Pharmacokinetics, immunogenicity and bioactivity of the therapeutic antibody catumaxomab intraperitoneally administered to cancer patients

Peter Ruf, Michael Kluge, Michael Jäger, Alexander Burges, Constantin Volovat, Markus Maria Heiss, Jürgen Hess, Pauline Wimberger, Birgit Brandt, Horst Lindhofer, Peter Ruf, Michael Kluge, Michael Jäger, Alexander Burges, Constantin Volovat, Markus Maria Heiss, Jürgen Hess, Pauline Wimberger, Birgit Brandt, Horst Lindhofer

Abstract

Aims: Catumaxomab is the first EMEA approved trifunctional anti-EpCAMxanti-CD3 antibody for the treatment of cancer patients with malignant ascites. A phase II pharmacokinetic study was conducted to determine local and systemic antibody concentrations and anti-drug antibody (ADA) development.

Methods: Thirteen cancer patients with symptomatic malignant ascites were treated with four ascending doses of 10, 20, 50, and 150 microg catumaxomab intraperitoneally (i.p.) infused on days 0, 3, 6 or 7 and 10. The pharmacokinetics of catumaxomab were studied by implementation of supportive data from a non clinical mouse tumour model. Additionally, ADA development was monitored.

Results: Ten out of 13 patients were evaluable for pharmacokinetic analysis. Catumaxomab became increasingly concentrated in ascites during the course of treatment, attaining effective concentrations in the ng ml(-1) range. Catumaxomab remained immunologically active even after several days in the circulation. The observed systemic catumaxomab exposure was low (<1%), with a maximal median plasma concentration (C(max)) of 403 pg ml(-1). The mean elimination half-life in the plasma was 2.13 days. All patients developed ADA, but not before the last infusion. High observed inter-individual variability and low systemic exposure may be explained by the inverse correlation between tumour burden, effector cell numbers and systemic antibody bioavailability as demonstrated in a defined mouse tumour model.

Conclusions: Based on the high and effective local concentrations, low systemic exposure and acceptable safety profile, we confirmed that the i.p. application scheme of catumaxomab for the treatment of malignant ascites is appropriate.

Keywords: catumaxomab; immunogenicity; intraperitoneal infusion; malignant ascites; pharmacokinetics.

Figures

Figure 1
Figure 1
Individual (symbols) and median (–) catumaxomab ascites concentrations observed during the course of treatment. Intraperitoneal infusions are indicated by arrows. The third infusion (50 µg) was administered either on day 6 or 7. Ascites samples were taken immediately before the second, third, and fourth infusions. 108/102 (); 118/101 (); 805/101 (); 805/102 (); 805/103 (); 805/107 (); 805/108 (); 805/112 (); 805/113 (); 808/101 (); Median ()
Figure 2
Figure 2
Individual catumaxomab plasma concentration vs. time profiles in 10 patients. Intraperitoneal infusions of catumaxomab are indicated by arrows. The third (50 µg) infusion was administered either on day 6 or 7. Catumaxomab was detectable in the plasma only after the third and fourth infusions. 108/102 (); 118/101 (); 805/101 (); 805/102 (); 805/103 (); 805/107 (); 805/108 (); 805/112 (); 805/113 (); 808/101 ()
Figure 3
Figure 3
Bioactivity of catumaxomab in ascites and plasma samples. Bioactivity was determined in a potency assay that evaluated ascites and plasma samples for the abilities to A) kill EpCAM-positive HCT-8 tumour cells and B) secrete TNF-α cytokine, relative to controls with freshly spiked catumaxomab. Ascites samples were obtained before the third or fourth infusion from five patients, and two plasma samples were obtained from one patient
Figure 4
Figure 4
Anti-drug antibody response in plasma vs. time profiles in 10 patients within the first 3 weeks after the beginning of the treatment. 108/102 (); 118/101 (); 805/101 (); 805/102 (); 805/103 (); 805/107 (); 805/108 (); 805/112 (); 805/113 (); 808/101 ()

References

    1. Zeidler R, Reisbach G, Wollenberg B, Lang S, Chaubal S, Schmitt B, Lindhofer H. Simultaneous activation of T cells and accessory cells by a new class of intact bispecific antibody results in efficient tumor cell killing. J Immunol. 1999;163:1246–52.
    1. Zeidler R, Mysliwietz J, Csanady M, Walz A, Ziegler I, Schmitt B, Wollenberg B, Lindhofer H. The Fc-region of a new class of intact bispecific antibody mediates activation of accessory cells and NK cells and induces direct phagocytosis of tumour cells. Br J Cancer. 2000;83:261–6.
    1. Shen J, Zhu Z. Catumaxomab, a rat/murine hybrid trifunctional bispecific monoclonal antibody for the treatment of cancer. Curr Opin Mol Ther. 2008;10:273–84.
    1. Ruf P, Lindhofer H. Induction of a long-lasting antitumor immunity by a trifunctional bispecific antibody. Blood. 2001;98:2526–34.
    1. Ruf P, Gires O, Jager M, Fellinger K, Atz J, Lindhofer H. Characterisation of the new EpCAM-specific antibody HO-3: implications for trifunctional antibody immunotherapy of cancer. Br J Cancer. 2007;97:315–21.
    1. Heiss MM, Strohlein MA, Jager M, Kimmig R, Burges A, Schoberth A, Jauch KW, Schildberg FW, Lindhofer H. Immunotherapy of malignant ascites with trifunctional antibodies. Int J Cancer. 2005;117:435–43.
    1. Parsons SL, Watson SA, Steele RJ. Malignant ascites. Br J Surg. 1996;83:6–14.
    1. Ayantunde AA, Parsons SL. Pattern and prognostic factors in patients with malignant ascites: a retrospective study. Ann Oncol. 2007;18:945–9.
    1. az-Arias AA, Loy TS, Bickel JT, Chapman RK. Utility of BER-EP4 in the diagnosis of adenocarcinoma in effusions: an immunocytochemical study of 232 cases. Diagn Cytopathol. 1993;9:516–21.
    1. De AM, Buley ID, Heryet A, Gray W. Immunocytochemical staining of serous effusions with the monoclonal antibody Ber-EP4. Cytopathology. 1992;3:111–7.
    1. Passebosc-Faure K, Li G, Lambert C, Cottier M, Gentil-Perret A, Fournel P, Perol M, Genin C. Evaluation of a panel of molecular markers for the diagnosis of malignant serous effusions. Clin Cancer Res. 2005;11:6862–7.
    1. Burges A, Wimberger P, Kumper C, Gorbounova V, Sommer H, Schmalfeldt B, Pfisterer J, Lichinitser M, Makhson A, Moiseyenko V, Lahr A, Schulze E, Jager M, Strohlein MA, Heiss MM, Gottwald T, Lindhofer H, Kimmig R. Effective relief of malignant ascites in patients with advanced ovarian cancer by a trifunctional anti-EpCAM x anti-CD3 antibody: a phase I/II study. Clin Cancer Res. 2007;13:3899–905.
    1. Sebastian M, Kiewe P, Schuette W, Brust D, Peschel C, Schneller F, Rühle KH, Nilius G, Ewert R, Lodziewski S, Passlick B, Sienel W, Wiewrodt R, Jäger M, Lindhofer H, Friccius-Quecke H, Schmittel A. Treatment of malignant pleural effusions with the trifunctional antibody catumaxomab (removab) (anti-EpCAM × anti-CD3): results of a phase 1/2 study. J Immunother. 2009;32:195–202.
    1. Parsons S, Murawa PX, Koralewski P, Kutarska E, Kolesnik OO, Stroehlein MA, Lahr A, Jaeger M, Heiss M. Intraperitoneal treatment of malignant ascites due to epithelial tumors with catumaxomab: a phase II/III study. J Clin Oncol. 2008;26(Suppl.) abstr 3000.
    1. Becker G, Galandi D, Blum HE. Malignant ascites: systematic review and guideline for treatment. Eur J Cancer. 2006;42:589–97.
    1. Went PT, Lugli A, Meier S, Bundi M, Mirlacher M, Sauter G, Dirnhofer S. Frequent EpCam protein expression in human carcinomas. Hum Pathol. 2004;35:122–8.
    1. Stroehlein MA, Gruetzner KU, Tarabichi A, Jauch KW, Bartelheim K, Lindhofer H, Von Roemeling R, Heiss MM. Efficacy of intraperitoneal treatment with the trifunctional antibody catumaxomab in patients with GI-tract cancer and peritoneal carcinomatosis: a matched-pair analysis. J Clin Oncol. 2006;24:2544. 2006 ASCO Annual Meeting Proceedings Part I. Vol 24, No. 18S (June 20 Supplement)
    1. Lobo ED, Hansen RJ, Balthasar JP. Antibody pharmacokinetics and pharmacodynamics. J Pharm Sci. 2004;93:2645–68.
    1. Frodin JE, Lefvert AK, Mellstedt H. Pharmacokinetics of the mouse monoclonal antibody 17-1A in cancer patients receiving various treatment schedules. Cancer Res. 1990;50:4866–71.
    1. Ternant D, Paintaud G. Pharmacokinetics and concentration-effect relationships of therapeutic monoclonal antibodies and fusion proteins. Expert Opin Biol Ther. 2005;5(Suppl 1):S37–47.
    1. Mobus VJ, Baum RP, Bolle M, Kreienberg R, Noujaim AA, Schultes BC, Nicodemus CF. Immune responses to murine monoclonal antibody-B43.13 correlate with prolonged survival of women with recurrent ovarian cancer. Am J Obstet Gynecol. 2003;189:28–36.
    1. Sebastian M, Passlick B, Friccius-Quecke H, Jager M, Lindhofer H, Kanniess F, Wiewrodt R, Thiel E, Buhl R, Schmittel A. Treatment of non-small cell lung cancer patients with the trifunctional monoclonal antibody catumaxomab (anti-EpCAM x anti-CD3): a phase I study. Cancer Immunol Immunother. 2007;56:1637–44.
    1. Davis TA, White CA, Grillo-Lopez AJ, Velasquez WS, Link B, Maloney DG, Dillman RO, Williams ME, Mohrbacher A, Weaver R, Dowden S, Levy R. Single-agent monoclonal antibody efficacy in bulky non-Hodgkin's lymphoma: results of a phase II trial of rituximab. J Clin Oncol. 1999;17:1851–7.
    1. Hale G, Rebello P, Brettman LR, Fegan C, Kennedy B, Kimby E, Leach M, Lundin J, Mellstedt H, Moreton P, Rawstron AC, Waldmann H, Osterborg A, Hillmen P. Blood concentrations of alemtuzumab and antiglobulin responses in patients with chronic lymphocytic leukemia following intravenous or subcutaneous routes of administration. Blood. 2004;104:948–55.
    1. Dayde D, Ternant D, Ohresser M, Lerondel S, Pesnel S, Watier H, Le PA, Bardos P, Paintaud G, Cartron G. Tumor burden influences exposure and response to rituximab: pharmacokinetic–pharmacodynamic modelling using a syngeneic bioluminescent murine model expressing human CD20. Blood. 2008;113:3765–72.

Source: PubMed

3
Iratkozz fel