The trifunctional antibody catumaxomab for the treatment of malignant ascites due to epithelial cancer: Results of a prospective randomized phase II/III trial

Markus M Heiss, Pawel Murawa, Piotr Koralewski, Elzbieta Kutarska, Olena O Kolesnik, Vladimir V Ivanchenko, Alexander S Dudnichenko, Birute Aleknaviciene, Arturas Razbadauskas, Martin Gore, Elena Ganea-Motan, Tudor Ciuleanu, Pauline Wimberger, Alexander Schmittel, Barbara Schmalfeldt, Alexander Burges, Carsten Bokemeyer, Horst Lindhofer, Angelika Lahr, Simon L Parsons, Markus M Heiss, Pawel Murawa, Piotr Koralewski, Elzbieta Kutarska, Olena O Kolesnik, Vladimir V Ivanchenko, Alexander S Dudnichenko, Birute Aleknaviciene, Arturas Razbadauskas, Martin Gore, Elena Ganea-Motan, Tudor Ciuleanu, Pauline Wimberger, Alexander Schmittel, Barbara Schmalfeldt, Alexander Burges, Carsten Bokemeyer, Horst Lindhofer, Angelika Lahr, Simon L Parsons

Abstract

Malignant ascites is a common manifestation of advanced cancers, and treatment options are limited. The trifunctional antibody catumaxomab (anti-epithelial cell-adhesion molecule x anti-CD3) represents a targeted immunotherapy for the intraperitoneal (i.p.) treatment of malignant ascites secondary to epithelial cancers. In this phase II/III trial (EudraCT 2004-000723-15; NCT00836654), cancer patients (n = 258) with recurrent symptomatic malignant ascites resistant to conventional chemotherapy were randomized to paracentesis plus catumaxomab (catumaxomab) or paracentesis alone (control) and stratified by cancer type (129 ovarian and 129 nonovarian). Catumaxomab was administered as an i.p. infusion on Days 0, 3, 7 and 10 at doses of 10, 20, 50 and 150 mug, respectively. The primary efficacy endpoint was puncture-free survival. Secondary efficacy parameters included time to next paracentesis, ascites signs and symptoms and overall survival (OS). Puncture-free survival was significantly longer in the catumaxomab group (median 46 days) than the control group (median 11 days) (hazard ratio = 0.254: p < 0.0001) as was median time to next paracentesis (77 versus 13 days; p < 0.0001). In addition, catumaxomab patients had fewer signs and symptoms of ascites than control patients. OS showed a positive trend for the catumaxomab group and, in a prospectively planned analysis, was significantly prolonged in patients with gastric cancer (n = 66; 71 versus 44 days; p = 0.0313). Although adverse events associated with catumaxomab were frequent, they were manageable, generally reversible and mainly related to its immunologic mode of action. Catumaxomab showed a clear clinical benefit in patients with malignant ascites secondary to epithelial cancers, especially gastric cancer, with an acceptable safety profile.

Figures

Figure 1
Figure 1
(a) Study design. (b) CONSORT flow diagram (only results for the intent-to-treat and safety populations are included in this article). *The main cancer types in the nonovarian stratum were gastric (n = 66, 51%), breast (n = 13, 10%), pancreas (n = 9, 7%), colon (n = 8, 6%) and endometrial (n = 6, 5%).
Figure 2
Figure 2
Kaplan-Meier estimates of puncture-free survival, time to next paracentesis and overall survival (intent-to-treat population). (a) Puncture-free survival in the pooled population; (b) time to next paracentesis in the pooled population; (c) overall survival in the pooled population; (d) overall survival in patients with ovarian cancer and (e) overall survival in patients with gastric cancer.
Figure 3
Figure 3
Patients without ascites signs and symptoms as assessed by interview 8 days after last infusion (catumaxomab) or 8 days after paracentesis (control group).
Figure 4
Figure 4
(a) Tumor-cell load in the ascites fluid before, during and after catumaxomab treatment. Number of EpCAM+ tumor cells per 106 total cells evaluated by immunocytochemistry. The median tumor-cell count at the repuncture visit was statistically significantly lower (p = 0.0012) in the catumaxomab group. (b) Fluorescent double staining of ascites fluid cells: evaluation of CD45+ leukocyte:EpCAM+ tumor cell ratio during catumaxomab therapy. (i) Malignant ascites harvested at screening puncture (before catumaxomab treatment). (ii) At Day 3 after 10 μg catumaxomab i.p. (iii) At Day 11 after a total of 230 μg catumaxomab i.p. Dotplot analysis: every plot resembles a fluorescence-labeled cell that was detected and counted by the computerized image analysis system.

References

    1. Parsons SL, Watson SA, Steele RJC. Malignant ascites. Br J Surg. 1996;83:6–14.
    1. Muir JC. Ascites. In: Von Roenn J, Smith TJ, Loprinzi CL, Von Gunten CF, editors. ASCO curriculum: optimizing cancer care. The importance of symptom management. Vol. 1. Dubuque, IA: Kendall/Hunt; 2001. pp. 1–31.
    1. Ayantunde AA, Parsons S. Pattern and prognostic factors in patients with malignant ascites: a retrospective study. Ann Oncol. 2007;18:945–9.
    1. Tamsma J. The pathogenesis of malignant ascites. Cancer Treat Res. 2007;134:109–18.
    1. Preston N. New strategies for the management of malignant ascites. Eur J Cancer Care. 1995;4:178–83.
    1. Hostetter RB, Marincola FM, Schwarzentruber DJ. Malignant ascites. In: deVita VT, Hellman S, Rosenberg SA, editors. Cancer: principles and practice of oncology. 7th edn. Philadelphia: Lippincott-Williams & Wilkins; 2005. pp. 2392–8.
    1. Becker G, Galandi D, Blum HE. Malignant ascites: systematic review and guideline for treatment. Eur J Cancer. 2006;42:589–97.
    1. Garrison RN, Kaelin LD, Galloway RH, Heuser LS. Malignant ascites. Clinical and experimental observations. Ann Surg. 1986;203:644–51.
    1. Ayhan A, Gultekin M, Taskiran C, Dursun P, Firat P, Bozdag G, Celik NY, Yuce K. Ascites and epithelial ovarian cancers: a reappraisal with respect to different aspects. Int J Gynecol Cancer. 2007;17:68–75.
    1. Lee CW, Bociek G, Faught W. A survey of practice in the management of malignant ascites. J Pain Symptom Manage. 1998;16:96–101.
    1. Smith EM, Jayson GC. The current and future management of malignant ascites. Clin Oncol (R Coll Radiol) 2003;15:59–72.
    1. Ruf P, Lindhofer H. Induction of a long-lasting antitumor immunity by a trifunctional bispecific antibody. Blood. 2001;98:2526–34.
    1. Zeidler R, Mysliwietz J, Csánady 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. Ruf P, Gires O, Jäger 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. 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. Passebosc-Faure K, Li G, Lambert C, Cottier M, Gentil-Perret A, Fournel P, Pérol 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. De Angelis M, Buley ID, Heryet A, Gray W. Immunocytochemical staining of serous effusions with the monoclonal antibody Ber-EP4. Cytopathology. 1992;3:111–7.
    1. Diaz-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. McLaughlin PM, Harmsen MC, Dokter WH, Kroesen BJ, van der Molen H, Brinker MG, Hollema H, Ruiters MH, Buys CH, de Leij LF. The epithelial glycoprotein 2 (EGP-2) promoter-driven epithelial-specific expression of EGP-2 in transgenic mice: a new model to study carcinoma-directed immunotherapy. Cancer Res. 2001;61:4105–11.
    1. Okamoto S, Ito K, Sasano H, Moriya T, Niikura H, Terada Y, Sato S, Okamura K, Yaegashi N. Ber-EP4 and anti-calretinin antibodies: a useful combination for differential diagnosis of various histological types of ovarian cancer cells and mesothelial cells. Tohoku J Exp Med. 2005;206:31–40.
    1. Heiss MM, Ströhlein MA, Jäger 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. Burges A, Wimberger P, Kümper C, Gorbounova V, Sommer H, Schmalfeldt B, Pfisterer J, Lichinitser M, Makhson A, Moiseyenko V, Lahr A, Schulze E, et al. 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. Likert R. A technique for the measurement of attitudes. Arch Psychol. 1932;140:1–55.
    1. Sartori S, Nielsen I, Tassinari D, Trevisani L, Abbasciano V, Malacarne P. Evaluation of a standardized protocol of intracavitary recombinant interferon alpha-2b in the palliative treatment of malignant peritoneal effusions. A prospective pilot study. Oncology. 2001;61:192–6.
    1. Stuart GC, Nation JG, Snider DD, Thunberg P. Intraperitoneal interferon in the management of malignant ascites. Cancer. 1993;71:2027–30.
    1. Hirte HW, Miller D, Tonkin K, Findlay B, Capstick V, Murphy J, Buckman R, Carmichael J, Levine M, Hill W. A randomized trial of paracentesis plus intraperitoneal tumor necrosis factor-alpha versus paracentesis alone in patients with symptomatic ascites from recurrent ovarian carcinoma. Gynecol Oncol. 1997;64:80–7.
    1. Räth U, Kaufmann M, Schmid H, Hofmann J, Wiedenmann B, Kist A, Kempeni J, Schlick E, Bastert G, Kommerell B, Männel D. Effect of intraperitoneal recombinant human tumour necrosis factor alpha on malignant ascites. Eur J Cancer. 1991;27:121–5.
    1. Parsons SL, Watson SA, Steele RJ. Phase I/II trial of batimastat, a matrix metalloproteinase inhibitor, in patients with malignant ascites. Eur J Surg Oncol. 1997;23:526–31.
    1. Katano M, Morisaki T. The past, the present and future of the OK-432 therapy for patients with malignant effusions. Anticancer Res. 1998;18:3917–25.
    1. Hamilton CA, Maxwell GL, Chernofsky MR, Bernstein SA, Farley JH, Rose GS. Intraperitoneal bevacizumab for the palliation of malignant ascites in refractory ovarian cancer. Gynecol Oncol. 2008;111:530–2.
    1. Kesterson JP, Mhawech-Fauceglia P, Lele S. The use of bevacizumab in refractory ovarian granulosa-cell carcinoma with symptomatic relief of ascites: a case report. Gynecol Oncol. 2008;111:527–9.
    1. Marmé A, Strauss G, Bastert G, Grischke EM, Moldenhauer G. Intraperitoneal bispecific antibody (HEA125xOKT3) therapy inhibits malignant ascites production in advanced ovarian carcinoma. Int J Cancer. 2002;101:183–9.
    1. Armstrong DK, Bundy B, Wenzel L, Huang HQ, Baergen R, Lele S, Copeland LJ, Walker JL, Burger RA, Gynecologic OncologyGroup Intraperitoneal cisplatin and paclitaxel in ovarian cancer. N Engl J Med. 2006;354:34–43.
    1. Jeyarajah DR, Thistlethwaite JR., Jr General aspects of cytokine-release syndrome: timing and incidence of symptoms. Transplant Proc. 1993;25:16–20.
    1. Winkler U, Jensen M, Manzke O, Schulz H, Diehl V, Engert A. Cytokine-release syndrome in patients with B-cell chronic lymphocytic leukemia and high lymphocyte counts after treatment with an anti-CD20 monoclonal antibody (rituximab. IDEC-C2B8) Blood. 1999;94:2217–24.
    1. Bokemeyer C, Heiss M, Gamperl H, Linke R, Schulze E, Friccius-Quecke H, Lindhofer H, Parsons S. Safety of catumaxomab: cytokine-release-related symptoms as a possible predictive factor for efficacy in a pivotal phase II/III trial in malignant ascites. J Clin Oncol. 2009;27(Suppl) Abstract 3036.
    1. Prandota J. Important role of proinflammatory cytokines/other endogenous substances in drug-induced hepatotoxicity: depression of drug metabolism during infections/inflammation states, and genetic polymorphisms of drug-metabolizing enzymes/cytokines may markedly contribute to this pathology. Am J Ther. 2005;12:254–61.
    1. Molema G, Cohen-Tervaer JW, Kroesen BJ, Helfrich W, Meijer DK, de Leij LF. CD3 directed bispecific antibodies induce increased lymphocyte-endothelia l cell interactions in vitro. Br J Cancer. 2000;82:472–9.
    1. DeNardo GL, Mirick GR, Kroger LA, Bradt BM, Lamborn KR, DeNardo SJ. Characterization of human IgG antimouse antibody in patients with B-cell malignancies. Clin Cancer Res. 2003;9:4013–4021S.
    1. Miotti S, Negri DR, Valota O, Calabrese M, Bolhuis RL, Gratama JW, Colnaghi MI, Canevari S. Level of anti-mouse-antibody response induced by bi-specific monoclonal antibody OC/TR in ovarian-carcinoma patients is associated with longer survival. Int J Cancer. 1999;84:62–8.

Source: PubMed

3
Prenumerera