A phase 1b study of humanized KS-interleukin-2 (huKS-IL2) immunocytokine with cyclophosphamide in patients with EpCAM-positive advanced solid tumors

Joseph P Connor, Mihaela C Cristea, Nancy L Lewis, Lionel D Lewis, Philip B Komarnitsky, Maria R Mattiacci, Mildred Felder, Sarah Stewart, Josephine Harter, Jean Henslee-Downey, Daniel Kramer, Roland Neugebauer, Roger Stupp, Joseph P Connor, Mihaela C Cristea, Nancy L Lewis, Lionel D Lewis, Philip B Komarnitsky, Maria R Mattiacci, Mildred Felder, Sarah Stewart, Josephine Harter, Jean Henslee-Downey, Daniel Kramer, Roland Neugebauer, Roger Stupp

Abstract

Background: Humanized KS-interleukin-2 (huKS-IL2), an immunocytokine with specificity for epithelial cell adhesion molecule (EpCAM), has demonstrated favorable tolerability and immunologic activity as a single agent.

Methods: Phase 1b study in patients with EpCAM-positive advanced solid tumors to determine the maximum tolerated dose (MTD) and safety profile of huKS-IL2 in combination with low-dose cyclophosphamide. Treatment consisted of cyclophosphamide (300 mg/m2 on day 1), and escalating doses of huKS-IL2 (0.5-4.0 mg/m2 IV continuous infusion over 4 hours) on days 2, 3, and 4 of each 21-day cycle. Safety, pharmacokinetic profile, immunogenicity, anti-tumor and biologic activity were evaluated.

Results: Twenty-seven patients were treated for up to 6 cycles; 26 were evaluable for response. The MTD of huKS-IL2 in combination with 300 mg/m2 cyclophosphamide was 3.0 mg/m2. At higher doses, myelosuppression was dose-limiting. Transient lymphopenia was the most common grade 3/4 adverse event (AE). Other significant AEs included hypotension, hypophosphatemia, and increase in serum creatinine. All patients recovered from these AEs. The huKS-IL2 exposure was dose-dependent, but not dose-proportional, accumulation was negligible, and elimination half-life and systemic clearance were independent of dose and time. Most patients had a transient immune response to huKS-IL2. Immunologic activity was observed at all doses. Ten patients (38%) had stable disease as best response, lasting for ≥ 4 cycles in 3 patients.

Conclusion: The combination of huKS-IL2 with low-dose cyclophosphamide was well tolerated. Although no objective responses were observed, the combination showed evidence of immunologic activity and 3 patients showed stable disease for ≥ 4 cycles.

Trial registration: ClinicalTrials.gov NCT00132522.

Figures

Figure 1
Figure 1
Mean (standard deviation) serum concentration profiles on day 2 of cycle 1.
Figure 2
Figure 2
Dose–response relationship of huKS-IL2. Day 2, cycle 1, as shown by Cmax(a) and AUC0–24h(b). Cmax, maximum or peak serum concentration; AUC0–24h, area under the concentration versus time curve from time zero to 24 hours after the start of infusion.
Figure 3
Figure 3
Serum levels of antibodies over time. (a) Anti-idiotype antibodies; (b) anti-Fc-IL2 antibodies; and (c) anti-IL2 antibodies.
Figure 4
Figure 4
Immune cell monitoring. Cytotoxic natural killer cells (a); CD4+ T-cells (b); CD8+ T-cells (c); and T-reg cells (d). C1D1, cycle 1 day 1; C1D2, cycle 2 day 2, etc.
Figure 5
Figure 5
Kaplan–Meier plot of overall survival. * The Kaplan–Meier method was applied only for dose-level groups with at least 6 patients.

References

    1. Trzpis M, McLaughlin PM, de Leij LM, Harmsen MC. Epithelial cell adhesion molecule: more than a carcinoma marker and adhesion molecule. Am J Pathol. 2007;171:386–395. doi: 10.2353/ajpath.2007.070152.
    1. van der Gun BT, Melchers LJ, Ruiters MH, de Leij LF, McLaughlin PM, Rots MG. EpCAM in carcinogenesis: the good, the bad or the ugly. Carcinogenesis. 2010;31:1913–1921. doi: 10.1093/carcin/bgq187.
    1. Connor JP, Felder M, Hank J, Harter J, Gan J, Gillies SD, Sondel P. Ex vivo evaluation of anti-EpCAM immunocytokine huKS-IL2 in ovarian cancer. J Immunother. 2004;27:211–219. doi: 10.1097/00002371-200405000-00005.
    1. Osta WA, Chen Y, Mikhitarian K, Mitas M, Salem M, Hannun YA, Cole DJ, Gillanders WE. EpCAM is overexpressed in breast cancer and is a potential target for breast cancer gene therapy. Cancer Res. 2004;64:5818–5824. doi: 10.1158/0008-5472.CAN-04-0754.
    1. Chaudry MA, Sales K, Ruf P, Lindhofer H, Winslet MC. EpCAM an immunotherapeutic target for gastrointestinal malignancy: current experience and future challenges. Br J Cancer. 2007;96:1013–1019. doi: 10.1038/sj.bjc.6603505.
    1. Kimura H, Kato H, Faried A, Sohda M, Nakajima M, Fukai Y, Miyazaki T, Masuda N, Fukuchi M, Kuwano H. Prognostic significance of EpCAM expression in human esophageal cancer. Int J Oncol. 2007;30:171–179.
    1. Kim Y, Kim HS, Cui ZY, Lee HS, Ahn JS, Park CK, Park K, Ahn MJ. Clinicopathological implications of EpCAM expression in adenocarcinoma of the lung. Anticancer Res. 2009;29:1817–1822.
    1. Songun I, Litvinov SV, van de Velde CJ, Pals ST, Hermans J, van Krieken JH. Loss of Ep-CAM (CO17-1A) expression predicts survival in patients with gastric cancer. Br J Cancer. 2005;92:1767–1772. doi: 10.1038/sj.bjc.6602519.
    1. Gastl G, Spizzo G, Obrist P, Dünser M, Mikuz G. Ep-CAM overexpression in breast cancer as a predictor of survival. Lancet. 2000;356:1981–1982. doi: 10.1016/S0140-6736(00)03312-2.
    1. MacDonald GC, Rasamoelisolo M, Entwistle J, Cizeau J, Bosc D, Cuthbert W, Kowalski M, Spearman M, Glover N. A phase I clinical study of VB4-845: weekly intratumoral administration of an anti-EpCAM recombinant fusion protein in patients with squamous cell carcinoma of the head and neck. Drug Des Devel Ther. 2009;2:105–114.
    1. Mosolits S, Markovic K, Frödin JE, Virving L, Magnusson CG, Steinitz M, Fagerberg J, Mellstedt H. Vaccination with Ep-CAM protein or anti-idiotypic antibody induces Th1-biased response against MHC class I- and II-restricted EpCAM epitopes in colorectal carcinoma patients. Clin Cancer Res. 2004;10:5391–5402. doi: 10.1158/1078-0432.CCR-04-0425.
    1. Schanzer JM, Fichtner I, Baeuerle PA, Kufer P. Antitumor activity of a dual cytokine/single-chain antibody fusion protein for simultaneous delivery of GM-CSF and IL-2 to EpCAM expressing tumor cells. J Immunother. 2006;29:477–488. doi: 10.1097/01.cji.0000210080.60178.fd.
    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;97:315–321. doi: 10.1038/sj.bjc.6603881.
    1. Salnikov AV, Groth A, Apel A, Kallifatidis G, Beckermann BM, Khamidjanov A, Ryschich E, Büchler MW, Herr I, Moldenhauer G. Targeting of cancer stem cell marker EpCAM by bispecific antibody EpCAMxCD3 inhibits pancreatic carcinoma. J Cell Mol Med. 2009;13:4023–4033. doi: 10.1111/j.1582-4934.2009.00723.x.
    1. Richter CE, Cocco E, Bellone S, Silasi DA, Rüttinger D, Azodi M, Schwartz PE, Rutherford TJ, Pecorelli S, Santin AD. High-grade, chemotherapy-resistant ovarian carcinomas overexpress epithelial cell adhesion molecule (EpCAM) and are highly sensitive to immunotherapy with MT201, a fully human monoclonal anti-EpCAM antibody. Am J Obstet Gynecol. 2010;203:582. e1-7.
    1. Patriarca C, Macchi RM, Marschner AK, Mellstedt H. Epithelial cell adhesion molecule expression (CD326) in cancer: a short review. Cancer Treat Rev. 2012;38:68–75. doi: 10.1016/j.ctrv.2011.04.002.
    1. Johnson EE, Lum HD, Rakhmilevich AL, Schmidt BE, Furlong M, Buhtoiarov IN, Hank JA, Raubitschek A, Colcher D, Reisfeld RA, Gillies SD, Sondel PM. Intratumoral immunocytokine treatment results in enhanced antitumor effects. Cancer Immunol Immunother. 2008;57:1891–1902. doi: 10.1007/s00262-008-0519-0.
    1. Holden SA, Lan Y, Pardo AM, Wesolowski JS, Gillies SD. Augmentation of antitumor activity of an antibody-interleukin 2 immunocytokine with chemotherapeutic agents. Clin Cancer Res. 2001;7:2862–2869.
    1. Ko YJ, Bubley GJ, Weber R, Redfern C, Gold DP, Finke L, Kovar A, Dahl T, Gillies SD. Safety, pharmacokinetics, and biological pharmacodynamics of the immunocytokine EMD 273066 (huKS-IL2): results of a phase I trial in patients with prostate cancer. J Immunother. 2004;27:232–239. doi: 10.1097/00002371-200405000-00008.
    1. Thompson JA, Lee DJ, Cox WW, Lindgren CG, Collins C, Neraas KA, Dennin RA, Fefer A. Recombinant interleukin 2 toxicity, pharmacokinetics, and immunomodulatory effects in a phase I trial. Cancer Res. 1987;47:4202–4207.
    1. King DM, Albertini MR, Schalch H, Hank JA, Gan J, Surfus J, Mahvi D, Schiller JH, Warner T, Kim K, Eickhoff J, Kendra K, Reisfeld R, Gillies SD, Sondel P. Phase I clinical trial of the immunocytokine EMD 273063 in melanoma patients. J Clin Oncol. 2004;22:4463–4473. doi: 10.1200/JCO.2004.11.035.
    1. Ribas A, Kirkwood JM, Atkins MB, Whiteside TL, Gooding W, Kovar A, Gillies SD, Kashala O, Morse MA. Phase I/II open-label study of the biologic effects of the interleukin-2 immunocytokine EMD 273063 (hu14.18-IL2) in patients with metastatic malignant melanoma. J Transl Med. 2009;7:68. doi: 10.1186/1479-5876-7-68.
    1. Quan WD Jr, Quan FM, King LA, Walker PR. Low-dose cyclophosphamide and continuous-infusion interleukin-2 with famotidine in previously treated metastatic melanoma or kidney cancer. Cancer Biother Radiopharm. 2008;23:108–113. doi: 10.1089/cbr.2007.0420.
    1. Freeman SM, Franco JL, Kenady DE, Baltzer L, Roth Z, Brandwein HJ, Hadden JW. A phase 1 safety study of an IRX-2 regimen in patients with squamous cell carcinoma of the head and neck. Am J Clin Oncol. 2011;34:173–178.
    1. Singh H, Hilf N, Mendrzyk R, Maurer D, Weinschenk T, Kirner A, Frisch J, Reinhardt C, Stenzl A, Walter S. Correlation of immune responses with survival in a randomized phase II study investigating multipeptide vaccination with IMA901 plus or minus low-dose cyclophosphamide in advanced renal cell carcinoma (RCC) J Clin Oncol. 2010;28(suppl):abstract 2587.
    1. Lindemann A, Höffken K, Schmidt RE, Diehl V, Kloke O, Gamm H, Hayungs J, Oster W, Böhm M, Kolitz JE. A phase-II study of low-dose cyclophosphamide and recombinant human interleukin-2 in metastatic renal cell carcinoma and malignant melanoma. Cancer Immunol Immunother. 1989;28:275–281.
    1. Alderson KL, Sondel PM. Clinical cancer therapy by NK cells via antibody-dependent cell-mediated cytotoxicity. J Biomed Biotechnol. 2011;2011:379123.
    1. Sondel PM, Gillies SD. Current and potential uses of immunocytokines as cancer immunotherapy. Antibodies. 2012;1:149–171. doi: 10.3390/antib1020149.

Source: PubMed

3
Subskrybuj