Skip to main content
Log in

Physiological Modeling of Altered Pharmacokinetics of a Novel Anticancer Drug, UCN-01 (7-Hydroxystaurosporine), Caused by Slow Dissociation of UCN-01 from Human α1-Acid Glycoprotein

  • Published:
Pharmaceutical Research Aims and scope Submit manuscript

Abstract

Purpose. The extremely low clearance and small distribution volumeof UCN-01 in humans could be partly due to the high degree of bindingto hAGP (1,2). The quantitative effects of hAGP on the pharmacokineticsof UCN-01 at several levels of hAGP and UCN-01 were estimatedin rats given an infusion of hAGP to mimic the clinical situation anda physiological model for analysis was developed.

Methods. The plasma concentrations of UCN-01 (72.5–7250 nmol/kgiv) in rats given an infusion of hAGP, 15 or 150 nmol/h/kg, weremeasured by HPLC. Pharmacokinetic analysis under conditionsassuming rapid equilibrium of protein binding and incorporating thedissociation rate was conducted.

Results. The Vdss and CLtot of UCN-01 (725 nmol/kg iv) in ratsgiven an infusion of hAGP, 150 nmol/h/kg, fell to about 1/250 and 1/700that in control rats. The Vdss and CLtot following 72.5–7250nmol/kg UCN-01 to rats given 150 nmol/h/kg hAGP were 63.9–688ml/kg and 3.18–32.9 ml/h/kg, respectively, indicating non-linearitydue to saturation of UCN-01 binding. The CLtot estimated by thephysiological model assuming rapid equilibrium of UCN-01 bindingto hAGP, was six times higher than the observed value while the CLtotestimated by the model incorporating koff, measured using DCC, wascomparable with the observed value.

Conclusions. These results suggest that the slow dissociation ofUCN-01 from hAGP limits its disposition and elimination.

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.

Similar content being viewed by others

REFERENCES

  1. E. Fuse, H. Tanii, N. Kurata, H. Kobayashi, Y. Shimada, T. Tamura, Y. Sasaki, Y. Tanigawara, R. D. Lush, D. Headlee, W. D. Figg, S. G. Arbuck, A. M. Senderowicz, E. A. Sausville, S. Akinaga, T. Kuwabara, and S. Kobayashi. Unpredicted clinical pharmacology of UCN-01 caused by specific binding to human α1-acid glycoprotein. Cancer Res. 58:3248–3253 (1998).

    Google Scholar 

  2. E. Fuse, H. Tanii, K. Takai, K. Asanome, N. Kurata, H. Kobayashi, T. Kuwabara, S. Kobayashi, and Y. Sugiyama. Altered pharmacokinetics of a novel anticancer drug, UCN-01 caused by specific high affinity binding to α1-acid glycoprotein in humans. Cancer Res. 59:1054–1060 (1999).

    Google Scholar 

  3. I. Takahashi, E. Kobayashi, K. Asano, M. Yoshida, and H. Nakano. UCN-01, a selective inhibitor of protein kinase C from Streptomyces. J. Antibiot. 40:1782–1784 (1987).

    Google Scholar 

  4. S. Akinaga, K. Gomi, M. Morimoto, T. Tamaoki, and M. Okabe. Antitumor activity of UCN-01, a selective inhibitor of protein kinase C, in murine and human tumor models. Cancer Res. 51:4888–4892 (1991).

    Google Scholar 

  5. C. M. Seynaeve, M. Stetler-Stevenson, S. Sebers, G. Kaur, E. A. Sausville, and P. J. Worland. Cell cycle arrest and inhibition by the protein kinase antagonist UCN-01 in human breast carcinoma cells. Cancer Res. 53:2081–2086 (1993).

    Google Scholar 

  6. S. Akinaga, K. Nomura, K. Gomi, and M. Okabe. Effect of UCN-01, a selective inhibitor of protein kinase C, on the cell-cycle distribution of human epidermoid carcinoma, A431 cells. Cancer Chemother. Pharmacol. 33:273–280 (1994).

    Google Scholar 

  7. K. Kawakami, H. Futami, J. Takahara, and K. Yamaguchi. UCN-01, 7-hydroxyl-staurosporine, inhibits kinase activity of cyclin-dependent kinases and reduces the phosphorylation of the retinoblastoma susceptibility gene product in A549 human lung cancer cell line. Biochem. Biophys. Res. Commun. 219:778–783 (1996).

    Google Scholar 

  8. T. Akiyama, T. Yoshida, T. Tsujita, M. Shimizu, T. Mizukami, M. Okabe, and S. Akinaga. G1 phase accumulation induced by UCN-01 is associated with dephosphorylation of Rb and CDK2 proteins as well as induction of CDK inhibitor p21/Cip/WAF1/ Sdi1 in p53-mutated human epidermoid carcinoma A431 cells. Cancer Res. 57:1495–1501 (1997).

    Google Scholar 

  9. S. Akinaga, K. Nomura, K. Gomi, and M. Okabe. Enhancement of antitumor activity of mitomycin C in vitro and in vivo by UCN-01, a selective inhibitor of protein kinase C. Cancer Chemother. Pharmacol. 32:183–189 (1993).

    Google Scholar 

  10. S. Akinaga, K. Nomura, K. Gomi, and M. Okabe. Synergistic antitumor effect of UCN-01, a protein kinase (C) inhibitor, combined with various anti-cancer agents. Proc. Am. Assoc. Cancer Res. 33:3072 (1992).

    Google Scholar 

  11. R. T. Bunch, and A. Eastman. Enhancement of cisplatin-induced cytotoxicity by 7-hydroxystaurosporine (UCN-01), a new G2-checkpoint inhibitor. Clin. Cancer Res. 2:791–797 (1996).

    Google Scholar 

  12. N. Kurata, T. Kuwabara, H. Tanii, E. Fuse, T. Akiyama, S. Akinaga, H. Kobayashi, K. Yamaguchi, and S. Kobayashi. Pharmacokinetics and pharmacodynamics of a novel protein kinase inhibitor, UCN-01. Cancer Chemother. Pharmacol. 44:12–18 (1999).

    Google Scholar 

  13. J. M. H. Kremer, J. Wilting, and L. H. M. Janssen. Drug binding to human alpha-1-acid glycoprotein in health and disease. Pharmacol. Rev. 40:1–47 (1988).

    Google Scholar 

  14. G. R. Wilkinson. Plasma and tissue binding considerations in drug disposition. Drug Metab. Rev. 14:427–465 (1983).

    Google Scholar 

  15. N. Kurata, T. Kuramitsu, H. Tanii, E. Fuse, T. Kuwabara, H. Kobayashi, and S. Kobayashi. Development of a highly sensitive high-performance liquid chromatographic method for measuring an anticancer drug, UCN-01 in human plasma or urine. J. Chromatogr. B 708:223–227 (1998).

    Google Scholar 

  16. M. Gibaldi and D. Perrier. Noncompartment analysis based on statistical moment theory. In M. Gibaldi, and D. Perrier (eds.), Pharmacokinetics, Marcel Dekker, New York, 1982, pp. 409–417.

    Google Scholar 

  17. N. Watari, and L. Z. Benet. Determinant of mean input time, mean residence time, and steady-state volume of distribution with multiple drug inputs. J. Pharmacokin. Biopharm. 17:593–599 (1989).

    Google Scholar 

  18. K. S. Pang, and M. Rawland. Hepatic clearance of drugs. I. Theoretical considerations of “well-stirred” model and a “parallel tube” model. Influence of hepatic blood flow, plasma and blood cell binding, and the hepatocellular enzymatic activity on hepatic drug clearance. J. Pharmacokin. Biopharm. 5:625–653 (1977).

    Google Scholar 

  19. B. Davies, and T. Morris. Physiological parameters in laboratory animals and humans. Pharm. Res. 10:1093–1095 (1993).

    Google Scholar 

  20. T. Fujita, and A. Koshiro. Development of the convenient and direct numerical analytical method of the pharmacokinetics of phenytoin by an ordinary and/or the Bayesian weighted least-squares method using the program MULTI2 (BAYES). J. Pharmacobio-Dyn. 12:717–725 (1989).

    Google Scholar 

  21. K. Yamaoka, and T. Nakagawa. A nonlinear least squares program based on differential equations, MULTI (RUNGE), for microcomputers. J. Pharmacobio-Dyn. 6:595–606 (1983).

    Google Scholar 

  22. E. Fuse, T. Kobayashi, M. Inaba, and Y. Sugiyama. Prediction of the maximal tolerated dose (MTD) and therapeutic effect of anticancer drugs in humans: integration of pharmacokinetics with pharmacodynamics and toxicodynamics. Cancer Treat. Rev. 21:133–157 (1995).

    Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Rights and permissions

Reprints and permissions

About this article

Cite this article

Fuse, E., Hashimoto, A., Sato, N. et al. Physiological Modeling of Altered Pharmacokinetics of a Novel Anticancer Drug, UCN-01 (7-Hydroxystaurosporine), Caused by Slow Dissociation of UCN-01 from Human α1-Acid Glycoprotein. Pharm Res 17, 553–564 (2000). https://doi.org/10.1023/A:1007512832006

Download citation

  • Issue Date:

  • DOI: https://doi.org/10.1023/A:1007512832006

Navigation