Cookies on this website

We use cookies to ensure that we give you the best experience on our website. If you click 'Accept all cookies' we'll assume that you are happy to receive all cookies and you won't see this message again. If you click 'Reject all non-essential cookies' only necessary cookies providing core functionality such as security, network management, and accessibility will be enabled. Click 'Find out more' for information on how to change your cookie settings.

Prof. Sibson together with her co-applicants Prof. Anthony, Dr Campbell and Prof. Middleton have now been awarded a second MRC DPFS grant, for £3.3 million, to acquire further preclinical data to support the case for clinical translation, to develop the mutTNF production for human use and to undertake pre-clinical toxicology.

Histology and magnetic resonance images from a mouse with breast cancer brain metastases, injected intravenously 2 hours previously with a TNFR1-selective mutant TNF protein. 

3 items in the picture:

Cresyl violet stained brain section showing location of a micrometastasis. 

Adjacent brain section stained with Hanker-Yates demonstrates HRP extravasation (brown staining) and, hence, BBB breakdown selectively at the site of metastasis. 

Post-contrast (gadolinium-DTPA) image from the same mouse showing extravasation of contrast at the metastatic site (hyperintensity) confirming local BBB breakdown. Note, the BBB remains intact in the rest of the brain.
Figure caption: Histology and magnetic resonance images from a mouse with breast cancer brain metastases, injected intravenously 2 hours previously with a TNFR1-selective mutant TNF protein. (A) Cresyl violet stained brain section showing location of a micrometastasis (boxed region and inset). (B) Adjacent brain section stained with Hanker-Yates demonstrates HRP extravasation (brown staining) and, hence, BBB breakdown selectively at the site of metastasis. (C) Post-contrast (gadolinium-DTPA) image from the same mouse showing extravasation of contrast at the metastatic site (hyperintensity) confirming local BBB breakdown. Note, the BBB remains intact in the rest of the brain.

Metastasis to the brain is a major challenge in cancer therapy and prognosis is poor. The blood-brain barrier (BBB) is a significant impediment to both early detection and effective treatment, as it is intact during the early stages of tumour development and only heterogeneously permeable at later stages. The presence of an intact BBB excludes both therapeutic and diagnostic imaging agents from metastases. However, Prof. Sibson and her colleagues have shown in mouse models that intravenous injection of the cytokine tumour necrosis factor (TNF) selectively permeabilises the BBB at sites of micrometastases in the brain, leaving the rest of the BBB intact (Connell et al., 2013, Journal of the National Cancer Institute). This permeabilisation is mediated primarily though activation of the TNF type 1 receptor (TNFR1), and it enables delivery of otherwise excluded therapies.

Although the permeabilising activity of TNF was shown to occur at concentrations below the maximum tolerated dose, to facilitate clinical translation Prof. Sibson and her colleagues wanted to reduce the potential for toxicity further. To this end, under an initial MRC Developmental Pathway Funding Scheme (DPFS) award, they have now developed a TNFR1-selective mutein (mutTNF) that will reduce the required dose even further and eliminate toxicity associated with activation of other TNF receptors. They have demonstrated binding specificity and permeabilising action of the mutTNF, both in vitro and in vivo, and have obtained preliminary toxicology data showing no contraindications to clinical translation.

On the basis of the above developments, Prof. Sibson together with her co-applicants Prof. Anthony, Dr Campbell and Prof. Middleton have now been awarded a second MRC Developmental Pathway Funding Scheme grant, for £3.3 million, to acquire further preclinical data to support the case for clinical translation, to develop the mutTNF production for human use and to undertake pre-clinical toxicology. At the end of this project, they will be ready to undertake a Phase I/IIa clinical trial in patients with brain metastases.

This solution to the problem of an intact BBB in brain metastasis, will open a treatment window that currently does not exist in a substantial number of cancer patients.

Similar stories

Robert Watson Awarded ACP McElwain and Presidents Prize

Dr Robert Watson has won one of three McElwain and President's Prizes from the Association of Cancer Physicians for his DPhil work undertaken with Professor Ben Fairfax.

Prof Anna Schuh launches first private specialised haematology clinic and laboratory in sub-Saharan Africa.

SerenOx Africa aims to address diagnostic gaps for common blood disorders through a testing facility for key underserved patient populations in Tanzania. This facility will also aim to provide early cancer detection for high-risk patients.

Professor Eileen Parkes appointed to lead Oxford’s Experimental Cancer Medicine Centre

Eileen Parkes, Associate Professor in Innate Tumour Immunology in the Department of Oncology, will lead the expansion of the centre’s programmes in early drug development and biomarker research.

Oxford to launch UK’s first trials unit dedicated to conducting precision prevention and early detection studies

Oxford researchers have been given a £1 million boost to support their strategy of developing cancer prevention treatments and early diagnostic tools for people at high risk of cancer.

Multi-cancer blood test shows real promise in NHS trial

An NHS trial of a new blood test for more than 50 types of cancer correctly revealed two out of every three cancers in more than 5,000 people who had visited their GP with suspected symptoms, in England or Wales. The test also correctly identified the original site of cancer in 85% of those cases.