The University of Southampton
University of Southampton Institutional Repository

Mathematical modeling predicts synergistic antitumor effects of combining a macrophage-based, hypoxia-targeted gene therapy with chemotherapy

Mathematical modeling predicts synergistic antitumor effects of combining a macrophage-based, hypoxia-targeted gene therapy with chemotherapy
Mathematical modeling predicts synergistic antitumor effects of combining a macrophage-based, hypoxia-targeted gene therapy with chemotherapy
Tumor hypoxia is associated with low rates of cell proliferation and poor drug delivery, limiting the efficacy of many conventional therapies such as chemotherapy. Because many macrophages accumulate in hypoxic regions of tumors, one way to target tumor cells in these regions could be to use genetically engineered macrophages that express therapeutic genes when exposed to hypoxia. Systemic delivery of such therapeutic macrophages may also be enhanced by preloading them with nanomagnets and applying a magnetic field to the tumor site. Here, we use a new mathematical model to compare the effects of conventional cyclophosphamide therapy with those induced when macrophages are used to deliver hypoxia-inducible cytochrome P450 to locally activate cyclophosphamide. Our mathematical model describes the spatiotemporal dynamics of vascular tumor growth and treats cells as distinct entities. Model simulations predict that combining conventional and macrophage-based therapies would be synergistic, producing greater antitumor effects than the additive effects of each form of therapy. We find that timing is crucial in this combined approach with efficacy being greatest when the macrophage-based, hypoxia-targeted therapy is administered shortly before or concurrently with chemotherapy. Last, we show that therapy with genetically engineered macrophages is markedly enhanced by using the magnetic approach described above, and that this enhancement depends mainly on the strength of the applied field, rather than its direction. This insight may be important in the treatment of nonsuperficial tumors, where generating a specific orientation of a magnetic field may prove difficult. In conclusion, we demonstrate that mathematical modeling can be used to design and maximize the efficacy of combined therapeutic approaches in cancer
0008-5472
2826-2837
Owen, Markus R.
3ef2b922-56a5-4758-a95d-135cbc1ccead
Stamper, I. Johanna
3e2545f6-875a-4460-8fa8-5dc47a1b9d2a
Muthana, Munitta
6cee15cb-6414-4575-b577-ef294c933c45
Richardson, Giles W.
3fd8e08f-e615-42bb-a1ff-3346c5847b91
Dobson, Jon
af3c1ce8-9631-407a-ad2f-be91a3a31b21
Lewis, Claire E.
22d58205-2f12-4465-850f-5db245d1904e
Byrne, Helen M.
3534c977-cb9a-4b70-b22a-25736767c47c
Owen, Markus R.
3ef2b922-56a5-4758-a95d-135cbc1ccead
Stamper, I. Johanna
3e2545f6-875a-4460-8fa8-5dc47a1b9d2a
Muthana, Munitta
6cee15cb-6414-4575-b577-ef294c933c45
Richardson, Giles W.
3fd8e08f-e615-42bb-a1ff-3346c5847b91
Dobson, Jon
af3c1ce8-9631-407a-ad2f-be91a3a31b21
Lewis, Claire E.
22d58205-2f12-4465-850f-5db245d1904e
Byrne, Helen M.
3534c977-cb9a-4b70-b22a-25736767c47c

Owen, Markus R., Stamper, I. Johanna, Muthana, Munitta, Richardson, Giles W., Dobson, Jon, Lewis, Claire E. and Byrne, Helen M. (2011) Mathematical modeling predicts synergistic antitumor effects of combining a macrophage-based, hypoxia-targeted gene therapy with chemotherapy. Cancer Research, 71 (8), 2826-2837. (doi:10.1158/0008-5472.CAN-10-2834). (PMID:21363914)

Record type: Article

Abstract

Tumor hypoxia is associated with low rates of cell proliferation and poor drug delivery, limiting the efficacy of many conventional therapies such as chemotherapy. Because many macrophages accumulate in hypoxic regions of tumors, one way to target tumor cells in these regions could be to use genetically engineered macrophages that express therapeutic genes when exposed to hypoxia. Systemic delivery of such therapeutic macrophages may also be enhanced by preloading them with nanomagnets and applying a magnetic field to the tumor site. Here, we use a new mathematical model to compare the effects of conventional cyclophosphamide therapy with those induced when macrophages are used to deliver hypoxia-inducible cytochrome P450 to locally activate cyclophosphamide. Our mathematical model describes the spatiotemporal dynamics of vascular tumor growth and treats cells as distinct entities. Model simulations predict that combining conventional and macrophage-based therapies would be synergistic, producing greater antitumor effects than the additive effects of each form of therapy. We find that timing is crucial in this combined approach with efficacy being greatest when the macrophage-based, hypoxia-targeted therapy is administered shortly before or concurrently with chemotherapy. Last, we show that therapy with genetically engineered macrophages is markedly enhanced by using the magnetic approach described above, and that this enhancement depends mainly on the strength of the applied field, rather than its direction. This insight may be important in the treatment of nonsuperficial tumors, where generating a specific orientation of a magnetic field may prove difficult. In conclusion, we demonstrate that mathematical modeling can be used to design and maximize the efficacy of combined therapeutic approaches in cancer

Text
Cancer research 2011 Owen-1.pdf - Other
Download (3MB)

More information

Published date: March 2011
Organisations: Applied Mathematics

Identifiers

Local EPrints ID: 202887
URI: http://eprints.soton.ac.uk/id/eprint/202887
ISSN: 0008-5472
PURE UUID: 3bf01bba-4198-46c1-bf65-655f2913911d
ORCID for Giles W. Richardson: ORCID iD orcid.org/0000-0001-6225-8590

Catalogue record

Date deposited: 10 Nov 2011 11:29
Last modified: 15 Mar 2024 03:33

Export record

Altmetrics

Contributors

Author: Markus R. Owen
Author: I. Johanna Stamper
Author: Munitta Muthana
Author: Jon Dobson
Author: Claire E. Lewis
Author: Helen M. Byrne

Download statistics

Downloads from ePrints over the past year. Other digital versions may also be available to download e.g. from the publisher's website.

View more statistics

Atom RSS 1.0 RSS 2.0

Contact ePrints Soton: eprints@soton.ac.uk

ePrints Soton supports OAI 2.0 with a base URL of http://eprints.soton.ac.uk/cgi/oai2

This repository has been built using EPrints software, developed at the University of Southampton, but available to everyone to use.

We use cookies to ensure that we give you the best experience on our website. If you continue without changing your settings, we will assume that you are happy to receive cookies on the University of Southampton website.

×