Abstract

Since trastuzumab monotherapy for treatment of breast cancer with HER2/ErbB2 over-expression has been shown to have limited efficacy, combined treatment of trastuzumab with chemotherapy is widely practiced in clinic. However, certain combination treatments of trastuzumab and chemotherapy (i.e. doxorubicin) are not recommended due to high risk of cardiotoxicity. Antibody-drug conjugates (ADCs) offer selective delivery of cytotoxic agents into targeted cancer cells, thereby allowing for reduced general cellular cytotoxicity caused by chemotherapeutic agents through antibody mediated specific recognition of tumor antigens. In this study, we constructed a trastuzumab-doxorubicin conjugate (T-Dox) using a thioether linkage and characterized both biophysical stability and anti-cancer potency of the T-Dox using a panel of HER2 expressing cancer cell lines. The T-Dox conjugate showed significantly improved anti-cancer potency in comparison with trastuzumab. The results demonstrated for the first time that there were significant differences in the uptake of T-Dox among high HER2 expression cancer cells and higher T-Dox uptake also showed stronger anti-cancer potency. Similar to trastuzumab, T-Dox selectively bound to HER2 overexpressing cancer cells and low HER2 expression cells had no detectable uptake of T-Dox. Consistent to the uptake data, human cardiomyocyte cells had no detectable HER2 expression and T-Dox showed minimal cytotoxic effects. On the contrary, a treatment with combination of trastuzumab and doxorubicin showed severe cytotoxicity to human cardiomyocytes (>90% cell death after 3 day exposure). This study demonstrated that trastuzumab conjugated with doxorubicin (T-Dox) can provide valuable alternative to the combination treatment with doxorubicin and trastuzumab for high HER2 expressing cancer patients.

Highlights

  • Doxorubicin is a DNA intercalating agent that has been used as an effective chemotherapeutic for treatment of many types of cancer, including breast cancers

  • Pursuing an analogous strategy to the trastuzumab-emtansine conjugate (T-DM1), we chose to couple the carbonyl function of doxorubicin to trastuzumab via a thioether linkage, using the maleimidohydrazide linker MMCCH (SMCC hydrazide) [22]

  • Characteristics of 4 batches of doxorubicin antibody conjugate preparations (2 batches for each of trastuzumab-doxorubicin conjugate (T-Dox) and IgGDox) are summarized in Supplemental Table S1; the molar ratio of doxorubicin coupled per molecule of antibody was between 4 and 5 for all four batches indicating the consistency of the conjugation process

Read more

Summary

Introduction

Doxorubicin is a DNA intercalating agent that has been used as an effective chemotherapeutic for treatment of many types of cancer, including breast cancers. Its use is severely limited due to cardiotoxicity, especially the risk of developing congestive heart failure with high cumulative doses (>400 mg/m2) [1]. Extensive efforts have been made to reduce cardiotoxicity and to improve the therapeutic index of doxorubicin [2,3,4,5,6,7,8,9]. Various delivery methods including liposomal polymers were investigated to improve the pharmacokinetics of doxorubicin in vivo [10,11,12,13,14,15]. Targeted delivery of doxorubicin using nanoparticles is being tested in clinical trials [16].

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