Skip to main content
Log in

An Integrated Multiple-Analyte Pharmacokinetic Model to Characterize Trastuzumab Emtansine (T-DM1) Clearance Pathways and to Evaluate Reduced Pharmacokinetic Sampling in Patients with HER2-Positive Metastatic Breast Cancer

  • Original Research Article
  • Published:
Clinical Pharmacokinetics Aims and scope Submit manuscript

Abstract

Background and Objective

Trastuzumab emtansine (T-DM1) is an antibody–drug conjugate recently approved by the US Food and Drug Administration for the treatment of human epidermal growth factor receptor 2 (HER2)–positive metastatic breast cancer previously treated with trastuzumab and taxane chemotherapy. It comprises the microtubule inhibitory cytotoxic agent DM1 conjugated to the HER2-targeted humanized monoclonal antibody trastuzumab via a stable linker. To characterize the pharmacokinetics of T-DM1 in patients with metastatic breast cancer, concentrations of multiple analytes were quantified, including serum concentrations of T-DM1 conjugate and total trastuzumab (the sum of conjugated and unconjugated trastuzumab), as well as plasma concentrations of DM1. The clearance of T-DM1 conjugate is approximately 2 to 3 times faster than its parent antibody, trastuzumab. However, the clearance pathways accounting for this faster clearance rate are unclear. An integrated population pharmacokinetic model that simultaneously fits the pharmacokinetics of T-DM1 conjugate and total trastuzumab can help to elucidate the clearance pathways of T-DM1. The model can also be used to predict total trastuzumab pharmacokinetic profiles based on T-DM1 conjugate pharmacokinetic data and sparse total trastuzumab pharmacokinetic data, thereby reducing the frequency of pharmacokinetic sampling.

Methods

T-DM1 conjugate and total trastuzumab serum concentration data, including baseline trastuzumab concentrations prior to T-DM1 treatment, from phase I and II studies were used to develop this integrated population pharmacokinetic model. Based on a hypothetical T-DM1 catabolism scheme, two-compartment models for T-DM1 conjugate and trastuzumab were integrated by assuming a one-step deconjugation clearance from T-DM1 conjugate to trastuzumab. The ability of the model to predict the total trastuzumab pharmacokinetic profile based on T-DM1 conjugate pharmacokinetics and various sampling schemes of total trastuzumab pharmacokinetics was assessed to evaluate total trastuzumab sampling schemes.

Results

The final model reflects a simplified catabolism scheme of T-DM1, suggesting that T-DM1 clearance pathways include both deconjugation and proteolytic degradation. The model fits T-DM1 conjugate and total trastuzumab pharmacokinetic data simultaneously. The deconjugation clearance of T-DM1 was estimated to be ~0.4 L/day. Proteolytic degradation clearances for T-DM1 and trastuzumab were similar (~0.3 L/day). This model accurately predicts total trastuzumab pharmacokinetic profiles based on T-DM1 conjugate pharmacokinetic data and sparse total trastuzumab pharmacokinetic data sampled at preinfusion and end of infusion in cycle 1, and in one additional steady state cycle.

Conclusions

This semi-mechanistic integrated model links T-DM1 conjugate and total trastuzumab pharmacokinetic data, and supports the inclusion of both proteolytic degradation and deconjugation as clearance pathways in the hypothetical T-DM1 catabolism scheme. The model attributes a faster T-DM1 conjugate clearance versus that of trastuzumab to the presence of a deconjugation process and suggests a similar proteolytic clearance of T-DM1 and trastuzumab. Based on the model and T-DM1 conjugate pharmacokinetic data, a sparse pharmacokinetic sampling scheme for total trastuzumab provides an entire pharmacokinetic profile with similar predictive accuracy to that of a dense pharmacokinetic sampling scheme.

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
Fig. 6
Fig. 7
Fig. 8

Similar content being viewed by others

References

  1. Siegel R, Naishadham D, Jemal A. Cancer statistics, 2012. CA Cancer J Clin. 2012; 62(1):10–29.

  2. Sjögren S, Inganäs M, Lindgren A, et al. Prognostic and predictive value of c-erbB-2 overexpression in primary breast cancer, alone and in combination with other prognostic markers. J Clin Oncol. 1998;16(2):462–9.

    PubMed  Google Scholar 

  3. Ross JS, Slodkowska EA, Symmans WF, et al. The HER-2 receptor and breast cancer: ten years of targeted anti-HER-2 therapy and personalized medicine. Oncologist. 2009;14(4):320–68.

    Article  PubMed  CAS  Google Scholar 

  4. Dawood S, Broglio K, Buzdar AU, et al. Prognosis of women with metastatic breast cancer by HER2 status and trastuzumab treatment: an institutional-based review. J Clin Oncol. 2010;28(1):92–8.

    Article  PubMed  CAS  Google Scholar 

  5. Slamon DJ, Clark GM, Wong SG, et al. Human breast cancer: correlation of relapse and survival with amplification of the HER-2/neu oncogene. Science. 1987;235(4785):177–82.

    Article  PubMed  CAS  Google Scholar 

  6. Seshadri R, Firgaira FA, Horsfall DJ, et al. Clinical significance of HER-2/neu oncogene amplification in primary breast cancer. The South Australian Breast Cancer Study Group. J Clin Oncol. 1993;11(10):1936–42.

    PubMed  CAS  Google Scholar 

  7. Gabos Z, Sinha R, Hanson J, et al. Prognostic significance of human epidermal growth factor receptor positivity for the development of brain metastases after newly diagnosed breast cancer. J Clin Oncol. 2006;24(36):5658–63.

    Article  PubMed  CAS  Google Scholar 

  8. Lewis Phillips GD, Li G, Dugger DL, et al. Targeting HER2-positive breast cancer with trastuzumab-DM1, an antibody-cytotoxic drug conjugate. Cancer Res. 2008;68(22):9280–90.

    Google Scholar 

  9. Junttila TT, Li G, Parsons K, et al. Trastuzumab-DM1 (T-DM1) retains all the mechanisms of action of trastuzumab and efficiently inhibits growth of lapatinib insensitive breast cancer. Breast Cancer Res Treat. 2011;128(2):347–56.

    Article  PubMed  CAS  Google Scholar 

  10. Barok M, Tanner M, Köninki K, et al. Trastuzumab-DM1 is highly effective in preclinical models of HER2-positive gastric cancer. Cancer Lett. 2011;306(2):171–9.

    Article  PubMed  CAS  Google Scholar 

  11. Burris HA 3rd, Rugo HS, Vukelja SJ, et al. Phase II study of the antibody drug conjugate trastuzumab-DM1 for the treatment of human epidermal growth factor receptor 2 (HER2)-positive breast cancer after prior HER2-directed therapy. J Clin Oncol. 2011;29(4):398–405.

    Article  PubMed  CAS  Google Scholar 

  12. Krop I, LoRusso P, Miller KD, et al. A phase II study of trastuzumab emtansine in patients with human epidermal growth factor receptor 2-positive metastatic breast cancer who were previously treated with trastuzumab, lapatinib, an anthracycline, a taxane, and capecitabine. J Clin Oncol. 2012;30(26):3234–41.

    Article  PubMed  CAS  Google Scholar 

  13. Hurvitz SA, Dirix L, Kocsis J, et al. A phase 2 randomized study of trastuzumab emtansine (T-DM1) versus trastuzumab + docetaxel in patients with HER2-positive metastatic breast cancer. J Clin Oncol. 2013;31(9):1157–63.

    Google Scholar 

  14. Verma S, Miles D, Gianni L, et al. Trastuzumab emtansine for HER2-positive advanced breast cancer. N Engl J Med. 2012;367(19):1783–91.

    Article  PubMed  CAS  Google Scholar 

  15. Leipold D, Bender B, Xu K, et al. (2009). Understanding the de-conjugation of Trastuzumab-MCC-DM1 through application of a multi-compartmental model of individual drug:antibody species in cynomolgus monkey [abstract no. 2914]. American Association for Cancer Research (AACR) Meeting; 2009 Apr 18; Denver (CO).

  16. Girish S, Gupta M, Wang B, et al. Clinical pharmacology of trastuzumab emtansine (T-DM1): an antibody-drug conjugate in development for the treatment of HER2-positive cancer. Cancer Chemother Pharmacol. 2012;69(5):1229–40.

    Article  PubMed  CAS  Google Scholar 

  17. Gupta M, LoRusso PM, Wang B, et al. Clinical implications of pathophysiological and demographic covariates on the population pharmacokinetics of trastuzumab-emtansine, a HER2-targeted antibody–drug conjugate, in patients with HER2-positive metastatic breast cancer. J Clin Pharmacol. 2012;52(5):691–703.

    Article  PubMed  CAS  Google Scholar 

  18. Bruno R, Washington CB, Lu JF, et al. Population pharmacokinetics of trastuzumab in patients with HER2 + metastatic breast cancer. Cancer Chemother Pharmacol. 2005;56(4):361–9.

    Article  PubMed  CAS  Google Scholar 

  19. Kuester K, Kloft C. Pharmacokinetics of monoclonal antibodies. In: Meibohm B, editor. Pharmacokinetics and pharmacodynamics of biotech drugs. 1st ed. Weinheim (Germany): Wiley-VCH GmbH & Co. KGaA; 2006. p. 45–91.

    Chapter  Google Scholar 

  20. Krop IE, Beeram M, Modi S, et al. Phase I study of trastuzumab-DM1, an HER2 antibody-drug conjugate, given every 3 weeks to patients with HER2-positive metastatic breast cancer. J Clin Oncol. 2010;28(16):2698–704.

    Article  PubMed  CAS  Google Scholar 

  21. Gupta M, Wang B, Carrothers TJ, et al. Effects of trastuzumab emtansine (T-DM1) on QT interval and safety of pertuzumab plus T-DM1 in patients with previously treated human epidermal growth factor receptor 2–positive metastatic breast cancer. Clin Pharmacol Drug Devel. 2013 Feb 26 (E-pub)

  22. Bonate PL. Pharmacokinetic-pharmacodynamic modeling and simulation. 2nd ed. New York: Springer; 2005.

    Google Scholar 

  23. Boeckman AJ, Beal SL, Sheiner LB, Beal. NONMEM User’s Guide—Part IV (online). San Francisco (CA): NONMEM Project Group, University of California at San Francisco, 1992. Available from URL: ftp://nonmem.iconplc.com/Public/nonmem720/guides/iv.pdf. Accessed 2012 Jul 18.

  24. Rowland M, Tozer TN. Clinical pharmacokinetics: concepts and applications. 3rd ed. Philadelphia: Lippincott Williams & Wilkins; 1995.

    Google Scholar 

  25. Hooker AC, Staatz CE, Karlsson MO. Conditional weighted residuals (CWRES): a model diagnostic for the FOCE method. Pharm Res. 2007;24(12):2187–97.

    Article  PubMed  CAS  Google Scholar 

  26. Karlsson MO, Savic RM. Diagnosing model diagnostics. Clin Pharmacol Ther. 2007;82(1):17–20.

    Article  PubMed  CAS  Google Scholar 

  27. Raemer DB, Buschman A, Varvel JR, et al. The prospective use of population pharmacokinetics in a computer-driven infusion system for alfentanil. Anesthesiology. 1990;73(1):66–72.

    Article  PubMed  CAS  Google Scholar 

  28. Varvel JR, Donoho DL, Shafer SL. Measuring the predictive performance of computer-controlled infusion pumps. J Pharmacokinet Biopharm. 1992;20(1):63–94.

    Article  PubMed  CAS  Google Scholar 

  29. Savic RM, Karlsson MO. Importance of shrinkage in empirical bayes estimates for diagnostics: problems and solutions. AAPS J. 2009;11(3):558–69.

    Article  PubMed  Google Scholar 

  30. Fukushima Y, Charoin J-E. Report No. 1034069. Population pharmacokinetic analysis of combined Phase II/III studies: BO15899, BO15935, WO16229, M77004 and MO16982. Basel: Hoffman-La Roche, Inc.;2009. Data on file.

  31. Meibohm B, editor. Pharmacokinetics and pharmacodynamics of biotech drugs: Principles and case studies in drug development. San Francisco: Wiley-VCH GmbH & Co.;2006.

    Google Scholar 

  32. Erickson HK, Widdison WC, Mayo MF, et al. Tumor delivery and in vivo processing of disulfide-linked and thioether-linked antibody-maytansinoid conjugates. Bioconjug Chem. 2010;21(1):84–92.

    Article  PubMed  CAS  Google Scholar 

  33. Shen BQ, Xu K, Liu L, et al. Conjugation site modulates the in vivo stability and therapeutic activity of antibody-drug conjugates. Nat Biotechnol. 2012;30(2):184–9.

    Article  PubMed  CAS  Google Scholar 

  34. Xie H, Audette C, Hoffee M, et al. Pharmacokinetics and biodistribution of the antitumor immunoconjugate, cantuzumab mertansine (huC242-DM1), and its two components in mice. J Pharmacol Exp Ther. 2004;308(3):1073–82.

    Article  PubMed  CAS  Google Scholar 

  35. Lu JF, Bruno R, Eppler S, et al. Clinical pharmacokinetics of bevacizumab in patients with solid tumors. Cancer Chemother Pharmacol. 2008;62(5):779–86.

    Article  PubMed  CAS  Google Scholar 

  36. Ng CM, Lum BL, Gimenez V, et al. Rationale for fixed dosing of pertuzumab in cancer patients based on population pharmacokinetic analysis. Pharm Res. 2006;23(6):1275–84.

    Article  PubMed  CAS  Google Scholar 

  37. Li J, Zhi J, Wenger M, et al. Population pharmacokinetics of rituximab in patients with chronic lymphocytic leukemia. J Clin Pharmacol. 2012;52(12):1918–26.

    Article  PubMed  CAS  Google Scholar 

  38. Lobo ED, Hansen RJ, Blathasar JP. Antibody pharmacokinetics and pharmacodynamics. J Pharm Sci. 2004;93(11):2645–68.

    Article  PubMed  CAS  Google Scholar 

  39. Garzone PD, Atkinson AJ Jr. In search of physiologically based distribution volume estimates for macromolecules. Clin Pharmacol Ther. 2012;92(4):419–21.

    Article  PubMed  CAS  Google Scholar 

  40. Chudasama VL, Schaedeli Stark F, Harrold JM, et al. Semi-mechanistic population pharmacokinetic model of multivalent trastuzumab emtansine in patients with metastatic breast cancer. Clin Pharmacol Ther. 2012;92(4):520–7.

    Article  PubMed  CAS  Google Scholar 

Download references

Acknowledgments

The authors acknowledge the investigators, patients and their families who participated in these clinical trials. The authors acknowledge Jin Yan Jin, PhD, for scientific input and helpful discussions during the preparation of this report. Support for third-party editorial assistance for this manuscript, furnished by Denise Chun, Ph.D. (CodonMedical), was provided by Genentech, Inc. The clinical studies reported here were sponsored by Genentech, Inc. Dan Lu, Amita Joshi, Bei Wang, Steve Olsen, Joo-Hee Yi and Sandhya Girish are employees of Genentech, Inc., and have Roche stock/stock options. Ian E. Krop receives clinical trial support from Genentech, Inc. Howard A. Burris has no conflicts of interest to declare.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Dan Lu.

Electronic supplementary material

Below is the link to the electronic supplementary material.

Supplementary material 1 (PDF 74 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Lu, D., Joshi, A., Wang, B. et al. An Integrated Multiple-Analyte Pharmacokinetic Model to Characterize Trastuzumab Emtansine (T-DM1) Clearance Pathways and to Evaluate Reduced Pharmacokinetic Sampling in Patients with HER2-Positive Metastatic Breast Cancer. Clin Pharmacokinet 52, 657–672 (2013). https://doi.org/10.1007/s40262-013-0060-y

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s40262-013-0060-y

Keywords

Navigation