Abstract

Fusion proteins combining hexavalent TRAIL with antibody fragments allow for a targeted delivery and efficient apoptosis induction in tumor cells. Here, we analyzed scFv-Fc-scTRAIL molecules directed against EGFR, HER2, HER3, and EpCAM as well as an untargeted Fc-scTRAIL fusion protein for their potentials to induce cell death both in vitro and in a xenograft tumor model in vivo. The scFv-Fc-scTRAIL fusion protein directed against EGFR as well as the fusion protein directed against EpCAM showed targeting effects on the two tested colorectal carcinoma cell lines Colo205 and HCT116, while a fusion protein targeting HER3 was more effective than untargeted Fc-scTRAIL only on Colo205 cells. Interestingly, another anti-HER3 scFv-Fc-scTRAIL fusion protein exhibiting approximately 10-fold weaker antigen binding as well as the HER2-directed molecule were unable to increase cytotoxicity compared to Fc-scTRAIL. A comparison of EC50 values of cell death induction and antigen binding supports the assumption that high affinity antigen binding is one of the requirements for in vitro targeting effects. Furthermore, a minimal number of expressed target antigens might be required for increased cytotoxicity of targeted compared to non-targeted molecules. In a Colo205 s.c. xenograft tumor model, strongest antitumor activity was observed for the anti-HER3 scFv-Fc-scTRAIL fusion protein based on antibody 3-43, with complete tumor remissions after six twice-weekly injections. Surprisingly, a similar in vivo activity was also observed for untargeted Fc-scTRAIL in this tumor model, indicating that additional factors contribute to the potent efficacy of targeted as well as untargeted hexavalent Fc-scTRAIL fusion proteins in vivo.

Highlights

  • Pro-apoptotic receptor agonists (PARAs) are powerful molecules to kill cancer cells independently of p53 and the intrinsic apoptosis pathway

  • Five different antibody moieties directed against four distinct tumorassociated antigens were employed, including the EGFRtargeting antibody hu225 derived from antibody C225 used in cetuximab [29] and humanized by CDR grafting [30], the trastuzumab-derived 4D5 directed against human epidermal growth factor receptor 2 (HER2) [31], the HER3-targeting antibodies 3M6 and 3-43 [33], as well as the humanized version 323/A3hu3 [34] of the anti-epithelial cell adhesion molecule (EpCAM) antibody 323/A3 [35, 36]

  • High molecular weight species were found for scFv4D5-fragment crystallizable (Fc)-single-chain variants of TNF-related apoptosis-inducing ligand (TRAIL) (scTRAIL), whereas fractions of smaller size were detected for scFv3M6-FcscTRAIL and scFv323/A3hu3-Fc-scTRAIL (Figure 2C)

Read more

Summary

Introduction

Pro-apoptotic receptor agonists (PARAs) are powerful molecules to kill cancer cells independently of p53 and the intrinsic apoptosis pathway. Despite promising anti-tumor effects in preclinical models [1, 2], soluble TRAIL (sTRAIL) as well as agonistic antibodies against the two death receptors TRAIL-R1 (DR4) and TRAIL-R2 (DR5) did not show therapeutic activity in clinical trials [3] This deficiency has been attributed to intrinsic or acquired resistance of many human tumors towards TRAILinduced apoptosis [4,5,6] as well as to a limited efficacy of the applied therapeutics. This insufficient activity has been linked to the lacking ability of sTRAIL and agonistic antibodies to induce higher order clustering of TRAIL receptors [7,8,9]. Based on the required clustering of death receptors for efficient apoptosis induction, several www.impactjournals.com/oncotarget strategies have been developed to enhance the activity of TRAIL-based therapeutics [8]

Methods
Results
Conclusion
Full Text
Paper version not known

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call