J G, Liston T E, Silber B M, Jones B C, MacIntyre F, Rance D J, Wastall P. The prediction of human pharmacokinetic parameters from preclinical and in vitro metabolism data.  J Pharmacol Exp Ther. 1997;  283 46-58
  • 32 Butterweck V, Derendorf H, Gaus W, Nahrstedt A, Schulz V, Unger M. Pharmacokinetic herb-drug interactions: are preventive screenings necessary and appropriate?.  Planta Med. 2004;  70 784-791
  • 33 CDER .Drug interaction studies: study design data analysis, and implications for dosing and labelling. Rockville; Food and Drug Administration 2004
  • 34 Liu Y, Yang L. Early metabolism evaluation making traditional Chinese medicine effective and safe therapeutics.  J Zhejiang Univ Sci B. 2006;  7 99-106
  • 35 Barone G W, Gurley B J, Ketel B L, Lightfoot M L, Abul-Ezz S R. Drug interaction between St. John's wort and cyclosporine.  Ann Pharmacother. 2000;  34 1013-1016
  • 36 Rosenblatt M, Mindel J. Spontaneous hyphema associated with ingestion of Ginkgo biloba extract.  N Engl J Med. 1997;  336 1108
  • 37 Usia T, Iwata H, Hiratsuka A, Watabe T, Kadota S, Tezuka Y. CYP3A4 and CYP2D6 inhibitory activities of Indonesian medicinal plants.  Phytomedicine. 2006;  13 67-73
  • 38 Huang J D, Oie S. Effect of altered disopyramide binding on its pharmacologic response in rabbits.  J Pharmacol Exp Ther. 1982;  223 469-471
  • 39 Qin M, Nilsson M, Oie S. Decreased elimination of drug in the presence of alpha-1-acid glycoprotein is related to a reduced hepatocyte uptake.  J Pharmacol Exp Ther. 1994;  269 1176-1181
  • 40 Shand D G, Cotham R H, Wilkinson G R. Perfusion-limited of plasma drug binding on hepatic drug extraction.  Life Sci. 1976;  19 125-130
  • 41 Jansen J A. Influence of plasma protein binding kinetics on hepatic clearance assessed from a “tube” model and a “well-stirred” model.  J Pharmacokinet Biopharm. 1981;  9 15-26

Shabana I. Khan

School of Pharmacy
National Center for Natural Products Research
University of Mississippi

University, MS 38677

USA

Phone: + 01 66 29 15 10 41

Fax: + 01 66 29 15 70 62

Email: skhan@olemiss.edu

    >