CD59 is a key regulator for restricting the formation of the membrane attack complex (MAC) of complement by binding to complement components C8 and C9 and preventing C9 incorporation and polymerization in the cell membrane. Complement is a key mediator for antibody-based cancer therapy such as complement-dependent cytotoxicity (CDC) and antibody-dependent-cell mediated cytotoxicity (ADCC). CD59 is universally expressed in all cells and highly expressed in many kinds of cancer cells including B-cell lymphoma. CD59 appears to be the membrane complement regulator that is most effective at protecting tumor cells from antibody-mediated CDC. Extensive studies indicate that upregulation of CD59 is responsible for lymphoma resistance to rituximab treatment. Therefore, it is imperative for us to develop a molecule capable of abrogating CD59 function in cancer cells and facilitating antibody-mediated cancer therapy. However, targeted toxicity effect from miniantibody specific against hCD59 and less efficacy of C8 or C9 peptides limit them for therapeutic purposes. Intermedilysin (ILY), a cytolytic pore-forming toxin secreted by Streptococcus intermedius, lyses only human cells due to its receptor specificity for human CD59 (hCD59). Recently, we used hCD59 transgenic mice to confirm the finding that ILY binds only to hCD59 in vivo. Domain 4 of ILY binds to AA42-58 in hCD59, which also participate in the binding to C8 and C9. Thus, we hypothesize that the truncated ILY presenting only domain 4 will specifically abrogate hCD59 function and facilitate antibody- mediated and complement-dependant cancer cytolysis. Consistently, our preliminary results have demonstrated that the recombinant protein (114AA) derived from the ILY domain 4 (rILYd4) specifically blocks hCD59 function ex vivo. Furthermore, we demonstrated that application of rILYd4 (IC50 = 33 nM in vitro and effective dose = 2.5 ug/g body weight in vivo ) to B lymphoma cell lines (RAMOS), which are resistant to Rituximab-mediated CDC (so called RRR cells), sensitized them to Rituximab-mediated CDC in lymphoma without off target toxicity effects. We propose to use rituximab as a therapeutic antibody in lymphoma xenografted athymic nude mice that express transgenic hCD59 to test this hypothesis. Specifically, we will determine and the efficacy and specificity of rILY4 (specific aim 1) and characterize rILYd4 immunogenicity (Aim 2). Success in this application will yield a newly-identified CD59 inhibitor, domain 4 of ILY as an adjunct to not only rituximab-based lymphoma therapy, but also other antibody-based cancer therapy.

Public Health Relevance

In this application, we will assess the in vivo efficacy of the nature product rILYd4 for antibody (rituximab)-mediated cancer (B-lymphoma) therapy. Success of this work will foster us to develop novel anti-cancer reagents, an anti-human CD59 inhibitor rILYd4, which can significantly facilitate antibody- mediated cancer therapy, not only in lymphoma, but also in other cancers.

Agency
National Institute of Health (NIH)
Institute
National Cancer Institute (NCI)
Type
Exploratory/Developmental Grants (R21)
Project #
5R21CA141324-02
Application #
8009892
Study Section
Cancer Immunopathology and Immunotherapy Study Section (CII)
Program Officer
Yovandich, Jason L
Project Start
2010-01-01
Project End
2012-06-30
Budget Start
2011-01-01
Budget End
2012-06-30
Support Year
2
Fiscal Year
2011
Total Cost
$214,562
Indirect Cost
Name
Harvard University
Department
Type
Organized Research Units
DUNS #
047006379
City
Boston
State
MA
Country
United States
Zip Code
02115
Liu, Fengming; Dai, Shen; Gordon, Jennifer et al. (2014) Complement and HIV-I infection/HIV-associated neurocognitive disorders. J Neurovirol 20:184-98
Cai, Bishuang; Xie, Shuwei; Liu, Fengming et al. (2014) Rapid degradation of the complement regulator, CD59, by a novel inhibitor. J Biol Chem 289:12109-25
Wu, Lin; Su, Sanbao; Liu, Fengming et al. (2012) Removal of the tag from His-tagged ILYd4, a human CD59 inhibitor, significantly improves its physical properties and its activity. Curr Pharm Des 18:4187-96
Amet, Tohti; Ghabril, Marwan; Chalasani, Naga et al. (2012) CD59 incorporation protects hepatitis C virus against complement-mediated destruction. Hepatology 55:354-63
Hu, Weiguo; Ge, Xiaowen; You, Tao et al. (2011) Human CD59 inhibitor sensitizes rituximab-resistant lymphoma cells to complement-mediated cytolysis. Cancer Res 71:2298-307
You, Tao; Hu, Weiguo; Ge, Xiaowen et al. (2011) Application of a novel inhibitor of human CD59 for the enhancement of complement-dependent cytolysis on cancer cells. Cell Mol Immunol 8:157-63
Ge, Xiaowen; Wu, Lin; Hu, Weiguo et al. (2011) rILYd4, a human CD59 inhibitor, enhances complement-dependent cytotoxicity of ofatumumab against rituximab-resistant B-cell lymphoma cells and chronic lymphocytic leukemia. Clin Cancer Res 17:6702-11