Phase I study of TrasGEX, a glyco-optimised anti-HER2 monoclonal antibody, in patients with HER2-positive solid tumours

Walter Fiedler, Herbert Stoeger, Antonella Perotti, Guenther Gastl, Jens Weidmann, Bruno Dietrich, Hans Baumeister, Antje Danielczyk, Steffen Goletz, Marc Salzberg, Sara De Dosso, Walter Fiedler, Herbert Stoeger, Antonella Perotti, Guenther Gastl, Jens Weidmann, Bruno Dietrich, Hans Baumeister, Antje Danielczyk, Steffen Goletz, Marc Salzberg, Sara De Dosso

Abstract

Purpose: TrasGEX is a second-generation monoclonal antibody of trastuzumab, glyco-optimised to enhance antibody-dependent cellular cytotoxicity while fully retaining trastuzumab's antigen-binding properties to human epidermal growth factor receptor 2 (HER2). A phase I dose-escalation study was conducted to establish the optimal TrasGEX dose and regimen for phase II studies and to define the safety, pharmacokinetics (PK) and preliminary antitumour activity of TrasGEX.

Patients and methods: A total of 37 patients with advanced HER2-positive carcinomas and progressive disease received TrasGEX intravenously every 3 weeks until disease progression in doses of 12-720 mg in a three-plus-three dose escalation design, including an expansion cohort at the highest dose.

Results: No dose limiting toxicity was observed, and no maximum tolerated dose was reached. Drug-related adverse events were mainly infusion-related reactions occurring during the first infusion in 51% of patients; all but two were mild-to-moderate. Compared with trastuzumab, the PK parameters were dose dependent, with a mean terminal half-life (t1/2) of 263±99 hours for the 720 mg dose. Clinical benefit in 15 out of 30 (50%) evaluable patients included one ongoing complete response, two partial remissions lasting 16 and 77 weeks and disease stabilisation (SD) in 12 patients lasting a median (range) of 17 (7-26) weeks; three of them had SD of 24, 25 and 26 weeks, respectively.

Conclusion: TrasGEX was safe, well-tolerated and showed antitumour activity in 50% of evaluable patients, all with progressive disease at study entry. Infusions at an interval of 2-3 weeks should achieve clinically relevant trough levels for future studies (NCT01409343).

Keywords: glycoengineered monoclonal antibody; her2; phase I; solid tumours; trasgex.

Conflict of interest statement

Competing interests: All authors have completed the ICMJE uniform disclosure form and have sent them to the corresponding author. BD, HB, AD and SG are/were employees of Glycotope GmbH. Employment or leadership position: BD, H, AD and SG. Patent holders: HB, SG and AD. Stock ownership: HB, SG and AD. MS was employee of Medpace Ltd, which received funding for the study from Glycotope GmbH. Other remunerations: WF, meeting participation to present the data at the American Society of Clinical Oncology 2013 Annual Meeting. All remaining authors declare no support from any organisation for the submitted work; no financial relationships with any organisations that might have an interest in the submitted work in the previous three years; no other relationships or activities that could appear to have influenced the submitted work.

Figures

Figure 1
Figure 1
Scatterplot of (A) terminal half-life (t1/2) and (B) clearance (CL) by dose cohort calculated for the first TrasGEX infusion.
Figure 2
Figure 2
Waterfall plots of the best per cent change from baseline in sum of longest diameters (SLD) for target lesions. Baseline is defined as the last non-missing value before the first dose of TrasGEX. Twenty-eight patients in the total population (n= 37) had valid baseline and postbaseline values. Tumour assessment was not performed in nine patients because of absence of target lesions (n= 2), or early withdrawal due to clinical deterioration (n= 3), adverse event (n=3) or withdrawal of informed consent (n= 1). The red dotted lines indicate the cut off for partial response (−30%) and progressive disease (+20%). Bars marked with an asterisk denote nine patients with stable target lesions but progressive disease because of progression of non-target lesions or appearance of new lesions. BC, breast cancer; bladder CA, urinary bladder cancer; CRC, colorectal cancer; EGCA, esophagogastric junction cancer; GCA, gastric cancer; NSCLC, non-small cell lung cancer; OVCA, ovarian cancer; PanC, pancreatic cancer; parotid CA, parotid gland cancer.

References

    1. Hudis CA. Trastuzumab--mechanism of action and use in clinical practice. N Engl J Med 2007;357:39–51. 10.1056/NEJMra043186
    1. Gianni L, Eiermann W, Semiglazov V, et al. . Neoadjuvant chemotherapy with trastuzumab followed by adjuvant trastuzumab versus neoadjuvant chemotherapy alone, in patients with HER2-positive locally advanced breast cancer (the NOAH trial): a randomised controlled superiority trial with a parallel HER2-negative cohort. Lancet 2010;375:377–84. 10.1016/S0140-6736(09)61964-4
    1. Bang YJ, Van Cutsem E, Feyereislova A, et al. . Trastuzumab in combination with chemotherapy versus chemotherapy alone for treatment of HER2-positive advanced gastric or gastro-oesophageal junction cancer (ToGA): a phase 3, open-label, randomised controlled trial. Lancet 2010;376:687–97. 10.1016/S0140-6736(10)61121-X
    1. Yarden Y, Sliwkowski MX. Untangling the ErbB signalling network. Nat Rev Mol Cell Biol 2001;2:127–37. 10.1038/35052073
    1. Spector NL, Blackwell KL. Understanding the mechanisms behind trastuzumab therapy for human epidermal growth factor receptor 2-positive breast cancer. J Clin Oncol 2009;27:5838–47. 10.1200/JCO.2009.22.1507
    1. Bianchini G, Gianni L. The immune system and response to HER2-targeted treatment in breast cancer. Lancet Oncol 2014;15:e58–e68. 10.1016/S1470-2045(13)70477-7
    1. Ferris RL, Jaffee EM, Ferrone S. Tumor antigen-targeted, monoclonal antibody-based immunotherapy: clinical response, cellular immunity, and immunoescape. J Clin Oncol 2010;28:4390–9. 10.1200/JCO.2009.27.6360
    1. Cartron G, Dacheux L, Salles G, et al. . Therapeutic activity of humanized anti-CD20 monoclonal antibody and polymorphism in IgG Fc receptor FcgammaRIIIa gene. Blood 2002;99:754–8. 10.1182/blood.V99.3.754
    1. Dall’Ozzo S, Tartas S, Paintaud G, et al. . Rituximab-dependent cytotoxicity by natural killer cells: influence of FCGR3A polymorphism on the concentration-effect relationship. Cancer Res 2004;64:4664–9. 10.1158/0008-5472.CAN-03-2862
    1. Musolino A, Naldi N, Bortesi B, et al. . Immunoglobulin G fragment C receptor polymorphisms and clinical efficacy of trastuzumab-based therapy in patients with HER-2/neu-positive metastatic breast cancer. J Clin Oncol 2008;26:1789–96. 10.1200/JCO.2007.14.8957
    1. Shields RL, Lai J, Keck R, et al. . Lack of fucose on human IgG1 N-linked oligosaccharide improves binding to human Fcgamma RIII and antibody-dependent cellular toxicity. J Biol Chem 2002;277:26733–40. 10.1074/jbc.M202069200
    1. Niwa R, Hatanaka S, Shoji-Hosaka E, et al. . Enhancement of the antibody-dependent cellular cytotoxicity of low-fucose IgG1 Is independent of FcgRIIIa functional polymorphism. Clin Cancer Res 2010;2004:6248–55.
    1. Suzuki E, Niwa R, Saji S, et al. . A nonfucosylated anti-HER2 antibody augments antibody-dependent cellular cytotoxicity in breast cancer patients. Clin Cancer Res 2007;13:1875–82. 10.1158/1078-0432.CCR-06-1335
    1. Junttila TT, Parsons K, Olsson C, et al. . Superior in vivo efficacy of afucosylated trastuzumab in the treatment of HER2-amplified breast cancer. Cancer Res 2010;70:4481–9. 10.1158/0008-5472.CAN-09-3704
    1. Eisenhauer EA, Therasse P, Bogaerts J, et al. . New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1). Eur J Cancer 2009;45:228–47. 10.1016/j.ejca.2008.10.026
    1. National Cancer Institute Common Terminology Criteria for Adverse Events version 4.0.
    1. Baselga J, Tripathy D, Mendelsohn J, et al. . Phase II study of weekly intravenous recombinant humanized anti-p185HER2 monoclonal antibody in patients with HER2/neu-overexpressing metastatic breast cancer. J Clin Oncol 1996;14:737–44. 10.1200/JCO.1996.14.3.737
    1. Cobleigh MA, Vogel CL, Tripathy D, et al. . Multinational study of the efficacy and safety of humanized anti-HER2 monoclonal antibody in women who have HER2-overexpressing metastatic breast cancer that has progressed after chemotherapy for metastatic disease. J Clin Oncol 1999;17:2639–48. 10.1200/JCO.1999.17.9.2639
    1. Tokuda Y, Watanabe T, Omuro Y, et al. . Dose escalation and pharmacokinetic study of a humanized anti-HER2 monoclonal antibody in patients with HER2/neu-overexpressing metastatic breast cancer. Br J Cancer 1999;81:1419–25. 10.1038/sj.bjc.6690343
    1. Kumai T, Oikawa K, Aoki N, et al. . Assessment of the change in cetuximab induced ADCC activity of NK cells by steroid. Head Neck 2016;38:410–6.
    1. Eisner F, Pichler M, Goletz S, et al. . A glyco-engineered anti-HER2 monoclonal antibody (TrasGEX) induces a long-lasting remission in a patient with HER2 overexpressing metastatic colorectal cancer after failure of all available treatment options. J Clin Pathol 2015;68:1044.1–6. 10.1136/jclinpath-2015-202996
    1. Leyland-Jones B, Gelmon K, Ayoub JP, et al. . Pharmacokinetics, safety, and efficacy of trastuzumab administered every three weeks in combination with paclitaxel. J Clin Oncol 2003;21:3965–71. 10.1200/JCO.2003.12.109
    1. Summary of product characteristics of Herceptin.
    1. Bruno R, Washington CB, Lu JF, Jf L, et al. . Population pharmacokinetics of trastuzumab in patients with HER2+ metastatic breast cancer. Cancer Chemother Pharmacol 2005;56:361–9. 10.1007/s00280-005-1026-z
    1. Niwa R, Sakurada M, Kobayashi Y, et al. . Enhanced natural killer cell binding and activation by low-fucose IgG1 antibody results in potent antibody-dependent cellular cytotoxicity induction at lower antigen density. Clin Cancer Res 2005;11:2327–36. 10.1158/1078-0432.CCR-04-2263
    1. Gennari R, Menard S, Fagnoni F, et al. . Pilot study of the mechanism of action of preoperative trastuzumab in patients with primary operable breast tumors overexpressing HER2. Clin Cancer Res 2004;10:5650–5. 10.1158/1078-0432.CCR-04-0225
    1. Hurvitz SA, Betting DJ, Stern HM, et al. . Analysis of Fcγ receptor IIIa and IIa polymorphisms: lack of correlation with outcome in trastuzumab-treated breast cancer patients. Clin Cancer Res 2012;18:3478–86. 10.1158/1078-0432.CCR-11-2294
    1. Norton N, Olson RM, Pegram M, et al. . Association studies of Fcγ receptor polymorphisms with outcome in HER2+ breast cancer patients treated with trastuzumab in NCCTG (Alliance) Trial N9831. Cancer Immunol Res 2014;2:962–9. 10.1158/2326-6066.CIR-14-0059
    1. Slamon DJ, Leyland-Jones B, Shak S, et al. . Use of chemotherapy plus a monoclonal antibody against HER2 for metastatic breast cancer that overexpresses HER2. N Engl J Med 2001;344:783–92. 10.1056/NEJM200103153441101
    1. Baselga J. Herceptin alone or in combination with chemotherapy in the treatment of HER2-positive metastatic breast cancer: pivotal trials. Oncology 2001;61 Suppl 2(Suppl 2):14–21. 10.1159/000055397
    1. Marty M, Cognetti F, Maraninchi D, et al. . Randomized phase II trial of the efficacy and safety of trastuzumab combined with docetaxel in patients with human epidermal growth factor receptor 2-positive metastatic breast cancer administered as first-line treatment: the M77001 study group. J Clin Oncol 2005;23:4265–74. 10.1200/JCO.2005.04.173

Source: PubMed

3
Subskrybuj