Rituximab (RTX) and Ofatumumab (OFA) efficacies in cancer therapy depend in part on the induction of complement-dependent cytotoxicity (CDC). Human CD59 (hCD59) is a key complement regulatory protein that restricts the formation of membrane attack complex (MAC), thereby inhibiting induction of CDC. hCD59 is highly expressed in B-cell malignancies such as non-Hodgkin's lymphoma (NHL) and chronic lymphocytic leukemia (CLL) and up-regulation of hCD59 expression is an important determinant of the sensitivity of those cancer cells to RTX treatment. Therefore, it is imperative for us to develop a molecule capable of abrogating CD59 function in cancer cells and thus facilitate antibody- mediated cancer therapy. However, targeted toxicity effect from antibody specific against hCD59 and less efficacy of C8 or C9 peptides limit them for therapeutic purposes. Recently, we developed a specific and potent hCD59 inhibitor, rILYd4 (114aa), and demonstrated that rILYd4 enhances CDC in vitro and in vivo, thereby sensitizing RTX resistant lymphoma cells and primary CLL to RTX and OFA treatment. By defining PK/PD profiles of rILYd4 in mice, we showed that by itself rILYd4 does not adversely mediate in vivo hemolysis of hCD59-expressing erythrocytes. We demonstrated that increasing expression level of hCD59 in RTX-resistant cells occurs due to selection of pre-existing clones, rather than de novo induction of hCD59. We also demonstrated that the sensitivity to CDC effects mediated by OFA or RTX on RTX-resistant B-cell malignant cell lines and CLL cells negatively correlated with the level of CD59 on the cell surface. Taken together, our results rationalize the use of rILYd4 as a therapeutic adjuvant for RTX and OFA treatment of RTX-resistant NHL and CLL. Based on these results, we propose to optimize rILYd4 and develop less or non immunogenic, potent and stable rILYd4-derived peptide. We hypothesize that the optimized rILYd4 will improve the physicochemical properties of parental rILYd4, eliminate or reduce markedly immunogenicity, increase metabolic stability and half life in circulation, and enhance therapeutic efficacy, thereb leading to effective rILYd4-related therapeutics for enhancing as a adjuvant to anti-cancer drugs such as OFA and RTX in B-cell malignancies that have relapsed after prior antibody-based therapies. We propose to 1) to identify the sequences in rILYd4 responsible for its anti-hCD59 activity by phage display and synthetic approaches, and use them to develop a small, potent and non-immunogenic rILYd4-derived peptide (derivative) (Aim 1) and 2) to study its immune responses in human HLA transgenic mice and characterize its PK/PD, efficacy and toxicity profiles in humanized hCD59 transgenic mice.
(Aim 2). Successful outcome of this research will generate a novel therapeutic rILYd4-derived agent for clinical trials that will test its efficacy a an adjuvant for RTX- or OFA-based B-cell malignancy therapy and provide the much needed means to overcome the devastating RTX- or OFA-resistant B-cell malignancies.

Public Health Relevance

In this application, we will propose to develop a therapeutic anti-human CD59 inhibitor for the treatment of therapeutic antibody resistant B-cell malignancies. Successful outcome of this research will generate a novel therapeutic hCD59 inhibitor for clinical trials that will test its efficacy as an adjuvant for RTX- or OFA-based B-cel malignancy therapy and provide the much needed means to overcome the devastating RTX- or OFA-resistant B-cell malignancies.

Agency
National Institute of Health (NIH)
Institute
National Cancer Institute (NCI)
Type
Research Project (R01)
Project #
7R01CA166144-02
Application #
8725332
Study Section
Drug Discovery and Molecular Pharmacology Study Section (DMP)
Program Officer
Yovandich, Jason L
Project Start
2013-02-01
Project End
2018-01-31
Budget Start
2013-10-01
Budget End
2014-01-31
Support Year
2
Fiscal Year
2013
Total Cost
$223,660
Indirect Cost
$79,363
Name
Temple University
Department
Neurosciences
Type
Schools of Medicine
DUNS #
057123192
City
Philadelphia
State
PA
Country
United States
Zip Code
19122
Kearns, Alison; Gordon, Jennifer; Burdo, Tricia H et al. (2017) HIV-1-Associated Atherosclerosis: Unraveling the Missing Link. J Am Coll Cardiol 69:3084-3098
Kearns, Alison; Burdo, Tricia H; Qin, Xuebin (2017) Editorial Commentary: Clinical management of cardiovascular disease in HIV-infected patients. Trends Cardiovasc Med 27:564-566
Zhao, Senlin; Tang, Huamei; Yan, Dongwang et al. (2016) DDA1 promotes stage IIB-IIC colon cancer progression by activating NF?B/CSN2/GSK-3? signaling. Oncotarget 7:19794-812
Fu, Xiaoyan; Ju, Jiyu; Lin, Zhijuan et al. (2016) Target deletion of complement component 9 attenuates antibody-mediated hemolysis and lipopolysaccharide (LPS)-induced acute shock in mice. Sci Rep 6:30239
Zhong, Lin; Li, Hao; Li, Zhiqiang et al. (2016) C7 genotype of the donor may predict early bacterial infection after liver transplantation. Sci Rep 6:24121
Cheng, Dantong; Zhao, Senlin; Tang, Huamei et al. (2016) MicroRNA-20a-5p promotes colorectal cancer invasion and metastasis by downregulating Smad4. Oncotarget 7:45199-45213
Hanson, Marina M; Liu, Fengming; Dai, Shen et al. (2016) Rapid conditional targeted ablation model for hemolytic anemia in the rat. Physiol Genomics 48:626-32
Feng, Dechun; Dai, Shen; Liu, Fengming et al. (2016) Cre-inducible human CD59 mediates rapid cell ablation after intermedilysin administration. J Clin Invest 126:2321-33
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