Abstract

Many cancer research efforts focus on exploiting genetic-level features that may be targeted for therapy. Tissue-level features of the tumour microenvironment also represent useful therapeutic targets. Here we investigate the presence of low oxygen tension and sensitivity to NOS inhibition of tumour vasculature as potential tumour-specific features that may be targeted by hypoxic cytotoxins, a class of therapeutics currently under investigation. We have previously demonstrated that tirapazamine (TPZ) mediates central vascular dysfunction in tumours. TPZ is a hypoxic cytotoxin that is also a competitive inhibitor of NOS. Here we further investigated the vascular-targeting activity of TPZ by combining it with NOS inhibitor L-NNA, or with low oxygen content gas breathing. Tumours were analyzed via multiplex immunohistochemical staining that revealed irreversible loss of perfusion and enhanced tumour cell death when TPZ was combined with either low oxygen or a NOS inhibitor. Tumour growth rate was reduced by TPZ + NOS inhibition, and tumours previously resistant to TPZ-mediated vascular dysfunction were sensitized by low oxygen breathing. Additional mapping analysis suggests that tumours with reduced vascular-associated stroma may have greater sensitivity to these effects. These results indicate that poorly oxygenated tumour vessels, also being abnormally organized and with inadequate smooth muscle, may be successfully targeted for significant anti-cancer effects by inhibition of NOS and hypoxia-activated prodrug toxicity. This strategy illustrates a novel use of hypoxia-activated cytotoxic prodrugs as vascular targeting agents, and also represents a novel mechanism for targeting tumour vessels.

Highlights

  • Identification of tumour-specific, targetable features for which effective anti-cancer therapeutics can be generated is an important focus in cancer research

  • HT29 xenografts treated with TPZ show no difference in their proportion of perfused vasculature (PF), a small but significant decrease in the proportion of viable tissue (VF) is seen (Fig. 1A, B; Table 1); mass for controls and TPZ-treated tumours is 163.5622.5 mg and 161.567.4 mg respectively

  • We have previously shown through tumour mapping and MRI studies that TPZ can cause catastrophic damage to the central blood vessels of tumour xenografts, killing the dependent tumour cells [8,9,10]

Read more

Summary

Introduction

Identification of tumour-specific, targetable features for which effective anti-cancer therapeutics can be generated is an important focus in cancer research. The variable tumour microenvironment presents opportunities for chemotherapeutic damage, with targets including hypoxic cells and the abnormal tumour vasculature. The presence and importance of hypoxia in tumours has been recognized for more than 50 years [1]. The supply of oxygen to tumours is compromised by low microvessel density, abnormal vascular architecture, low blood oxygenation and slow or stagnant blood flow [2]. Tirapazamine (TPZ; SR4233; 3-amino-1,2,4benzotriazine 1,4-dioxide) is a hypoxic cytotoxin thought to damage poorly oxygenated tumour cells [3]. TPZ has demonstrated an unexpected ability to effect a dose-dependent, large area of vascular dysfunction in the central regions of tumour xenografts [8,9,10]

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