Skip to main content
Erschienen in: Cancer Chemotherapy and Pharmacology 5/2015

01.05.2015 | Original Article

A phase I trial of intravenous catumaxomab: a bispecific monoclonal antibody targeting EpCAM and the T cell coreceptor CD3

verfasst von: Morten Mau-Sørensen, Christian Dittrich, Rodrigo Dienstmann, Ulrik Lassen, Wilfried Büchler, Holger Martinius, Josep Tabernero

Erschienen in: Cancer Chemotherapy and Pharmacology | Ausgabe 5/2015

Einloggen, um Zugang zu erhalten

Abstract

Purpose

The aim of the study was to evaluate the safety and determine the maximum tolerated dose (MTD) of intravenous catumaxomab, a trifunctional bispecific antibody that binds to EpCAM on epithelial cancer cells and CD3 on T cells.

Methods

The trial was a dose-escalation study with a 3 + 3 design in epithelial cancers with known EpCAM expression. The dose-limiting toxicity (DLT) period consisted of 4 weeks, with weekly intravenous administration of catumaxomab. Key DLTs were ≥grade 3 optimally treated non-hematological toxicity; ≥grade 3 infusion-related reactions refractory to supportive care; ≥grade 3 increase in liver enzymes and/or bilirubin not resolving to grade 2.

Results

Sixteen patients were enrolled receiving doses of 2 (n = 5), 4 (n = 3), 7 (n = 7) and 10 µg catumaxomab (n = 1). The most common treatment-emergent adverse events (TEAEs) were chills (93.8 %) and pyrexia (87.5 %). The most common TEAE of grade ≥3 was transient dose-dependent increases in aspartate aminotransferase (56.3 %). The intensity of toxicities decreased with the number of infusions. Also, serum IL-6 increased in a dose-dependent manner and reverted to low or undetectable levels after four infusions. A reversible decrease in liver function test (prothrombin time) at the 7-µg dose level was considered a DLT. The first patient at 10 µg experienced a fatal hepatic failure related to catumaxomab that led to the termination of the study.

Conclusions

The MTD of weekly intravenous catumaxomab was 7 µg. Major toxicities were cytokine release-related symptoms and hepatotoxicity.
Literatur
1.
Zurück zum Zitat Went P, Lugli A, Meier S et al (2004) Frequent EpCAM protein expression in human carcinomas. Hum Pathol 35:122–128CrossRefPubMed Went P, Lugli A, Meier S et al (2004) Frequent EpCAM protein expression in human carcinomas. Hum Pathol 35:122–128CrossRefPubMed
2.
Zurück zum Zitat Went P, Vasei M, Bubendorf L et al (2006) Frequent high-level expression of the immunotherapeutic target EpCAM in colon, stomach, prostate and lung cancers. Br J Cancer 94:128–135CrossRefPubMedCentralPubMed Went P, Vasei M, Bubendorf L et al (2006) Frequent high-level expression of the immunotherapeutic target EpCAM in colon, stomach, prostate and lung cancers. Br J Cancer 94:128–135CrossRefPubMedCentralPubMed
3.
Zurück zum Zitat Spizzo G, Went P, Dirnhofer S et al (2004) High EpCAM expression is associated with poor prognosis in node-positive breast cancer. Breast Cancer Res Treat 86:207–213CrossRefPubMed Spizzo G, Went P, Dirnhofer S et al (2004) High EpCAM expression is associated with poor prognosis in node-positive breast cancer. Breast Cancer Res Treat 86:207–213CrossRefPubMed
4.
Zurück zum Zitat Spizzo G, Went P, Dirnhofer S et al (2006) Overexpression of epithelial cell adhesion molecule (EpCAM) is an independent prognostic marker for reduced survival of patients with epithelial ovarian cancer. Gynecol Oncol 103:483–488CrossRefPubMed Spizzo G, Went P, Dirnhofer S et al (2006) Overexpression of epithelial cell adhesion molecule (EpCAM) is an independent prognostic marker for reduced survival of patients with epithelial ovarian cancer. Gynecol Oncol 103:483–488CrossRefPubMed
5.
Zurück zum Zitat Balzar M, Winter MJ, de Boer CJ et al (1999) The biology of the 17-1A antigen (EpCAM). J Mol Mod 77:699–712CrossRef Balzar M, Winter MJ, de Boer CJ et al (1999) The biology of the 17-1A antigen (EpCAM). J Mol Mod 77:699–712CrossRef
6.
Zurück zum Zitat Pauli C, Muenz M, Kieu C et al (2003) Tumor-specific glycosylation of the carcinoma-associated epithelial cell adhesion molecule EpCAM in head and neck carcinomas. Cancer Lett 193:25–32CrossRefPubMed Pauli C, Muenz M, Kieu C et al (2003) Tumor-specific glycosylation of the carcinoma-associated epithelial cell adhesion molecule EpCAM in head and neck carcinomas. Cancer Lett 193:25–32CrossRefPubMed
7.
Zurück zum Zitat Lindhofer H, Mocikat R, Steipe B et al (1995) Preferential species-restricted heavy/light chain pairing in rat/mouse quadromas. Implications for a single-step purification of bispecific antibodies. J Immunol 155:219–225PubMed Lindhofer H, Mocikat R, Steipe B et al (1995) Preferential species-restricted heavy/light chain pairing in rat/mouse quadromas. Implications for a single-step purification of bispecific antibodies. J Immunol 155:219–225PubMed
8.
Zurück zum Zitat Zeidler R, Reisbach G, Wollenberg B et al (1999) Simultaneous activation of T cells and accessory cells by a new class of intact bispecific antibody results in efficient tumor cell killing. J Immunol 163:1246–1252PubMed Zeidler R, Reisbach G, Wollenberg B et al (1999) Simultaneous activation of T cells and accessory cells by a new class of intact bispecific antibody results in efficient tumor cell killing. J Immunol 163:1246–1252PubMed
9.
Zurück zum Zitat Zhang S, Zhang HS, Cordon-Cardo C et al (1998) Selection of tumor antigens as targets for immune attack using immunohistochemistry: protein antigens. Clin Cancer Res 4:2669–2676PubMed Zhang S, Zhang HS, Cordon-Cardo C et al (1998) Selection of tumor antigens as targets for immune attack using immunohistochemistry: protein antigens. Clin Cancer Res 4:2669–2676PubMed
10.
Zurück zum Zitat Riesenberg R, Buchner A, Pohla H et al (2001) Lysis of prostate carcinoma cells by trifunctional bispecific antibodies (alpha EpCAM x alpha CD3). J Histochem Cytochem 49:911–917CrossRefPubMed Riesenberg R, Buchner A, Pohla H et al (2001) Lysis of prostate carcinoma cells by trifunctional bispecific antibodies (alpha EpCAM x alpha CD3). J Histochem Cytochem 49:911–917CrossRefPubMed
11.
Zurück zum Zitat Zeidler R, Mysliwietz J, Csanady M et al (2000) 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 83:261–266CrossRefPubMedCentralPubMed Zeidler R, Mysliwietz J, Csanady M et al (2000) 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 83:261–266CrossRefPubMedCentralPubMed
12.
Zurück zum Zitat Schmitt M, Schmitt A, Reinhardt P et al (2004) Opsonization with a trifunctional bispecific (alphaCD3 x alphaEpCAM) antibody results in efficient lysis in vitro and in vivo of EpCAM positive tumor cells by cytotoxic T lymphocytes. Int J Oncol 25:841–848PubMed Schmitt M, Schmitt A, Reinhardt P et al (2004) Opsonization with a trifunctional bispecific (alphaCD3 x alphaEpCAM) antibody results in efficient lysis in vitro and in vivo of EpCAM positive tumor cells by cytotoxic T lymphocytes. Int J Oncol 25:841–848PubMed
13.
Zurück zum Zitat Seimetz D, Lindhofer H, Bokemeyer C (2010) Development and approval of the trifunctional antibody catumaxomab (anti-EpCAM x anti-CD3) as a targeted cancer immunotherapy. Cancer Treat Rev 36:458–467CrossRefPubMed Seimetz D, Lindhofer H, Bokemeyer C (2010) Development and approval of the trifunctional antibody catumaxomab (anti-EpCAM x anti-CD3) as a targeted cancer immunotherapy. Cancer Treat Rev 36:458–467CrossRefPubMed
14.
Zurück zum Zitat Heiss MM, Murawa P, Koralewski P et al (2010) The trifunctional antibody catumaxomab for the treatment of malignant ascites due to epithelial cancer: results of a prospective randomized phase II/III trial. Int J Cancer 127:2209–2221CrossRefPubMedCentralPubMed Heiss MM, Murawa P, Koralewski P et al (2010) The trifunctional antibody catumaxomab for the treatment of malignant ascites due to epithelial cancer: results of a prospective randomized phase II/III trial. Int J Cancer 127:2209–2221CrossRefPubMedCentralPubMed
15.
Zurück zum Zitat Petrelli F, Borgonovo K, Lonati V et al (2013) Regression of liver metastases after treatment with intraperitoneal catumaxomab for malignant ascites due to breast cancer. Target Oncol 8:291–294CrossRefPubMed Petrelli F, Borgonovo K, Lonati V et al (2013) Regression of liver metastases after treatment with intraperitoneal catumaxomab for malignant ascites due to breast cancer. Target Oncol 8:291–294CrossRefPubMed
16.
Zurück zum Zitat Bezan A, Hohla F, Meissnitzer T et al (2013) Systemic effect of catumaxomab in a patient with metastasized colorectal cancer: a case report. BMC Cancer 13:618CrossRefPubMedCentralPubMed Bezan A, Hohla F, Meissnitzer T et al (2013) Systemic effect of catumaxomab in a patient with metastasized colorectal cancer: a case report. BMC Cancer 13:618CrossRefPubMedCentralPubMed
17.
Zurück zum Zitat Sebastian M, Passlick B, Friccius-Quecke H et al (2007) 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 56:1637–1644CrossRefPubMed Sebastian M, Passlick B, Friccius-Quecke H et al (2007) 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 56:1637–1644CrossRefPubMed
19.
Zurück zum Zitat Ruf P, Kluge M, Jäger M et al (2010) Pharmacokinetics, immunogenicity and bioactivity of the therapeutic antibody catumaxomab intraperitoneally administered to cancer patients. Br J Clin Pharmacol 69:617–625CrossRefPubMedCentralPubMed Ruf P, Kluge M, Jäger M et al (2010) Pharmacokinetics, immunogenicity and bioactivity of the therapeutic antibody catumaxomab intraperitoneally administered to cancer patients. Br J Clin Pharmacol 69:617–625CrossRefPubMedCentralPubMed
20.
Zurück zum Zitat Amacher DE (1998) Serum transaminase elevations as indicators of hepatic injury following the administration of drugs. Regul Toxicol Pharmacol 27:119–130CrossRefPubMed Amacher DE (1998) Serum transaminase elevations as indicators of hepatic injury following the administration of drugs. Regul Toxicol Pharmacol 27:119–130CrossRefPubMed
21.
Zurück zum Zitat Suntharalingam G, Perry MR, Ward S et al (2006) Cytokine storm in a phase 1 trial of the anti-CD28 monoclonal antibody TGN1412. N Engl J Med 355:1018–1028CrossRefPubMed Suntharalingam G, Perry MR, Ward S et al (2006) Cytokine storm in a phase 1 trial of the anti-CD28 monoclonal antibody TGN1412. N Engl J Med 355:1018–1028CrossRefPubMed
22.
Zurück zum Zitat Riechelmann H, Wiesneth M, Schauwecker P et al (2007) Adoptive therapy of head and neck squamous cell carcinoma with antibody coated immune cells: a pilot clinical trial. Cancer Immunol Immunother 56:1397–1406CrossRefPubMed Riechelmann H, Wiesneth M, Schauwecker P et al (2007) Adoptive therapy of head and neck squamous cell carcinoma with antibody coated immune cells: a pilot clinical trial. Cancer Immunol Immunother 56:1397–1406CrossRefPubMed
23.
Zurück zum Zitat Bugelski PJ, Achuthanandam R, Capocasale RJ et al (2009) Monoclonal antibody-induced cytokine-release syndrome. Expert Rev Clin Immunol 5:499–521CrossRefPubMed Bugelski PJ, Achuthanandam R, Capocasale RJ et al (2009) Monoclonal antibody-induced cytokine-release syndrome. Expert Rev Clin Immunol 5:499–521CrossRefPubMed
24.
Zurück zum Zitat Burges A, Wimberger P, Kümper C et al (2007) 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 13:3899–3905CrossRefPubMed Burges A, Wimberger P, Kümper C et al (2007) 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 13:3899–3905CrossRefPubMed
25.
Zurück zum Zitat Pietzner K, Vergote I, Santoro A et al (2014) Re-challenge with catumaxomab in patients with malignant ascites: results from the SECIMAS study. Med Oncol 31:308CrossRefPubMed Pietzner K, Vergote I, Santoro A et al (2014) Re-challenge with catumaxomab in patients with malignant ascites: results from the SECIMAS study. Med Oncol 31:308CrossRefPubMed
26.
Zurück zum Zitat Thomaidis T, Wörns MA, Galle PR et al (2014) Treatment of malignant ascites with a second cycle of catumaxomab in gastric signet cell carcinoma—a report of 2 cases. Oncol Res Treat 37:674–677CrossRefPubMed Thomaidis T, Wörns MA, Galle PR et al (2014) Treatment of malignant ascites with a second cycle of catumaxomab in gastric signet cell carcinoma—a report of 2 cases. Oncol Res Treat 37:674–677CrossRefPubMed
Metadaten
Titel
A phase I trial of intravenous catumaxomab: a bispecific monoclonal antibody targeting EpCAM and the T cell coreceptor CD3
verfasst von
Morten Mau-Sørensen
Christian Dittrich
Rodrigo Dienstmann
Ulrik Lassen
Wilfried Büchler
Holger Martinius
Josep Tabernero
Publikationsdatum
01.05.2015
Verlag
Springer Berlin Heidelberg
Erschienen in
Cancer Chemotherapy and Pharmacology / Ausgabe 5/2015
Print ISSN: 0344-5704
Elektronische ISSN: 1432-0843
DOI
https://doi.org/10.1007/s00280-015-2728-5

Weitere Artikel der Ausgabe 5/2015

Cancer Chemotherapy and Pharmacology 5/2015 Zur Ausgabe

Adjuvante Immuntherapie verlängert Leben bei RCC

25.04.2024 Nierenkarzinom Nachrichten

Nun gibt es auch Resultate zum Gesamtüberleben: Eine adjuvante Pembrolizumab-Therapie konnte in einer Phase-3-Studie das Leben von Menschen mit Nierenzellkarzinom deutlich verlängern. Die Sterberate war im Vergleich zu Placebo um 38% geringer.

Alectinib verbessert krankheitsfreies Überleben bei ALK-positivem NSCLC

25.04.2024 NSCLC Nachrichten

Das Risiko für Rezidiv oder Tod von Patienten und Patientinnen mit reseziertem ALK-positivem NSCLC ist unter einer adjuvanten Therapie mit dem Tyrosinkinase-Inhibitor Alectinib signifikant geringer als unter platinbasierter Chemotherapie.

Bei Senioren mit Prostatakarzinom auf Anämie achten!

24.04.2024 DGIM 2024 Nachrichten

Patienten, die zur Behandlung ihres Prostatakarzinoms eine Androgendeprivationstherapie erhalten, entwickeln nicht selten eine Anämie. Wer ältere Patienten internistisch mitbetreut, sollte auf diese Nebenwirkung achten.

ICI-Therapie in der Schwangerschaft wird gut toleriert

Müssen sich Schwangere einer Krebstherapie unterziehen, rufen Immuncheckpointinhibitoren offenbar nicht mehr unerwünschte Wirkungen hervor als andere Mittel gegen Krebs.

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.