Abstract

Simple SummaryIL13Rα2 has been repeatedly reported as an excellent therapeutic target for multiple types of advanced cancers. However, previous IL13Rα2 targeting attempts have been mostly unsuccessful. Here, we describe a novel strategy based on the blocking of the IL-13 tumorigenic activity using a highly preserved D1 peptide selected from the IL13Rα2 binding site for mouse immunization and the inhibition of the cell invasion capacity for antibody screening. The IL13Rα2 D1 peptide-specific monoclonal antibody 5.5.4 has demonstrated a large capacity for blocking IL13Rα2 signaling capacity and protecting mice against established and non-established liver metastasis in colorectal cancer. These positive results predict a potential application to other IL13Rα2 positive cancers.Background: IL13Rα2 is reportedly a promising therapeutic target in different cancers. Still, no specific antagonists have reached the clinics yet. We investigated the use of a IL-13/IL13Rα2 binding motif, called D1, as a new target for the development of therapeutic monoclonal antibodies (mAbs) for colorectal cancer (CRC) metastasis. Methods: IL13Rα2 D1 peptides were prepared and used for immunization and antibody development. Antibodies were tested for inhibition of cellular invasion through Matrigel using CRC cell lines. Effects of the mAbs on cell signaling, receptor internalization and degradation were determined by western blot and flow cytometry. Swiss nude mice were used for survival analysis after treatment with IL13Rα2-specific mAbs and metastasis development. Results: IL13Rα2 D1 peptides were used to generate highly selective mAbs that blocked IL13/IL13Rα2-mediated SRC activation and cell invasion in colorectal cancer cells. Antibodies also provoked a significant reduction in cell adhesion and proliferation of metastatic cancer cells. Treatment with mAbs impaired the FAK, SRC and PI3K/AKT pathway activation. Blocking effectivity was shown to correlate with the cellular IL13Rα2 expression level. Despite mAb 5.5.4 partially blocked IL-13 mediated receptor internalization from the cancer cell surface it still promotes receptor degradation. Compared with other IL13Rα2-specific antibodies, 5.5.4 exhibited a superior efficacy to inhibit metastatic growth in vivo, providing a complete mouse survival in different conditions, including established metastasis. Conclusions: Monoclonal antibody 5.5.4 showed a highly selective blocking capacity for the interaction between IL-13 and IL13Rα2 and caused a complete inhibition of liver metastasis in IL13Rα2-positive colorectal cancer cells. This capacity might be potentially applicable to other IL13Rα2-expressing tumors.

Highlights

  • Colorectal cancer (CRC) is among the four most lethal tumors worldwide [1]

  • Mice were immunized with a 19-mer D1-derived IL-13 receptor α2 (IL13Rα2)-peptide coupled to OVA (Figure S1)

  • Serum #2 showed a 40% inhibition of the cellular invasive capacity (Figure 1A). This mouse was selected for spleen fusion and antibody production after a final boost with a recombinant IL13Rα2-Fc protein (Figure S1)

Read more

Summary

Introduction

Colorectal cancer (CRC) is among the four most lethal tumors worldwide [1]. Europe shows a high incidence, with the majority of its countries having a rate of 26 deaths per 100,000 people [1]. We investigated the use of a IL-13/IL13Rα2 binding motif, called D1, as a new target for the development of therapeutic monoclonal antibodies (mAbs) for colorectal cancer (CRC) metastasis. Compared with other IL13Rα2-specific antibodies, 5.5.4 exhibited a superior efficacy to inhibit metastatic growth in vivo, providing a complete mouse survival in different conditions, including established metastasis. Conclusions: Monoclonal antibody 5.5.4 showed a highly selective blocking capacity for the interaction between IL-13 and IL13Rα2 and caused a complete inhibition of liver metastasis in IL13Rα2-positive colorectal cancer cells. This capacity might be potentially applicable to other IL13Rα2-expressing tumors

Methods
Results
Conclusion
Full Text
Published version (Free)

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