Abstract

Tumour resistance to radiotherapy remains a barrier to improving cancer patient outcomes. To overcome radioresistance, certain drugs have been found to sensitize cells to ionizing radiation (IR). In theory, more potent radiosensitizing drugs should increase tumour kill and improve patient outcomes. In practice, clinical utility of potent radiosensitizing drugs is curtailed by off-target side effects. Here we report potent anti-tubulin drugs conjugated to anti-ErbB antibodies selectively radiosensitize to tumours based on surface receptor expression. While two classes of potent anti-tubulins, auristatins and maytansinoids, indiscriminately radiosensitize tumour cells, conjugating these potent anti-tubulins to anti-ErbB antibodies restrict their radiosensitizing capacity. Of translational significance, we report that a clinically used maytansinoid ADC, ado-trastuzumab emtansine (T-DM1), with IR prolongs tumour control in target expressing HER2+ tumours but not target negative tumours. In contrast to ErbB signal inhibition, our findings establish an alternative therapeutic paradigm for ErbB-based radiosensitization using antibodies to restrict radiosensitizer delivery.

Highlights

  • Tumour resistance to radiotherapy remains a barrier to improving cancer patient outcomes

  • On the basis of these findings, we propose antibody drug conjugate based chemoradiotherapy paradigms designed to focus on antibody directed delivery of highly potent radiosensitizing chemotherapies as an alternative to receptor signal inhibition

  • Cetuximab and trastuzumab were labelled at endogenous cysteines by selective reduction of the four disulfides in the hinge region and conjugation confirmed by ES-HPLC, with drug loading measured as B3.7 and B3.2 monomethyl auristatin E (MMAE) per molecule of cetuximab and trastuzumab, respectively and with B1 Cy5

Read more

Summary

Introduction

Tumour resistance to radiotherapy remains a barrier to improving cancer patient outcomes. To test if ADC can restrict MMAE radiosensitization to tumours, we conjugated MMAE to cetuximab (C-MMAE) and trastuzumab (T-MMAE) and labelled them with Cy5 for tracking (Supplementary Figs 1a and 2). Irradiation of vehicle-treated CAL-27 cells resulted in a 3.6-fold increase in comet tail length that was significantly increased by 0.5 nM C-MMAE but not by 5 nM cetuximab (Fig. 2d).

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

Disclaimer: All third-party content on this website/platform is and will remain the property of their respective owners and is provided on "as is" basis without any warranties, express or implied. Use of third-party content does not indicate any affiliation, sponsorship with or endorsement by them. Any references to third-party content is to identify the corresponding services and shall be considered fair use under The CopyrightLaw.