Targeting the tumour microenvironment : characterizing the anti-vascular effects of the hypoxic cytotoxin tirapazamine

Tirapazamine (TPZ) is a bioreductive prodrug with greater toxicity to hypoxic cells in vitro, and anti-vascular activity in tumours grown in vivo in mice. Considerable inter- and intra-tumour heterogeneity occurs in response to the anti-vascular effects of TPZ. The main hypothesis for the work descr...

Full description

Bibliographic Details
Main Author: Baker, Jennifer Hazel Elizabeth
Language:English
Published: University of British Columbia 2010
Online Access:http://hdl.handle.net/2429/23242
id ndltd-UBC-oai-circle.library.ubc.ca-2429-23242
record_format oai_dc
spelling ndltd-UBC-oai-circle.library.ubc.ca-2429-232422018-01-05T17:24:10Z Targeting the tumour microenvironment : characterizing the anti-vascular effects of the hypoxic cytotoxin tirapazamine Baker, Jennifer Hazel Elizabeth Tirapazamine (TPZ) is a bioreductive prodrug with greater toxicity to hypoxic cells in vitro, and anti-vascular activity in tumours grown in vivo in mice. Considerable inter- and intra-tumour heterogeneity occurs in response to the anti-vascular effects of TPZ. The main hypothesis for the work described in this thesis is that features of the tumour microenvironment confer tumour sensitivity to TPZ-mediated vascular dysfunction. Tumours exhibiting less sensitivity to the anti-vascular effects of TPZ had evidence of greater pre-treatment vascular function, including greater blood flow or permeability measured using DCE-MRI derived biomarkers and tumour mapping data of high molecular weight fluorescent dyes injected intravenously. Modulation of nitric oxide (NO) levels decreased the density of perfused blood vessels and sensitized tumours to the anti-vascular effects of TPZ. Additional vascular phenotype features such as relatively poor vascular maturity were also found to correlate with greater tumour sensitivity to TPZ-mediated vascular dysfunction. Greater toxicity of TPZ to hypoxic cells in vitro led to the hypothesis that blood oxygenation may have an impact on tumour sensitivity to TPZ-mediated vascular dysfunction in vivo. Tumours from mice that had moderate bleeding-induced anemia or that were breathing lowered (7-10%) oxygen were sensitized to the anti-vascular effects of TPZ. In vitro assays showed that human microvascular tube structures are sensitive to damage by TPZ at clinically relevant concentrations and oxygen levels. TPZ may be reduced by cellular nitric oxide synthase (NOS), and NOS is competitively inhibited by TPZ to result in decreased amounts of NO. Enhanced anti-cancer effects have previously been observed for other vascular disrupting agents (VDAs) and hypoxic cytotoxins when combined with NOS inhibitors. These findings led to studies presented in this thesis, which show that TPZ-mediated vascular dysfunction is enhanced by co-administration with a NOS inhibitor, and this combined activity can lead to reduced cancer growth. Results from this thesis suggest that features of the tumour microenvironment, including tumour vascular phenotype, blood oxygenation and tumour hypoxia impact tumour sensitivity to TPZ-mediated vascular dysfunction. In addition, inhibiting NOS in combination with TPZ is a therapeutically advantageous strategy that merits further investigation. Medicine, Faculty of Graduate 2010-04-01T14:39:37Z 2010-04-01T14:39:37Z 2010 2010-05 Text Thesis/Dissertation http://hdl.handle.net/2429/23242 eng Attribution-NonCommercial-NoDerivatives 4.0 International http://creativecommons.org/licenses/by-nc-nd/4.0/ University of British Columbia
collection NDLTD
language English
sources NDLTD
description Tirapazamine (TPZ) is a bioreductive prodrug with greater toxicity to hypoxic cells in vitro, and anti-vascular activity in tumours grown in vivo in mice. Considerable inter- and intra-tumour heterogeneity occurs in response to the anti-vascular effects of TPZ. The main hypothesis for the work described in this thesis is that features of the tumour microenvironment confer tumour sensitivity to TPZ-mediated vascular dysfunction. Tumours exhibiting less sensitivity to the anti-vascular effects of TPZ had evidence of greater pre-treatment vascular function, including greater blood flow or permeability measured using DCE-MRI derived biomarkers and tumour mapping data of high molecular weight fluorescent dyes injected intravenously. Modulation of nitric oxide (NO) levels decreased the density of perfused blood vessels and sensitized tumours to the anti-vascular effects of TPZ. Additional vascular phenotype features such as relatively poor vascular maturity were also found to correlate with greater tumour sensitivity to TPZ-mediated vascular dysfunction. Greater toxicity of TPZ to hypoxic cells in vitro led to the hypothesis that blood oxygenation may have an impact on tumour sensitivity to TPZ-mediated vascular dysfunction in vivo. Tumours from mice that had moderate bleeding-induced anemia or that were breathing lowered (7-10%) oxygen were sensitized to the anti-vascular effects of TPZ. In vitro assays showed that human microvascular tube structures are sensitive to damage by TPZ at clinically relevant concentrations and oxygen levels. TPZ may be reduced by cellular nitric oxide synthase (NOS), and NOS is competitively inhibited by TPZ to result in decreased amounts of NO. Enhanced anti-cancer effects have previously been observed for other vascular disrupting agents (VDAs) and hypoxic cytotoxins when combined with NOS inhibitors. These findings led to studies presented in this thesis, which show that TPZ-mediated vascular dysfunction is enhanced by co-administration with a NOS inhibitor, and this combined activity can lead to reduced cancer growth. Results from this thesis suggest that features of the tumour microenvironment, including tumour vascular phenotype, blood oxygenation and tumour hypoxia impact tumour sensitivity to TPZ-mediated vascular dysfunction. In addition, inhibiting NOS in combination with TPZ is a therapeutically advantageous strategy that merits further investigation. === Medicine, Faculty of === Graduate
author Baker, Jennifer Hazel Elizabeth
spellingShingle Baker, Jennifer Hazel Elizabeth
Targeting the tumour microenvironment : characterizing the anti-vascular effects of the hypoxic cytotoxin tirapazamine
author_facet Baker, Jennifer Hazel Elizabeth
author_sort Baker, Jennifer Hazel Elizabeth
title Targeting the tumour microenvironment : characterizing the anti-vascular effects of the hypoxic cytotoxin tirapazamine
title_short Targeting the tumour microenvironment : characterizing the anti-vascular effects of the hypoxic cytotoxin tirapazamine
title_full Targeting the tumour microenvironment : characterizing the anti-vascular effects of the hypoxic cytotoxin tirapazamine
title_fullStr Targeting the tumour microenvironment : characterizing the anti-vascular effects of the hypoxic cytotoxin tirapazamine
title_full_unstemmed Targeting the tumour microenvironment : characterizing the anti-vascular effects of the hypoxic cytotoxin tirapazamine
title_sort targeting the tumour microenvironment : characterizing the anti-vascular effects of the hypoxic cytotoxin tirapazamine
publisher University of British Columbia
publishDate 2010
url http://hdl.handle.net/2429/23242
work_keys_str_mv AT bakerjenniferhazelelizabeth targetingthetumourmicroenvironmentcharacterizingtheantivasculareffectsofthehypoxiccytotoxintirapazamine
_version_ 1718582379639996416