Abstract

Hypoxia is a central problem in tumor treatment because hypoxic cells are less sensitive to chemo- and radiotherapy than normoxic cells. Radioresistance of hypoxic tumor cells is due to reduced sensitivity towards low Linear Energy Transfer (LET) radiation. High LET α-emitters are thought to eradicate tumor cells independent of cellular oxygenation. Therefore, the aim of this study was to demonstrate that cell-bound α-particle emitting 213Bi immunoconjugates kill hypoxic and normoxic CAL33 tumor cells with identical efficiency. For that purpose CAL33 cells were incubated with 213Bi-anti-EGFR-MAb or irradiated with photons with a nominal energy of 6 MeV both under hypoxic and normoxic conditions. Oxygenation of cells was checked via the hypoxia-associated marker HIF-1α. Survival of cells was analysed using the clonogenic assay. Cell viability was monitored with the WST colorimetric assay. Results were evaluated statistically using a t-test and a Generalized Linear Mixed Model (GLMM). Survival and viability of CAL33 cells decreased both after incubation with increasing 213Bi-anti-EGFR-MAb activity concentrations (9.25 kBq/ml–1.48 MBq/ml) and irradiation with increasing doses of photons (0.5–12 Gy). Following photon irradiation survival and viability of normoxic cells were significantly lower than those of hypoxic cells at all doses analysed. In contrast, cell death induced by 213Bi-anti-EGFR-MAb turned out to be independent of cellular oxygenation. These results demonstrate that α-particle emitting 213Bi-immunoconjugates eradicate hypoxic tumor cells as effective as normoxic cells. Therefore, 213Bi-radioimmunotherapy seems to be an appropriate strategy for treatment of hypoxic tumors.

Highlights

  • In solid tumors hypoxia results from accelerated proliferation combined with high metabolic activities and poor oxygenation due to insufficient blood supply [1], [2]

  • In CAL33 cells that had been incubated under hypoxic conditions for 3 hours, release from hypoxia and cultivation under normoxic conditions resulted in a decrease of the hypoxia inducible factors (HIFs)-1a signal

  • HIF-1a appeared 30 min after incubation of CAL33 cells under hypoxia and reached maximum expression after 3 h

Read more

Summary

Introduction

In solid tumors hypoxia results from accelerated proliferation combined with high metabolic activities and poor oxygenation due to insufficient blood supply [1], [2]. Radioresistance is supposed to appear at p[O2] ,10 mmHg [4], [5]. It can be quantified by the oxygen enhancement ratio (OER) expressing the ratio of radiation dose required under hypoxia and normoxia to produce the same biological effect [6]. Lower sensitivity towards ionizing radiation is explained by the oxygen effect [7]. Hypoxia-related tumor radioresistance is triggered by biological signaling pathways. HIF-1 induces cytokines, which are involved in protection of endothelial cells from the effects of radiation [9]. HIF-1 activation leads to an increased resistance to radio- and chemotherapy, increased local aggressive growth and an increased risk of metastatic disease [7], [8]

Objectives
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