2011 - Cancer Research UK Cambridge Institute

Transcription

2011 - Cancer Research UK Cambridge Institute
Cambridge Research Institute Scientific Report 2011
Cambridge
Research
Institute
Scientific Report 2011
Cancer Research UK Cambridge Research Institute
Li Ka Shing Centre
Robinson Way
Cambridge CB2 0RE
Telephone +44 (0) 1223 404209
www.cambridgecancer.org.uk
Cover images
Top
Primary human prostate cells grown in culture in
order to identify progenitor cells. Cells were
dissociated from benign patient tissue and were
seeded on growth medium, giving rise to the large
colony shown. The cells were stained for human
epithial markers cytokeratin 5 (green) and
cytokeratin 18 (red). Nuclear staining with DAPI
(blue). Image provided by Ajoeb Baridi (Stingl and
Neal laboratories).
Bottom
Podocyte cells wrap around the capillaries of the
glomerulus within the kidney and are a core element
of the filtration barrier that is the first stage in
removing waste products from the blood to form
urine. Podocytes play a critical role in the constant
turnover of the glomerular basement membrane as
well as endothelium maintenance by secreting
extracellular matrix components, vascular endothelial
growth factor (VEGF) and many other factors.
These cells stain positive for the recently described
TOR autophagy spatial coupling compartment
(TASCC). This intracellular ‘factory’ compartment
may conceivably allow them to maintain their high
levels of secretion. Image provided by Andy Young
(Narita laboratory).
Cancer Research UK Cambridge Research Institute
Scientific Report 2011
Editor: Laura Blackburn
Page setting: Charles D N Thomson
Cancer Research UK Cambridge Research Institute
Li Ka Shing Centre
Robinson Way
Cambridge CB2 0RE
ISSN 1756-8994
Copyright © 2011 Cancer Research UK
Scientific Report 2011
Cambridge Research Institute
Confocal image of the TOR-autophagy spatial coupling
compartment (TASCC) in Ras-induced senescent IMR90 cells.
p62 (red), trans-Golgi network (pink) and Vimentin (green)
were immunolabelled. DNA was counterstained with DAPI.
Image provided by Masako Narita (Narita laboratory).
2 | Cambridge Research Institute Scientific Report 2011
Contents
Director’s Introduction Research Highlights 5
7
Research Groups
Shankar Balasubramanian
12
Chemical Biology of Nucleic Acids
James Brenton 14
Functional Genomics of Ovarian Cancer
Kevin Brindle
16
Molecular Imaging of Cancer
Carlos Caldas
18
Functional Genomics of Breast Cancer
Jason Carroll
20
Nuclear Receptor Transcription
Douglas Fearon
22
Tumour Immunology and the FAP+ Stromal Cell
Fanni Gergely
24
Centrosomes, Microtubules and Cancer
John Griffiths 26
Magnetic Resonance Imaging and Spectroscopy (MRI
and MRS)
Duncan Jodrell
28
Pharmacology and Drug Development
Florian Markowetz
30
Computational Biology
Gillian Murphy
32
Proteases and the Tumour Microenvironment
Adele Murrell
34
Genomic Imprinting and Cancer
Masashi Narita
36
Mechanisms of Cellular Senescence
David Neal
38
Prostate Research
Duncan Odom
40
Regulatory Systems Biology
Bruce Ponder
42
Polygenic Predisposition to Breast Cancer
Nitzan Rosenfeld
44
Molecular and Computational Diagnostics
John Stingl
46
Mammary Stem Cell Biology
Simon Tavaré
48
Computational Biology and Statistics
David Tuveson
50
Tumour Modelling and Experimental Medicine
Fiona Watt
52
Keratinocytes in Normal Tissue and in Tumours
Doug Winton
54
Cancer and Intestinal Stem Cells
Core Facilities
Matthew Eldridge
Bioinformatics
Allen Hazlehurst
Biological Resources Unit
Bob Geraghty
Biorepository and Cell Services
Jane Gray
Equipment Park
Richard Grenfell
Flow Cytometry
James Hadfield
Genomics
Will Howat
Histopathology and In Situ Hybridisation
Stefanie Reichelt
Light Microscopy
Donna Smith
Pharmacokinetics and Pharmacodynamics
Kevin Brindle and John Griffiths
Pre-clinical Imaging
Clive D’Santos
Proteomics
Institute Information
Research Publications External Funding Seminars and Conferences Cambridge Cancer Centre
Outreach and Fundraising
Academic Administration
Institute Administration
Theses Contact Details
58
59
60
61
62
63
64
65
66
67
68
72
84
86
88
89
90
92
95
96
Contents | 3
The Cancer Research UK Cambridge Research Institute
The Institute’s location on the Cambridge Biomedical Campus
facilitates collaborations with Addenbrooke’s Hospital and
other Institutes and University of Cambridge Departments on
the site.
4 | Cambridge Research Institute Scientific Report 2011
Director’s Introduction
Professor Sir Bruce Ponder
This year, our fifth since the opening of the
Institute, we had our first Quinquennial Review
by a strong international panel. The results were
extremely positive. We were congratulated
on our ‘truly remarkable achievements’, and in
only four years the CRI was considered to be
‘a model for how to enable the translation of
elegant basic science into potentially powerful
clinical discovery’. I will step down as Director
within the next two years, which will bring
the opportunities that come with change; but
I hope that with this endorsement from the
Review Committee, the shape of the CRI as
an Institute that builds from a platform of basic
science towards innovative practical application
is firmly established. The Review also endorsed
the view that, if it is to fulfil its potential, the
CRI must not be developed in isolation but in
close partnership with the University, the NHS,
and other research funders in Cambridge. This
partnership is already developing well, and
strengthening it further is a priority for the
immediate future. To give one example, over
half of our group leaders have collaborative
projects with the Sanger Institute or the
European Bioinformatics Institute at the nearby
Hinxton campus. This year Duncan Odom was
appointed as joint faculty at the Sanger Institute,
while Ultan McDermott, currently a Cancer
Research UK Clinician Scientist at the Sanger, has
held an appointment in Oncology since 2009.
The CRI was launched in 2007 with a very
strong group of junior group leaders, and this
year another of these, James Brenton, achieved
tenure. James is a medical oncologist, well
trained in cancer biology following a PhD
as a CRC Clinical Fellow with Azim Surani,
and a CRC Senior Clinical Fellowship. He
has an increasing international reputation
for his research into the genomics of ovarian
cancer and its practical clinical applications.
He will further strengthen the team of CRI
Group Leaders who are both active clinicians
and leaders of laboratory research that is
internationally competitive and directly related
to their clinical activity. Ovarian cancer will
become established as one of the ‘focus’ cancers
through which we will build the bridge between
the CRI and the clinic.
Because of the impending change of Director,
senior recruitments to the CRI have been on
hold. Nevertheless, we are moving forward
with several new developments. Kevin Brindle
has taken a major step in bringing his novel 13C
hyperpolarised MRI imaging to the clinic. A
£4.3M Strategic Translation Award from the
Wellcome Trust will provide the running costs
for a 5-year clinical programme which will be
led by Kevin Brindle and Ferdia Gallagher, a
Cancer Research UK clinical scientist, in the
Radiology Department in the Cambridge
University Teaching Hospital, on the same
site as the CRI. Initial studies will focus on
lymphoma, glioma and breast cancer. We are
also planning for a possible future expansion
and consolidation of bioinformatics. Both the
Quinquennial Review and an outstandingly
successful review of our bioinformatics core
endorsed a proposal that we should open one
half of the third floor – currently mothballed –
to consolidate and expand our bioinformatics
capability. We have been running at a wet:dry
lab ratio of about 88:12 in terms of staff, which
has proved nowhere near enough to support
our extensive programmes in genomics,
transcriptional regulation, molecular pathology
and imaging. Many groups have recruited their
own additional bioinformatics staff, creating a
diaspora that is inefficient in use of space and
unhelpful for the training and development of
the staff themselves. We are preparing plans
so that, should the new director agree with this
assessment, work can proceed with a minimum
delay.
We have formed a link with the Haematological
Oncology Department in the hospital, to
extend their excellent regional molecular
diagnostic service to solid tumours. Although
not successful in our bid to be a Genomics
Centre in the Cancer Research UK Stratified
Medicines Initiative, we have set up a nextgeneration sequencing pipeline for a wide range
of gene mutations within a set of samples. This
will contribute comparative data to the Initiative,
and kick-start the molecular pathology that
is essential to underpin our programmes in
experimental cancer medicine.
Director’s Introduction | 5
Cambridge University has designated a number
of ‘Strategic Initiatives’ that are intended to
catalyse interactions across the University that
will exploit the unusual range of expertise that
is available, and that may be a future focus
for fundraising. ‘Cancer’ is one of the seven
themes so far selected in competition. This has
provided funding to appoint a senior facilitator,
Dr Kenneth Seamon, formerly Global VP for
Drug Development at Amgen. He is leading
a process for the Cancer Centre that will first
provide a clear and comprehensive description
of our activities in different areas such as early
detection or molecular imaging or quantitative
biology. This will provide a ‘shop window’ for
potential pharma partners, and progress to
successive levels of scientific interaction, joint
projects (stimulated by Cancer Centre pumppriming funds) and responses to large-scale
funding opportunities.
In parallel, we have agreed with the hospital
another senior appointment, reporting jointly
to me as Director of the Cancer Centre and
to the CEO of the NHS Trust, to develop joint
planning between the hospital and the Cancer
Centre. Simon Oberst has a background in
accountancy and long experience of interaction
with the NHS in a senior position within a
national medical charity. Together with the
NHS Clinical Director of the Cancer Service,
he is leading a detailed analysis benchmarked
to objective criteria of our current service
provision in cancer, and of the support that
it provides for the clinical components of our
major research themes. We will identify the
weaknesses and opportunities and set priorities
to address them, informed by a clear picture of
the financial implications.
Now that the CRI is launched and its main
research directions are clear, we hope that
these two interlocking initiatives will inform
the strategy for the future development of the
Cancer Centre.
Professor John Griffiths with his
Gold Medal of the International
Society for Magnetic Resonance in
Medicine.
We have continued our annual round of CRI
activities. The CRI Annual Symposium on
‘Unanswered Questions’ was held in November
on the topic of Transcription. We attracted a
star line-up of speakers and a capacity audience.
The format of half-day sessions on each of four
topics, each session ending with a structured
45-minute discussion of the ‘Unanswered
Questions’, was again so successful that lunches
were taken at 2 pm on both days. Next year
the topic will be ‘Unanswered Questions in
Cancer Sequencing’.
The Cancer Centre Annual Symposium in June
attracted over 300 participants from within
Cambridge, with talks spanning many disciplines.
The keynote was given by Robert Weinberg
6 | Cambridge Research Institute Scientific Report 2011
from MIT, and was a great success both
because of the lecture itself, but also because
Dr Weinberg so enjoyed his two days with us
that he is returning in April for a four-week
sabbatical.
Finally, congratulations to the CRI members
who won prizes or distinctions during the year.
Among our post-docs, Daniele Perna (Tuveson
lab) won an International Fellowship from the
Italian Association for Cancer Research and
a Marie Curie European Fellowship, and Hiro
Fujiwara (Watt lab) won the first prize for
his poster at the EMBO Conference on Stem
Cell Research. Among PhD students, Natalie
Cook (Tuveson lab) won the McElwain Prize
of the Association of Cancer Physicians, Simon
Buczacki (Winton lab) won both the best
oral presentation prize and the top poster
prize at the Association of Coloproctology
of Great Britain and Ireland Annual Meeting,
Michelle Ward (Odom lab) has won an EMBO
short-term Fellowship, Roheet Rao (Neal
lab) has won an AACR Scholar-in-Training
Award; three students – Sarah Kozar (Winton
lab), Dominic Schmidt (Odom/Carroll labs)
and Chris Tape (Murphy lab) – have given
invited platform presentations at international
meetings. Among our Group Leaders, Simon
Tavaré was elected to the Royal Society, and
Shankar Balasubramanian to the Academy of
Medical Sciences. David Neal was elected to
the European Academy of Cancer Sciences and
Carlos Caldas was elected Fellow of the Society
of Biology. John Griffiths was awarded the Gold
Medal of the International Society for Magnetic
Resonance in Medicine. The Gold Medal is
the highest distinction in this field, and reflects
John’s many distinguished contributions since
he introduced the use of magnetic resonance
spectroscopy to evaluate cancers over 30 years
ago. Nitzan Rosenfeld won the Young Award
CNAPS VII for the best free communication at
the CNAPS meeting in Madrid, and finally Bruce
Ponder was invited to become the President of
the British Association for Cancer Research.
Research Highlights
Sir JH, Barr AR, Nicholas AK, Carvalho OP,
Khurshid M, Sossick A, Reichelt S, D’Santos C,
Woods CG, Gergely F. A primary microcephaly
protein complex forms a ring around parental
centrioles. Nat Genet 2011; 43: 1147-53
Loss of genomic stability is an attribute of many
cancers. Equal partitioning of chromosomes
between two daughter cells is critical for maintaining
genome integrity. This process is largely dependent
on a functional bipolar mitotic spindle. The two
poles of the spindle are formed by proteinaceous
organelles called centrosomes. Concerted
duplication of chromatin with the centrosome
ensures that a cell enters mitosis containing two
functional centrosomes. Centrosome duplication
is a multi-step event involving several regulators
and structural components. We have discovered a
new protein complex formed by two centrosomal
proteins, CEP63 and CEP152, that is required for
initiating centrosome duplication. Cells lacking
CEP63 display deregulated centrosome duplication
cycles leading to mitotic spindle abnormalities,
aberrant growth and impaired clonal expansion.
Indeed, weak hypomorphic mutations in the CEP63
gene cause mental retardation and reduced brain
size in humans, indicative of a vital role of CEP63
and perhaps the centrosome in neural stem cells.
Bapiro TE, Richards FM, Goldgraben MA, Olive KP,
Madhu B, Frese KK, Cook N, Jacobetz MA,
Smith DM, Tuveson DA, Griffiths JR, Jodrell DI.
A novel method for quantification of gemcitabine
and its metabolites 2′,2′-difluorodeoxyuridine
and gemcitabine triphosphate in tumour tissue by
LC‑MS/MS: comparison with 19F NMR spectroscopy.
Cancer Chemother Pharmacol 2011; 68: 1243-53A
This paper describes for the first time that
gemcitabine triphosphate, the active metabolite
of gemcitabine, has been measured in tumour
tissue. Gemcitabine is a drug used in the treatment
of pancreatic, non-small cell lung, ovary, bladder
and breast cancer. The gemcitabine compound
is a prodrug that is metabolised into its active
components in the body, and these metabolites
work by interfering with DNA synthesis in tumour
cells. The activity of the drug is limited by poor drug
delivery, however giving gemcitabine and a drug that
depletes the tumour stroma improves both drug
delivery and its efficacy. Developing methods to
measure the levels of gemcitabine and metabolites in
tumours, therefore, enables researchers to test drug
delivery in tumours and assess whether combination
treatment strategies increase the amount of active
drug reaching the tumour, increasing efficacy. The
authors compared 19F NMR and LC-MS/MS (liquid
chromatography-mass spectroscopy) methods
and found a sensitive LC-MS/MS method that
could measure the levels of gemcitabine and its
metabolites in tumour tissue. The advantage of
the method is that it requires only 10 mg of tissue,
meaning that multiple areas from the same tumour
can be analysed, leaving tissue spare for other assays.
Yuan Y, Savage RS, Markowetz F. Patient-specific
data fusion defines prognostic cancer subtypes.
PLoS Comput Biol 2011; 7: e1002227
The goal of personalised medicine is to develop
accurate diagnostic tests that identify patients who
can benefit from targeted therapies. To achieve
this goal it is necessary to stratify cancer patients
into homogeneous subtypes according to which
molecular aberrations their tumours exhibit.
Prominent approaches for subtype definition
combine information from different molecular levels,
for example data on DNA copy number changes
with data on mRNA expression changes. This is
called data fusion. We contribute to this field by
proposing a unified model that fuses different data
types, finds informative features and estimates the
number of subtypes in the data. The main strength
of our model comes from the fact that we assess
for each patient whether the different data agree on
a subtype or not. Competing methods combine the
data without checking for concordance of signals.
On a breast cancer and a prostate cancer data set
we show that concordance of signals has a strong
influence on subtype definition and that our model
allows us to define prognostic subtypes that would
have been missed otherwise.
Narita M, Young AR, Arakawa S, Samarajiwa SA,
Nakashima T, Yoshida S, Hong S, Berry LS,
Reichelt S, Ferreira M, Tavaré S, Inoki K,
Shimizu S, Narita M. Spatial coupling of mTOR and
autophagy augments secretory phenotypes. Science
2011; 332: 966-70
Protein synthesis and autophagic degradation
are typically regulated in an opposite manner by
TOR (Target of Rapamycin), which senses cellular
nutrients as the master regulator of protein
metabolism. This paper identified a unique
cellular compartment, the TOR-autophagy spatial
coupling compartment (TASCC), where mTOR
and autolysosomes (the end stage of autophagy)
Research Highlights | 7
are both enriched. Because mTOR inhibits the
initial step of autophagy, and amino acids (end
products of autophagy) activate mTOR, TASCC
formation allows simultaneous activation of anabolic
(mediated by mTOR) and catabolic (autophagy)
processes. Such locally active protein turnover
facilitates rapid en masse production of secretory
proteins during oncogene-induced senescence.
Massie CE, Lynch A, Ramos-Montoya A, Boren J,
Stark R, Fazli L, Warren A, Scott H, Madhu B,
Sharma N, Bon H, Zecchini V, Smith DM,
Denicola GM, Mathews N, Osborne M, Hadfield J,
Macarthur S, Adryan B, Lyons SK, Brindle KM,
Griffiths J, Gleave ME, Rennie PS, Neal DE,
Mills IG. The androgen receptor fuels prostate
cancer by regulating central metabolism and
biosynthesis. EMBO J 2011; 30: 2719-33
Prostate cancer is the most common solid cancer
in men and accounts for 37,000 new cases and
10,000 deaths each year in the UK. Its growth and
development is strongly dependent on signalling
by the male sex hormone testosterone through its
receptor (the androgen receptor, AR). In advanced
cases, the standard treatment is to reduce the
levels of testosterone and most tumours respond,
although eventually most of them recur. Even in
such castration resistant prostate cancer (CRPC),
continued signalling through the AR is critically
important. Prior to this study we did not know the
binding sites for the AR within the genome, and which
genes were most important in the continued growth
of prostate cells. We carried out a genome-wide,
unbiased study looking for these genes and binding
sites. A major discovery was that the AR coordinates
a strong metabolic response and is responsible for the
Warburg effect, where cancer cells produce energy
by glycolysis, despite normal levels of oxygen. We
also discovered a novel gene CAMKK2 (calcium /
calmodulin kinase kinase 2), which is overexpressed in
human CRPC. We showed that blocking CAMKK2
can reduce prostate cancer growth in model systems.
We now believe that on the back of this work,
studies in man are justified in terms of inhibiting
metabolic pathways and offer a new approach to the
management of this disease.
Kutter C, Brown GD, Goncalves A, Wilson MD,
Watt S, Brazma A, White RJ, Odom DT. Pol III
binding in six mammals shows conservation among
amino acid isotypes despite divergence among
tRNA genes. Nat Genet 2011; 43: 948-55.
RNA polymerase III (Pol III) transcription of
tRNA genes is essential for generating the tRNA
adaptor molecules that link genetic sequence and
protein translation. By mapping Pol III occupancy
genome-wide in mouse, rat, human, macaque, dog
and opossum livers, we found that Pol III binding
to individual tRNA genes varies substantially in
strength and location. However, when we took
into account tRNA redundancies by grouping Pol III
occupancy into 46 anticodon isoacceptor families or
21 amino acid-based isotype classes, we discovered
8 | Cambridge Research Institute Scientific Report 2011
strong conservation. Similarly, Pol III occupancy
of amino acid isotypes is almost invariant among
transcriptionally and evolutionarily diverse tissues in
mouse. Thus, synthesis of functional tRNA isotypes
has been highly constrained, although the usage of
individual tRNA genes has evolved rapidly.
DeNicola GM, Karreth FA, Humpton TJ,
Gopinathan A, Wei C, Frese K, Mangal D, Yu KH,
Yeo CJ, Calhoun ES, Scrimieri F, Winter JM,
Hruban RH, Iacobuzio-Donahue C, Kern SE,
Blair IA, Tuveson DA. Oncogene-induced Nrf2
transcription promotes ROS detoxification and
tumorigenesis. Nature 2011; 475: 106-9
The role of reactive oxygen species (ROS) in
carcinogenesis is controversial, as ROS can promote
both mutagenesis and cellular senescence. This
paper shows that near endogenous expression of
Myc and oncogenic Kras and Braf decreases ROS by
inducing expression of the transcription factor Nrf2.
Nrf2 is a master regulator of cellular detoxification,
by directing the coordinate transcription of many
genes involved in the synthesis and recycling of
glutathione, thioredoxin, and endogenous and
exogenous toxins such as heme and certain
chemicals. During Kras-driven transformation, Nrf2
is a requisite pathway both in cell culture and in
developing lung and pancreatic neoplasms. Using
alternative pharmacological approaches, we also
found that depleting cells’ glutathione during tumour
initiation blocked the proliferation of incipient
malignant lung and pancreatic cells. Therefore, the
Nrf2 pathway is a new pathway to consider for
therapeutic intervention in certain malignancies.
Fujiwara H, Ferreira M, Donati G, Marciano DK,
Linton JM, Sato Y, Hartner A, Sekiguchi K,
Reichardt LF, Watt FM. The basement membrane
of hair follicle stem cells is a muscle cell niche. Cell
2011; 144: 577-89
The importance of microenvironmental (“niche”)
signals in regulating stem cell behaviour is well
established. However, the concept that stem cells
provide a niche for neighbouring cells is quite new.
We show that by depositing the ECM protein
nephronectin in the basement membrane,
hair follicle stem cells stimulate neighbouring
mesenchymal cells to differentiate into the muscle
that controls whether body hairs stand on end.
Nephronectin is a Wnt target gene and there is
aberrant deposition of nephronectin and remodelling
of the stroma in Wnt-driven hair follicle tumours.
March HN, Rust AG, Wright NA, Ten Hoeve J,
de Ridder J, Eldridge M, van der Weyden L,
Berns A, Gadiot J, Uren A, Kemp R, Arends MJ,
Wessels LF, Winton DJ, Adams DJ. Insertional
mutagenesis identifies multiple networks of
cooperating genes driving intestinal tumorigenesis.
Nat Genet 2011; 43: 1202-9
Here we describe exploiting random insertional
mutagenesis by transposable DNA elements and
high throughput sequencing to identify which
reduced microenvironment (see figure). We believe
that this will be a sensitive probe for detecting very
early responses of tumours to treatment, since we
know that therapeutic agents can have a dramatic
and early effect on tumour redox status.
genes drive cancer development in the intestine.
The surprise finding from this essentially unbiased
approach is that many hundreds of genes are
potentially implicated. Mechanistically this is most
likely explained by ongoing selection for mutations
that modulate the level of available signalling of
known oncogenic pathways. The results raise
important questions concerning the extent to
which cancers develop due to a multiplicity of such
fine-tuning mutations as opposed to a small number
of known driver genes with large effects.
Hegde NS, Sanders DA, Rodriguez R,
Balasubramanian S. The transcription factor
FOXM1 is a cellular target of the natural product
thiostrepton. Nat Chem 2011; 3: 725-31
This paper shows how the antibiotic thiostrepton,
which was first discovered in bacteria, blocks
the activity of FOXM1, a protein that attaches to
particular stretches of DNA and triggers the growth
and division of cells, as well as tumour angiogenesis.
Higher levels of FOXM1 are present in breast
cancer cells. Therefore, blocking the activity of
FOXM1 could prevent the development of cancer
at an early stage as well as block cancer growth and
spread. While thiostrepton doesn’t have all of the
right properties to be used as a cancer drug, the
information will help researchers to design drugs
based on its structure that can block FOXM1.
Robinson JL, Macarthur S, Ross-Innes CS,
Tilley WD, Neal DE, Mills IG, Carroll JS.
Androgen receptor driven transcription in
molecular apocrine breast cancer is mediated by
FoxA1. EMBO J 2011; 30: 3019-27
This paper explored a recently characterised breast
cancer subtype called molecular apocrine tumours.
These are unusual in that they express the classic
ER target genes, but they are in fact ER negative.
However, these molecular apocrine breast cancers
express androgen receptor (AR) and AR highjacks
the pathways normally used by ER, associating
with DNA in the same locations that ER normally
occupies. As such, AR can mimic ER and can switch
on the genes normally regulated by ER. AR utilises
the pioneer factor, FoxA1, which directs AR to the
same regions in the genome that it would normally
direct ER to. These findings suggest that molecular
apocrine breast cancer patients, which make up
5% of all breast cancers, should be treated with
anti‑androgens, rather than anti-estrogens, since it
is AR that should be blocked, not ER.
Holland DG, Burleigh A, Git A, Goldgraben MA,
Perez-Mancera PA, Chin SF, Hurtado A, Bruna A,
Ali HR, Greenwood W, Dunning MJ, Samarajiwa S,
Menon S, Rueda OM, Lynch AG, McKinney S,
Ellis IO, Eaves CJ, Carroll JS, Curtis C, Aparicio S,
Caldas C. ZNF703 is a common Luminal B breast
cancer oncogene that differentially regulates
luminal and basal progenitors in human mammary
epithelium. EMBO Mol Med 2011; 3: 167-80
This paper describes the first new breast cancer
oncogene discovered in the last five years, called
ZNF703. It is predominant in luminal B breast
cancers – a more aggressive oestrogen positive form
of the disease – and it has been shown to drive
luminal differentiation. The authors used microarray
technology to narrow down the location of the
gene to a region on chromosome 8. It is thought
that up to a third of more aggressive oestrogen
positive breast cancers could have multiple copies
of the ZNF703 gene. If this finding is confirmed
then this could pave the way for the development
of treatments targeting ZNF703. Testing patients’
tumours for this gene would help identify those with
more aggressive tumours, so that their treatment
can be tailored accordingly.
Bohndiek SE, Kettunen MI, Hu DE, Kennedy BW,
Boren J, Gallagher FA, Brindle KM. Hyperpolarized
[1-13C]-ascorbic and dehydroascorbic acid: vitamin
C as a probe for imaging redox status in vivo. J Am
Chem Soc 2011; 133: 11795-801
Vitamin C is the body’s natural redox buffer, which
mops up reactive oxygen species. We have used
hyperpolarised 13C-labelled vitamin C (ascorbic
acid) and its oxidised product, dehydroascorbic
acid, to image tumour redox status in vivo. We
demonstrated, for the first time, that tumours very
rapidly take up dehydroascorbic acid and convert
it back to ascorbic acid, which is consistent with an
emerging picture in which tumours maintain a very
HO
HO
HO
13
O 13 O [1- C]-Ascorbic Acid
OH
HO
HO
[1-13C]-Dehydroascorbic Acid O
[1-13C]-DHA
[1-13C]-AA
O 13 O
O
Reduction of oxidised vitamin C (DHA) to reduced vitamin C (AA) in a tumour
180
175
Chemical shift (ppm)
170
Research Highlights | 9
10 | Cambridge Research Institute Scientific Report 2011
Research Groups
The CRI’s laboratories undertake research in four main areas:
1.Basic research into the cellular and molecular biology of
cancer.
2.Research in molecular imaging, genomics, bioinformatics and
biomolecular modelling.
3.Research focussed on specific cancer sites, which form a
bridge between laboratory and clinic.
4.Clinical investigations including experimental medicine
based clinical studies, conducted jointly with the University
of Cambridge and National Health Service (NHS) clinical
departments.
Tumour cells showing nuclei (blue - dapi) and lectin (green).
Lectin binds to sialic acids and β1,4 N-acetylglucosamine
(GlcNAc) oligomers at the outside of the cell.
Image by Esther Arwert (Watt laboratory)
Research Groups | 11
Chemical Biology of Nucleic Acids
www.cambridgecancer.org.uk/shankarbalasubramanian
Recent advances in the understanding of nucleic acid function
have shown that non-coding sequences have key roles in
regulating many cellular processes, from transcription and
translation to cell division and genome stability.
Group Leader
Shankar
Balasubramanian
Associate Scientist
David Tannahill
Postdoctoral Scientists
Dario Beraldi*
Enid Lam
Keith McLuckie
Graduate Students
Giulia Biffi
Debbie Sanders
Visiting Workers
Sina Berndl*
Sanne Hindriksen*
Ramon Kranaster
Chris Lowe
Mehran Nikan
Eun-Ang Raiber
Beth Thomas*
*
joined in 2011
†
left in 2011
While nucleic acids generally adopt a
well‑known double helical structure through
guanine-cytosine and adenine-thymine base
pairing, some sequences can take on alternative
structures. In guanine (G)-rich regions, G bases
can adopt stable intra-molecular arrangements
mediated by Hoogsteen hydrogen bonding
to form several stacked G-tetrads. Within
the human genome, we have shown that
potential G-quadruplex forming sequences,
with the consensus G3-6N1-7G3-6N1-7G3-6N1-7G3-6 ,
are common (Figure 1). These sequences
show particular concentrations near or in the
promoters and first introns of many genes,
including oncogenes such as MYC, KIT and RAS.
The accumulated evidence for G-quadruplex
structure and function is based largely on data
from biophysical, structural and in vitro studies.
We are therefore investigating the existence of
G-quadruplex nucleic acids in living systems, and
are seeking robust evidence of their biological
function and their validity as drug targets.
DNA G-quadruplexes
DNA G-quadruplexes are implicated in a
range of biological processes from the control
of cell division to the regulation of gene
transcription. We are investigating several
12 | Cambridge Research Institute Scientific Report 2011
aspects of DNA G-quadruplex biology. First,
we wish to prove the existence and survey
the extent of G-quadruplex formation in
living cells, and how this might be regulated in
cancer phenotypes. To do this we are using a
variety of probes to stabilise G-quadruplexes
in cells. We have therefore synthesised a
number of small molecules with high affinity
for G-quadruplex over duplex DNA. We
are also using protein tools as probes,
and are exploiting natural G-quadruplexbinding proteins, such as helicases, that
resolve G-quadruplex structures, as well as
engineered recombinant proteins, such as
specific antibodies and zinc-finger proteins
that specifically bind G-quadruplexes. By
isolating genomic DNA bound to these probes,
we can use chromatin immunoprecipitation
together with next‑generation sequencing
(ChIP‑seq) technologies to determine the sites
of G-quadruplex structure formation across the
genome. In addition, we are using our chemical
biology probes in high-resolution imaging and
chemical mapping approaches to definitively
demonstrate the formation of G-quadruplex
structures in the genome.
We are also exploring how G-quadruplexes
in promoters and gene bodies influence
gene transcription and DNA replication in
cancer cells. Many groups including ours have
intensely studied the biophysical and structural
H
R
N
N
N
N
H
H
N
H
O
N
O
N
N
R
cation
N
R
N
H
N
N
H
H
H
Stability: K+ >NH4+ > Na+ > Li+
Sr2+ >> Ba2+ > Ca2+ > Mg2+
N
H
N
N
N
X1-3
N
O
O
H
H
N
N
N
G
H
G
G
G
G
G
G
G
X 1-3
Figure 1
G-quadruplex formation mediated
by Hoogsteen hydrogen bonding
(left). Stacked G-tetrads in an
intramolecular G-quadruplex (top
right) and the putative consensus
sequence for G-quadruplex
formation (bottom right) (see
Huppert and Balasubramanian,
Nucleic Acids Res 2005; 33: 2908).
Rather than having typical Watson-Crick
base pairs that form a double helix, many
non‑coding sequences in DNA and RNA display
non‑standard structural features. For example,
guanine-rich sequences can adopt stable
four‑stranded structures called G-quadruplexes.
We hypothesise that the formation of such
structures in vivo is critical to biological function
and medicine. We aim to elucidate the role of
such structures in cancer and in normal cells. By
the application of small chemical molecules that
selectively target such non-canonical structural
elements, we further aim to develop novel
approaches that could be used in the treatment
of cancer.
G
G
G
G
X1-3
R
Consensus Putative Quadruplex Sequence:
G3-6N1-7G3-6N1-7G3-6N1-7G3-6
Figure 2
DNA damage and uncapping
of telomeres is induced by a
G-quadruplex-binding small
molecule. In treated cells (b), POT1
(green spots) is lost from telomeres
compared to untreated cells (a). The
compound induces DNA damage
as measured by gamma-H2AX foci
(red spots in c). This suggests that a
DNA damage response is stimulated
by loss of POT1 from telomeres.
Double staining, under conditions of
partial POT1 loss shows that DNA
damage occurs at telomeres (see
Rodriguez et al., J Am Chem Soc
2008; 130: 15758).
characteristics of predicted G-quadruplexes
found in human oncogenes and we have used
this information to design chemical biology
studies on cancer cells. We have now shown
that one of our G-quadruplex-binding small
molecules, called pyridostatin, induces growth
arrest in human cancer cells through the
induction of a replication- and transcriptiondependent DNA damage response. Genomewide approaches showed that the sites of
damage induced by pyridostatin were located
in gene bodies, including oncogenes such as
SRC, and we found that this also resulted
in suppression of SRC expression and a
concomitant inhibition of SRC-dependent
cellular motility. This work provides a novel
framework for defining functional drug-DNA
interactions for cancer therapies.
During cell division, it is vital the ends of
chromosomes do not become shortened or
are recognised by DNA damage response
pathways, otherwise genome instability will be
induced. Telomeres protect chromosomes
from damage by virtue of their DNA sequence.
This sequence, comprised of tandem TTAGGG
repeats, is required to recruit a protective
RNA G-quadruplex
GGGAGGGGCGGGUCUGGG
UTRQ
NRAS 5′ UTR (1–254)
Luciferase
5′ cap
DelQ
NRAS 5′ UTR (30–254)
Luciferase
5′ cap
AAAAGGGGCGGGUCUGGG
MutQ
NRAS 5′ UTR (1–254)
Luciferase
5′ cap
450
400
Relative luciferase activity (%)
Figure 3
G-quadruplexes in the 5′UTR of
NRAS mRNA modulate translation.
Luciferase reporter constructs
containing a G-quadruplex
upstream of the translation start
site show significantly reduced
translation levels as compared to
mRNAs containing no or mutated
G-quadruplexes (see Kumari et al.,
Nat Chem Biol 2007; 3: 218).
protein complex, known as shelterin, to
telomeres. The TTAGGG sequence is capable
of forming stable DNA G-quadruplex structures
in vitro, and others have observed the cell
cycle regulation of G-quadruplex formation at
telomeres in living invertebrate cells. Telomeres
are also actively transcribed into telomeric
(TERRA) RNA. While TERRA RNA can form
stable G-quadruplexes in vitro, it is not known if
this is true in vivo or whether this is needed for
normal telomere function. Also, of note is the
observation that 85% of primary tumours show
increased expression of the enzyme telomerase,
which is required to maintain telomeres. By
applying small molecule and protein probes,
together with genetic approaches, we aim
to prove the existence and understand the
regulation of DNA telomeric G-quadruplexes
and the role of TERRA RNA in human cells.
Indeed, we have shown that the application of
G-quadruplex-binding small molecules results
in the release the shelterin complex from the
telomeres to cause DNA damage (Figure 2).
RNA G-quadruplexes
There is much evidence that G-quadruplexes
are widely present in RNA and that they may
be associated with several key aspects of
RNA biology. For example, G-quadruplexes
in the 3′‑UTR of insulin-like growth factor
II mRNA play a role in post-transcriptional
endonucleolytic cleavage. Furthermore, we
have recently shown that a conserved RNA
G-quadruplex motif in the 5′-UTR of the human
NRAS proto-oncogene can modulate protein
translation (Figure 3). We have also shown that
small molecule ligands can target such RNA
G-quadruplexes and thus influence translation.
As RNA helicases with G-quadruplex resolving
activity have recently been identified, this
suggests that RNA quadruplexes exist normally
in vivo. Our bioinformatics analysis highlights
that large numbers of human RNA transcripts
contain a potential G-quadruplex forming
region. This raises important questions:
how widespread are G-quadruplexes in
RNA transcripts, and what is their functional
relevance? To address this we are therefore
using a combination of genome-wide ChIP-seq
and chemical biology approaches to identify and
map the existence of G-quadruplex structures
within the transcriptome and investigate their
role in cancer cells.
Publications listed on page 72
350
300
250
200
150
100
50
0
UTRQ
DelQ
MutQ
Research Groups | 13
Functional Genomics of Ovarian Cancer
www.cambridgecancer.org.uk/jamesbrenton
Group Leader
James Brenton
Bioinformatician
Lorna Morris† (with C Caldas)
Clinical PhD Student
Ioannis Gounaris*
Clinician Scientist
Christine Parkinson†
Graduate Students
Charlotte Ng
Elke Van Oudenhove*
Siru Virtanen (with J Stingl)
Feng Wang †
Postdoctoral Scientists
Susannah Cooke†
Scott Newman*†
Principal Scientific Officer
Jian Xian
Scientific Officer
Jill Temple
Summer Placement Student
Jessica Unger *†
Temporary Staff
Amin Ahmadnia*
Visiting Workers
Tom Beale†
Heather Biggs
Lily Chan*†
Steve Charnock-Jones*
Claire Dawson*
Merche Jimenez-Linan
James Shearman†
*
joined in 2011
†
left in 2011
Our laboratory focuses on discovering improved treatments
for epithelial ovarian cancer using laboratory and clinical
studies. Ovarian cancer has a high healthcare burden because
of low cure rates and frequent recurrent disease that causes
significant symptoms for patients. This is despite the fact that
ovarian cancer is initially sensitive to systemic treatments and
most patients are free of disease after completing initial surgery
and chemotherapy. The fundamental problem that we are
addressing is to understand how ovarian cancer cells escape
initial treatment and the molecular mechanisms by which
they acquire resistance to further therapy. Using genomic
and functional studies we are identifying new biomarkers and
treatment targets for testing in clinical trials.
Genomic studies of chemotherapy response in
vivo
To identify genetic alterations that are selected
for during the acquisition of drug resistance
we are carrying out prospective clinical studies
that collect cancer samples before and during
neoadjuvant treatment. Our initial studies
have focused on the drugs carboplatin and
paclitaxel as these are the most important
therapies in ovarian cancer. By using expression
analysis and bioinformatics methods that have
been developed to model the acquisition of
resistance, we have identified clinically relevant
biomarkers that overlap with independently
identified genes from RNA interference screens
(Swanton et al., Cancer Cell 2007; 11: 498).
Our studies depend upon having homogeneous
patient cohorts with similar clinical characteristics.
However, response to treatment in tumour
masses can be heterogeneous and mixed
response frequently occurs at different
anatomical sites. For example, primary ovarian
masses may respond better than peritoneal
metastases. This differential response may be a
result of variable blood supply and hypoxia that
limits delivery and efficacy of chemotherapy.
We have confirmed these observations using
functional magnetic resonance imaging for
perfusion (Sala et al., Eur Radiol 2010; 20: 491)
14 | Cambridge Research Institute Scientific Report 2011
and diffusion and are now using imaging data to
target the collection of tissues from responding
and non-responding areas. This will allow
us to calibrate genomic profiles much more
precisely and to better identify the molecular
determinants of resistance. High throughput
sequencing with Illumina technologies is being
used to quantitate expression and genomic
changes and to identify novel fusion transcripts
and mutations (Figure 1).
Differential sensitivity to paclitaxel as compared
to carboplatin may depend on cellular pathways
involved in maintaining chromosomal stability
(CIN). To ask whether this may be clinically
relevant we have tested surrogate expression
markers of CIN in samples from a prospective
neoadjuvant study and have shown that high
measures of CIN predict resistance to paclitaxel
and increased sensitivity to carboplatin
(Swanton et al., PNAS 2009; 106: 8671)
(Figure 2). Thus, measuring CIN pre-treatment
may optimise choice of treatment for patients.
The key oncogenic and tumour suppressor
genes for high-grade ovarian serous carcinoma
have not been identified as this type of tumour
has high rates of genomic instability, where many
of the described alterations may be passenger
mutations. Numerous studies have tested the
association between TP53 mutations in ovarian
Log2 Ratio
A
Allele frequency
B
C
Copy number
Position on chromosome
cancer and prognosis but these have been
consistently confounded by limitations in study
design, methodology and/or heterogeneity in the
sample cohort. To identify the true prevalence
of TP53 mutations in high-grade pelvic serous
carcinoma, we sequenced exons 2–11 and
intron-exon boundaries in tumour DNA from
145 patients with invasive serous carcinoma of
the ovary, fallopian tube and primary peritoneal
cancer. Surprisingly, pathogenic TP53 mutations
were identified in 97% (n = 119/123) of HGS
cases (Ahmed et al., J Pathol 2010; 221: 49). This
is the first comprehensive mapping of TP53
mutation rate in a homogeneous group of
high-grade pelvic carcinoma patients and shows
that mutant TP53 is a driver mutation in the
pathogenesis of HGS cancers.
Mechanisms of taxane resistance and the role
of extracellular matrix
Taxanes, such as paclitaxel, interfere with the
dynamic growth of microtubules by directly
binding to them, leading to mitotic arrest and
apoptosis. Paclitaxel is widely used to treat
ovarian and breast cancers but drug resistance
limits its clinical usefulness to only half of
patients who receive it.
Alterations in the ratio of tubulin isoforms or
mutations in tubulin can alter microtubule stability
and sensitivity to taxane drugs. By studying
Current projects are characterising how TGFBI
interacts with integrins and other cell surface
receptors and how this may be modulated
therapeutically. It is now clear that TGFBI
exerts its effects specifically through beta-3
integrins but is also co-regulated, and interacts
with, other ECM proteins implicated in drug
resistance. To identify the downstream
pathways from FAK and RHO that alter
microtubule stability, we have generated
knock-out somatic cell lines using homologous
recombination. These knock-out models
have provided a powerful system to identify
microtubule associated proteins responsible
for effects on paclitaxel resistance. As TGFBI
has complex roles in organising interactions
between cells and ECM, we have studied its
function in early development in Xenopus to
identify how it may affect cell migration. Both
loss and gain of function experiments have
shown that TGFBI is required for somite
development in Xenopus.
Publications listed on page 72
●
9
●
●
●
●
8
●
●
● ●
●
●
●
●
●
●
●
●
● ● ●
●
●
●
●
●
● ●
●
●● ●
●
●
●
●
● ●
7
●
●
●
●● ● ● ●
●
●
●
●
●
●
●
●
●●
6
●
●
6
7
8
9
10
Median CIN70 log intensity in Paclitaxel−sensitive tumours
10
●
●
●
●
●
9
●
●
●
●
●
●
●
8
●
●
●
●
● ●
●
●
7
Median CIN70 log intensity in Carboplatin−resistant tumours
10
●
Median CIN70 log intensity in Paclitaxel−resistant tumours
Figure 2
Expression of CIN70 genes
determines sensitivity to paclitaxel
and carboplatin. The figure
contrasts basal median gene
expression for each CIN70
gene in tumours with differing
responses to paclitaxel and
carboplatin. Paclitaxel-resistant
tumours exhibited a higher median
log‑intensity of the CIN70 signature
compared with paclitaxel-sensitive
tumours (P=0.043). CIN70 gene
expression differed significantly
between tumours subsequently
resistant to paclitaxel and tumours
resistant to carboplatin (P=0.044;
Student 2-sided t-test).
cell line models of taxane resistance along with
clinical samples we have recently shown that
loss of the ECM protein, transforming growth
factor beta induced (TGFBI), was sufficient to
induce paclitaxel resistance in cells and ovarian
cancer tissues (Ahmed et al., Cancer Cell 2007; 12:
514). We have also shown that TGFBI induces
microtubule stabilisation that is dependent
upon integrin-mediated FAK and RHO signalling
pathways. Extracellular matrix proteins have been
implicated in the acquisition of drug resistance in
ovarian cancer although the mechanism by which
this is achieved is unclear. Loss of TGFBI induces
resistance by altering microtubules which are the
direct pharmacodynamic target of paclitaxel. This
work shows that the effects of ECM proteins on
drug resistance may be very specific to particular
cytotoxic treatments. As 30% of ovarian cancers
do not express TGFBI, it may be an important
biomarker for paclitaxel response.
●
●
●
●
●
●●
●
6
Figure 1
High-resolution array CGH analysis
of ovarian tumours. The Illumina
1M SNP array gives (A) Copy
number data and (B) SNP allele
calls. Applying the QuantiSNP
segmentation algorithm provides (C)
copy number calls. Blue, four copies;
Green, three copies; Yellow,
one copy. We are using this along
with high-throughput sequencing
data to characterise ovarian tumour
heterogeneity and evolution towards
chemotherapy resistant disease.
●
●
●
●●
●
●●
●
●
●
●
●
●
● ● ● ●●
● ● ●●
●
●
●
●
● ●
●
●
●
●
●
●
6
7
8
9
10
Median CIN70 log intensity in Carboplatin−sensitive tumours
Research Groups | 15
Molecular Imaging of Cancer
www.cambridgecancer.org.uk/kevinbrindle
The primary aim of our laboratory is to develop imaging methods
that can be used in the clinic to detect early tumour responses
to treatment. These could be used in early stage clinical trials of
new drugs to get an indication of efficacy and subsequently, in the
clinic, to guide therapy in individual patients.
Group Leader
Kevin Brindle
Clinical Fellows
Sarah Fawcett
Zoltan Szucs
Clinician Scientist
Ferdia Gallagher
Graduate Students
Thomas Booth
Piotr Dzien
Brett Kennedy
Joe Chin-Han Kuo
Henning Stöckmann
Postdoctoral Scientists
Sarah Bohndiek
Joan Boren
Kathrin Heinzmann*
Tim Larkin
David Lewis
Tiago Rodrigues
Esther Rodriguez
Principal Scientific Officers
De-En Hu
André Neves
Scientific Officers
Paula D’Santos
Rebecca Harmston
Liz Mannion
Helen Sladen
Senior Staff Scientist
Dmitry Soloviev
Staff Scientists
Mikko Kettunen
Scott Lyons
Visiting Workers
Elizabeth Bird-Lieberman†
Holly Canuto†
Michaela de Clare*†
William Hughes*
Irene Marco Ruis*
Brad O’Dell
Giuseppe Pileio*
*
joined in 2011
†
left in 2011
Patients with similar tumour types can show
markedly different responses to the same
therapy. The development of new treatments
would benefit, therefore, from the introduction
of imaging methods that allow an early
assessment of treatment response in individual
patients, allowing rapid selection of the most
effective treatment for a specific patient
(Brindle, Nat Rev Cancer 2008; 8: 1).
A targeted imaging agent for detecting cell
death
We have continued development of an agent
based on the 14 kDa C2A domain of the
protein synaptotagmin, which binds to the
phosphatidylserine (PS) exposed by dying cells.
Last year we reported a site-directed mutant of
the protein (C2Am) in which the introduction
of a cysteine residue (S78C) distant from
the active site allows site-specific attachment
of imaging labels using sulphydryl‑selective
reagents. We also showed that this agent
had better specificity for detecting dying cells
in vitro than Annexin V since it showed less
binding to viable cells. Annexin V has already
been trialled in the clinic for detecting therapy
response in tumours, however there were
problems with non-specific binding. This
year, using a fluorescently labelled derivative,
we have shown that C2Am shows excellent
selectivity for detecting tumour cell death in
vivo post‑treatment (Figure 1). We believe,
therefore, that radiolabelled derivatives of
C2Am may have some significant advantages
for detecting therapy response in the clinic,
particularly in tumours in the abdomen.
Imaging metabolism with hyperpolarised
13
C-labelled cell substrates
MRI gives excellent images of soft tissues,
such as tumours. The technique works by
mapping, in 3D, the distribution and MR
properties of tissue water protons, which are
16 | Cambridge Research Institute Scientific Report 2011
very abundant (60 – 70 M in tissues). However,
we can also use MR to detect metabolites
in vivo. The problem is that these molecules
are present at 10,000× lower concentration
than the protons in tissue water, which makes
them hard to detect and almost impossible
to image, except at very low resolution. We
have been collaborating with GE Healthcare
in the development of a technique, termed
‘hyperpolarisation’, that increases sensitivity in
the MRI experiment by more than 10,000×.
With this technique we inject a hyperpolarised
13
C-labelled molecule and now have sufficient
sensitivity to image its distribution in the
body and the distribution of the metabolites
produced from it. In the past year we have
developed an exciting new probe for imaging
tumour redox status. Vitamin C is the body’s
natural redox buffer, which mops up reactive
oxygen species. We have used hyperpolarised
13
C-labelled vitamin C (ascorbic acid) and
its oxidised product, dehydroascorbic acid,
to image tumour redox status in vivo. We
demonstrated, for the first time, that tumours
very rapidly take up dehydroascorbic acid
and convert it back to ascorbic acid, which
is consistent with an emerging picture in
which tumours maintain a very reduced
microenvironment (Bohndiek et al., J Am Chem
Soc 2011; 133: 11795). We believe that it will be
a sensitive probe for detecting early responses
of tumours to treatment, since we expect some
therapeutic agents to have a dramatic and early
effect on tumour redox status.
Imaging tumour cell glycosylation
Aberrant glycosylation is a hallmark of cancer. In
collaboration with Rebecca Fitzgerald’s group
in the Hutchison/MRC Research Centre we
have developed a fluorescently-labelled lectin
for endoscopic detection of early dysplasia in
the oesophagus (Bird-Lieberman et al., Nat Med
2012; Epub Jan 15). We are also developing
Figure 1
Imaging cell death in a genetically
engineered mouse model of
lymphoma using fluorescentlylabelled C2Am. Representative
fluorescence images (the
fluorophore emits at 750 nm) of
untreated (A) and drug-treated
(B) animals following injection of
labelled C2Am. The agent detects
cell death in tumours around the
neck and in lymphomatous disease
within the thorax in the treated
animal. There was also some cell
death in the neck tumour of the
untreated animal. Cell death was
confirmed by histology. The signal
at the bottom of both images is
from urine in the bladder and at the
urethra.
A
Visiting Workers ctd
Maxim Rossman*
Eva Serrao*
Shaun Stairs*
Sui Seng Tee
Kerstin Timm*
Yelena Wainman*
Yimao Zhang*
a novel molecular imaging platform for the
dynamic non-invasive assessment of tumour
glycosylation state, in which sugar analogues
are incorporated metabolically by tumour cells
in vivo and detected subsequently by a highly
selective chemical reaction (“click chemistry”)
with a reporter probe that has been labelled
with an imaging agent. We have demonstrated,
for the first time, that this technique can be
used to image tumour glycans in vivo, using
both fluorescence and radionuclide (SPECT)
imaging (Neves et al., FASEB J 2011; 25: 2528).
This year we have produced a range of novel
small molecule click reagents that should
improve the image contrast obtainable with this
technique (Stöckmann et al., Chem Sci 2011; 2:
932; Stöckmann et al., Chem Comm 2011; 47:
7203; Stöckmann et al., Org Biomol Chem 2011;
9: 7303). The methodology could potentially
be used for tumour detection, imaging tumour
cell proliferation and detecting response to
therapy. The technique also has the potential
for subsequent translation into a clinical setting,
using nuclear imaging techniques.
B
Future directions
We will continue development of the C2Am
agent for detecting cell death, particularly
with radionuclide-labelled derivatives. We
will conduct further studies with fluorescently
labelled lectins in the oesophagus and also
the colon. We will use our newly developed
click reagents to obtain better glycan image
contrast in vivo. We will explore the potential of
hyperpolarised 13C-labelled vitamin C to give us
new information on tumour redox status in vivo
and anticipate taking the first steps in translating
hyperpolarised 13C technology to the clinic
with the installation of a clinical polariser in the
Department of Radiology.
Publications listed on page 72
Research Groups | 17
Functional Genomics of Breast Cancer
www.cambridgecancer.org.uk/carloscaldas
The characterization of the molecular heterogeneity of
breast cancer is leading to increasingly personalized cancer
management and better understanding of the biology of the
disease. Our laboratory has continued to make advances on
both of these fronts.
Group Leader
Carlos Caldas
Associate Scientist
Suet-Feung Chin
Bioinformatician
Bin Liu
Clinical Fellows
Jean Abraham
Raza Ali
Sarah Jane Dawson
Clinician Scientist
John Le Quesne
Graduate Students
Mae Goldgraben
Angelika Modelska
Jose Sandoval
Ana Tufegdžić Vidaković
Postdoctoral Scientists
Alejandra Bruna
Anna Git
Stefan Gräf
Oscar Rueda
Research Assistant
Josephine Beaton*
Scientific Officers
Helen Bardwell
Wendy Greenwood
Sarah McGuire
Rebecca Sargeant
Joanna Warren†
Visiting Workers
Katy Bird
Heidi Dvinge
Mahesh Iddawela†
Lorna Morris
Claire Pike
Stephen Sammut
Ina Schulte
Katy Teo*†
Pauline Traynard*†
Hans Kristian Moen Vollan
Jamie Weaver
*
joined in 2011
†
left in 2011
Translational breast cancer genomics:
applications of molecular profiling in prognosis,
prediction and novel therapeutics
We have completed the analysis of the genomic
and transcriptomic landscapes of 2000 breast
cancers with linked clinical follow-up using
high-resolution SNP arrays (Affymetrix SNP6.0)
and gene expression arrays (Illumina). This
allowed us to do an eQTL analysis revealing that
inherited variants (CNVs, SNPs) and acquired
somatic copy number aberrations (CNAs)
were associated with ~40% of genes, although
the landscape was dominated by CNAs. By
delineating expression outlier genes driven in
cis by CNAs, we identified several novel driver
cancer genes, including deletions in PPP2R2A,
MTAP, NCOR1 and MAP2K4. Integrative
clustering analysis of DNA/RNA profiles revealed
novel subgroups with distinct clinical outcomes.
These include a high-risk, ER-positive 11q13/14
cis-acting subgroup and a favourable prognosis
subgroup devoid of CNAs. Trans-acting
aberration ‘hotspots’ were found to modulate
subgroup specific gene networks such as a TCR
deletion-mediated adaptive immune response in
the ‘CNA-devoid’ subgroup and a Basal-specific
chromosome 5 deletion-driven mitotic network.
Using the same cohort we have also shown
that a complex arm aberration index (CAAI),
derived from the SNP array segmented data,
and adapted from what we previously published
in collaboration with the Borresen-Dale
group, validates as an independent prognostic
marker in both ER+ and ER− disease. Finally,
we have obtained miRNA expression profiles
from ~1,300 of the same cases allowing for an
integrated analysis that has revealed patterns
of mRNA/miRNA interactions that modulated
cancer signalling pathways differentially (Figure 1).
We have completed, in collaboration with
Sam Aparicio in Vancouver, the sequencing
of around 100 exomes and transcriptomes
18 | Cambridge Research Institute Scientific Report 2011
from triple-negative (ER−/PR−/Her2−) breast
cancers, revealing complex clonal heterogeneity
in these tumours at clinical presentation. We
also contributed to the International Cancer
Genome Consortium (ICGC) effort to
characterise 100 breast cancer exomes to single
nucleotide resolution, which has revealed several
novel cancer genes. We sequenced DNA from
selected breast cancers for which we have
collected serial plasma samples in the metastatic
phase of the disease to compare the clinical
utility of circulating tumour DNA, circulating
tumour cells and imaging for tumour monitoring.
We have expanded our efforts in both systems
pathology and digital pathology, aiming at
integrating molecular profiling with tissue
architecture. We developed, in collaboration
with Florian Markowetz, a computational
approach to deconvolute cellular heterogeneity
and detect subtle genomic aberrations, to
boost the comparability of copy-number
profiles between samples. The method utilises
standard H&E-stained sections and employs an
algorithm based on tumour cellularity. We also
demonstrated that a lymphocytic infiltration
predictor for survival which integrates imagebased and molecular features significantly
outperforms classifiers based on single data
types.
Finally we continue to use the tissue
microarray (TMA) resource we have built,
which to date includes ~9,000 samples from
a population‑based cohort and from four
randomised clinical trials. Using this unique
resource of clinically-annotated tumours we
can perform studies that rigorously conform to
the REMARK (REporting recommendations for
tumour MARKer prognostic studies) guidelines.
For example, we have shown that Ki67 and
BCL2 can be effectively combined to produce
an index which is an independent predictor of
HES1
CD3E
CD3E
HES1
PLS<0
CTNNA1
DYRK2
CTNNA1
IFT52
DYRK2
KIAA1278
IFT52
GLI3
IFT172
PLS>0
KIAA1278
IFT172
GLI3
ULK3
HIPK2
ULK3
POR
TULP3
HIPK2
TULP3
POR
GAS1
STIL
STIL
SFRP1
BOC
SFRP1
PRRX1
PRRX1
BOC
DISP1
GPC3
GLI2
RUNX2
BMP4
DISP1
GPC3
GLI2
RUNX2
GAS1
GO:0007224 smoothened signaling pathway
ER−
BMP4
PRRX2
ER+
PRRX2
Figure 1
The interaction between the
miRNome and signalling pathways
is different in ER-positive and
ER-negative breast cancer. Green
and pink lines depict correlation
or anti-correlation (measured
by partial least squares; PLS),
respectively, between individual
miRNAs (grey vertical bars, middle)
and mRNAs classified under the
Gene Ontology term “smoothened
signalling pathway” (yellow or blue
rectangles in ER negative or positive
disease, respectively). The mRNAs
are listed in the same order in the
ER+ and ER− rows and the widths
of the boxes are proportionate to
the number of miRNA connections.
Only mRNAs with at least one
significant miRNA connection are
shown.
nc=1, nr=1
Breast Cancer Specific Survival (BCSS) in ER+
cases, thus enhancing their potential prognostic
utility. We have also conducted the largest
study to date on cancer stem cell markers and
their prognostic value.
Collaborators: Sam Aparicio (University of British
Columbia), Simon Tavaré and Florian Markowetz
(CRI), Paul Pharoah (Strangeways Research
Laboratory), Helena Earl (Department of Oncology
and Addenbrooke’s Hospital), the Molecular
Taxonomy of Breast Cancer International Consortium
(METABRIC), Anne-Lise Borresen-Dale (Oslo)
Functional breast cancer genomics:
characterising tumour initiating/cancer stem
cells in breast cancer subtypes
We have now demonstrated that ZNF703
is a common Luminal B breast cancer
oncogene that differentially regulates luminal
and basal progenitors in human mammary
epithelium. The interactions of ZNF703 with
ER and HDACs, in a chromatin-associated
complex, suggest that in the mammary
epithelium ZNF703 might differentially regulate
stem/progenitor cells at the transcriptional level.
This complex might be potentially specifically
targeted, for example by HDAC‑inhibitors,
exclusively in the luminal progenitor
compartment.
The differential effects of TGFβ on breast
cancer stem cells have not been previously
characterised and we have now shown, using
rigorous in vitro (mammosphere and CFCs) and
in vivo (limited dilution xenografting) assays,
that it promotes self-renewal exclusively in
claudin‑low breast cancers, and this effect
is distinct from its EMT-inducing properties.
Furthermore we showed mechanistically
that this effect is mediated by cross-talk with
Rho-Rac-SRF, opening new possibilities for
therapeutic strategies in these aggressive
tumours.
Using similar assays for breast cancer stem cells
we have shown that IER5, a novel Notch target
gene which we have identified, contributes
to the effects of Notch on breast cancer
stem/progenitor cells.
Collaborators: John Stingl and Jason Carroll (CRI)
Publications listed on page 73
Research Groups | 19
Nuclear Receptor Transcription
www.cambridgecancer.org.uk/jasoncarroll
We are interested in defining the genomic and molecular
features of oestrogen receptor (ER)-mediated transcription
in breast cancer cells. We are specifically interested in
understanding how these events and the machinery involved
cause breast cancer cells to grow.
Group Leader
Jason Carroll
Clinical Fellow
Kamarul Zaki*
Graduate Students
Kamila Jozwik *
Hisham Mohammed
Jessica Robinson (with D Neal)
Caryn Ross-Innes† (with D Odom)
Dominic Schmidt † (with D Odom)
Senior Scientific Officer
Kelly Holmes
Postdoctoral Scientists
Antoni Hurtado-Rodriguez †
Aisling Redmond†
Vasiliki Theodorou
Wilbert Zwart †
Scientific Officer
Rosalind Launchbury
Visiting Worker
Jo Leeper *
Oestrogen receptor is the defining feature
of luminal breast cancers, where it functions
as a transcription factor to induce cell cycle
progression. ER is also the target of most
endocrine therapies, including tamoxifen and
aromatase inhibitors, which are effective
treatments. However, some women can
develop resistance to these drugs and in many
cases, ER simply gets switched back on again,
despite the presence of the drug. Therefore,
understanding how ER functions is an important
issue and one that has not been completely
resolved. ER transcriptional activity requires
a number of co-factors and co-operating
transcription factors that possess enzymatic
activity to alter chromatin structure, the
outcome of which determines transcriptional
activity. It is currently known that a number of
ER co-factors can either assist in transcription
(including SRC-1 and AIB-1) or are involved in
gene repression by tamoxifen (including N-CoR
and SMRT).
Recently, using chromatin immunoprecipitation
(ChIP) combined with high-throughput
sequencing (ChIP-seq), we mapped all ER
binding sites in a breast cancer cell model
after oestrogen treatment. This unbiased
identification of the genomic contact points of
ER revealed a number of surprising features
about ER biology. These included the
observation that ER rarely regulates genes from
promoter regions, but instead utilises distal
enhancers. We also identified the role of a
‘pioneer factor’ called FoxA1, which is critical
for ER to function. Our lab is interested in
extending these findings to fully define the
cis- and trans-elements that contribute to ER
activity in breast cancer cells, with particular
emphasis on the pioneer factors that stabilise
ER-DNA interactions.
*
joined in 2011
†
left in 2011
20 | Cambridge Research Institute Scientific Report 2011
Characterisation of the role of pioneer factors
in ER biology
We are interested in identifying and
characterising the role of the pioneer factor
FoxA1 in regulating ER activity. We have
found that FoxA1 is required for all ER-DNA
interactions and for ER to promote cell growth.
In the absence of FoxA1, ER function is blocked
and cells do not proliferate. We have also
been interested in finding additional ER pioneer
factors. We recently discovered a role for the
Groucho protein, TLE1, as a pioneer factor
for ER. Unlike FoxA1, TLE1 appears to be a
pioneer factor only at a subset of ER binding
events. When TLE1 is specifically silenced,
ER binding to the chromatin is depleted at
approximately half of all ER binding events. This
results in changes in gene expression profiles
and cell cycle arrest. As such TLE1 is essential
for some ER binding events and for effective
functioning of the ER complex. Interestingly, the
ER binding events that require TLE1 tend not to
be the regions that are co-occupied by FoxA1.
As such, there is likely to be interplay between
the pioneer factors, with FoxA1 regulating
ER binding, but simultaneously and indirectly
influencing other pioneer factors, such as TLE1.
This gives insight into the factors that allow ER
to make contact with the DNA and provides
opportunities for targeting these pioneer factors
in breast cancers that have acquired drug
resistance.
Genomic analysis of ER function in primary
breast cancer
All ER genomic studies to date have been
limited to breast cancer cell line models, yet
they have revealed extraordinary features
about ER biology. We have now been able to
extend genomic transcription factor mapping
experiments into frozen primary breast cancer
Figure 1
In breast cancer cells, the male
hormone receptor androgen
receptor (AR) can mimic estrogen
receptor (ER).
(A) Estrogen receptor negative
breast cancer cells that express
androgen receptor (termed
molecular apocrine breast cancers)
require AR for growth.
(B) In breast cancer cells, AR
associated with the genome in
locations that are normally ER
interaction sites. As such, AR can
behave differently in breast cancer
cells to mimic the behaviour of ER.
This results in the unusual situation
where a breast cancer is driven by
AR, instead of ER.
A
B
Cell growth
Protein-DNA interactions (ChIP-seq)
100 kb
128800000
chr8:
44 -
siControl
siAR
128850000
128900000
128950000
AR
MDA-MB-453
2_
2057 -
ER
MCF7
2_
40 -
AR
LNCaP
2_
RefSeq Genes
samples, by performing ER ChIP-sequencing
in luminal breast cancer material. The data
confirm that ER ChIP-seq can be performed
in primary breast cancer samples and that the
ER binding events accurately represent the
binding sites in the cell lines. However, there
are significant numbers of ER binding events
that are acquired in tumours with a poor
clinical outcome and in metastatic material that
originated from an ER positive breast cancer.
The novel ER binding events correlate with
genes that have predictive value in independent
breast cancer cohorts. We can model these
events using drug sensitive or resistant cell line
models, where ER binding events are dynamic
and can be reprogrammed with growth factor
stimulation. The reprogrammed ER binding
events are mediated by changes in the pioneer
factor FoxA1. We are currently exploring
what enables changes in FoxA1, since these
mediate the changes in ER binding events and
subsequently influence the transcriptome.
BC042052
MYC
MYC
PVT1
PVT1
PVT1
(termed molecular apocrine) express gene
patterns that are normally expressed in tumours
driven by ER. We have shown that in molecular
apocrine breast cancer cells, AR can substitute
for ER, go to the same regions in the genome
that ER normally goes to, and can switch on the
same genes that ER normally regulates (Figure
1). As such, when ER is absent, AR can highjack
the mechanisms used by ER and can mimic
ER. We have shown that AR uses the pioneer
factor FoxA1, which directs AR to the regions
in the genome that ER is normally directed to.
These findings suggest that molecular apocrine
breast cancers, which make up ~5% of all breast
cancers, probably would not benefit from
ER antagonists, but may in fact benefit from
prostate cancer drugs that specifically target AR.
Publications listed on page 74
Understanding the role of androgen receptor
in breast cancer
Most ER positive breast cancers also express
androgen receptor (AR), the male hormone
receptor. The parallels between ER biology
in breast cancer and AR biology in prostate
cancer are very high. Mechanisms are common
and interacting proteins are similar. What is
unknown is how AR behaves in breast cancers.
Recently, an unusual breast cancer subtype has
been described, which are AR positive, but ER
negative. Unexpectedly, these specific tumours
Research Groups | 21
Tumour Immunology and the FAP+ Stromal Cell
www.cambridgecancer.org.uk/dougfearon
Principal Investigator
Douglas Fearon*
Graduate Students
James Jones*
Lukasz Magiera*
Laura Mears*
Edward Roberts*
Andy Watts*
Richard Wells*
Postdoctoral Scientists
James Arnold*
Alice Denton*
Matthew Kraman*
Visiting Workers
Dylan MacLochlainn*
James Thaventhiran*
Liora Vilmousky *
*
joined in 2011
†
left in 2011
Even though spontaneous or vaccine-induced systemic immune
responses to cancers occur, the stromal microenvironment of
tumours protects cancer cells from immune attack. We have
recently found that a stromal cell identified by its expression
of fibroblast activation protein-α (FAP) mediates immune
suppression in murine tumours. We seek ways to block its
immune suppressive functions to improve clinical tumour
immunotherapy.
The FAP+ stromal cell in tumours
The proposal of the immune surveillance of
cancer, as put forward by Macfarlane Burnet and
Lewis Thomas, hypothesizes that cancers may
sufficiently differ from normal cells so that they
would be recognized by the immune system
and eliminated. Today we know that cancers,
either because they are virally induced and
express foreign viral antigens, or are genetically
unstable and express mutated self antigens, do
induce systemic immune responses, but we also
recognize that cancers usually escape immune
control.
on the possibility that the cancer cell itself was
responsible for tumoural immune suppression,
gaps in our understanding of how the immune
system worked, and the complexity of the
tumour stroma.
Two general mechanisms have been proposed
for the ability of cancers to circumvent an
immune response: establishing an immune
suppressive microenvironment within the
tumour, and the generation and immune
selection of cancer cell variants that are not
immunologically recognized. Evidence for
both exists, but we decided to concentrate on
immune suppression because it would dominate
over immune selection, and it offered the
possibility of therapeutic approaches.
The tumour stroma is comprised of three
general cell types, those involved with forming
the tumour vasculature, cells of the innate and
adaptive immune systems, and mesenchymal
cells, or fibroblasts. Most work has been
directed to understanding the roles of the cells
of the immune systems, with the reasonable
rationale that the processes intrinsic to this
system, which control auto-immunity, also
would be involved in immune suppression in
the tumour microenvironment. This approach
has been productive and has led to the
development of a clinically approved treatment
for metastatic melanoma, ipilimumab, an
antibody that blocks the function of CTLA-4, a
lymphocyte receptor. This treatment, however,
causes systemic autoimmunity because it does
not selectively target immune suppression in the
tumour microenvironment.
Early evidence for an immune suppressive
microenvironment within tumours was the
observation that an established tumour,
containing not only cancer cells but also
non‑cancer “stromal cells”, was resistant to killing
by tumour antigen-specific T cells. However,
cancer cells alone, without an accompanying
stroma, were eliminated. This finding was made
more than a quarter of a century ago, but the
realization that attention must be directed to
the tumour stroma has been slow to develop,
perhaps for three reasons: a continued emphasis
To determine the cellular basis for
immune suppression within the tumour
microenvironment, we focused on stromal
cells of mesenchymal origin, which have
usually been referred to as myofibroblasts
or carcinoma‑associated fibroblasts (CAFs).
These cells have been examined for their
ability to promote tumour growth, but not by
an immunological mechanism. Over the last
20 years, however, an interesting correlation
was found between the occurrence of chronic
inflammatory lesions of various types, such as
22 | Cambridge Research Institute Scientific Report 2011
The FAP+ stromal cell and normal tissues
The possibility that FAP+ stromal cells might have
functions in normal tissues was raised by their
presence in the somites of developing mouse
embryos, and in the uterus and placenta. To
examine this possibility, we developed a mouse
in which luciferase was expressed in FAP+ cells,
which has revealed that FAP+ cells are present in
almost all tissues of the adult mouse. Thus, FAP
expression may denote a mesenchymal lineage
with both shared and tissue-specific homeostatic
functions, as well as its immune suppressive
function in tumours, which may be an elicited
activity that is potentially available to “injured”
tissues throughout the body.
Figure 1
A mouse pancreatic ductal
adenocarcinoma showing the FAP+
stromal cells (red) surrounding
the ductal cancer cells (green)
with Trp53R172H+ nuclei (white).
Photograph courtesy of James Jones.
atherosclerosis, rheumatoid arthritis, cirrhosis,
and dermal scars, and the presence of a
mesenchymal cell that was first observed in
most human adenocarcinomas by its expression
of a membrane protein, FAP. The recognition
that tumours contain the same inflammatory
cells that characterize these chronic lesions, the
likelihood that these lesions represent attempts
at tissue repair, and the possibility that immune
suppression is a normal component of tissue
repair led to the consideration that the FAP+
stromal cell might have a role in tumoural
immune suppression.
We tested this possibility by developing a
mouse line in which the primate diphtheria
toxin receptor is expressed in FAP+ stromal
cells to enable their conditional depletion by
the administration of diphtheria toxin. The
experiment was informative in that depleting
FAP+ cells from the stroma of established
tumours caused immune control of tumour
growth. This finding was initially made with
immunogenic, ectopic tumours caused by
injecting cultured cancer cells, but has been
extended now to the Tuveson laboratory
model of spontaneous pancreatic ductal
adenocarcinoma in which cancer cells express
mutant Trp53R172H and KrasG12D alleles (Figure
1). The FAP+ stromal cell is a non‑redundant
element of tumoural immune suppression,
and the presence of these cells in human
adenocarcinomas suggests that these findings in
the mouse may be relevant to human cancer.
We have begun to define the functions of FAP+
cells in several normal tissues, including skeletal
muscle in which we have shown that they are
required for the maintenance of normal muscle
mass. Remarkably, cancer may also affect some
tissue homeostatic functions of FAP+ cells, in that
in two mouse models of cancer-induced cachexia,
which is the loss of skeletal muscle mass that
may occur independently of food intake, FAP+
cell numbers are decreased in skeletal muscle,
perhaps accounting for the cachexia. Cachexia is
a serious clinical problem, and these findings may
lead to an improved understanding of this process.
Next steps
The depletion of FAP+ cells is not a reasonable
option for enhancing the ability of the immune
system to control tumour growth because
they are necessary for the functions of normal
tissues. Therefore, we must determine the
molecular basis of the immune suppressive
function of the tumoural FAP+ cell, and develop
therapies that will interrupt this function. Our
strategy is to identify among the genes that are
selectively expressed in the tumoural FAP+ cells
candidates for immune suppression. We are also
determining how FAP+ cells accumulate in the
tumour. Conceivably, they may be generated
by replication from FAP+ cells in the local tissue,
or they may come from another site, such as
the bone marrow, where we have shown them
to proliferate. Either of these two research
directions may lead to therapeutic opportunities
for enhancing immune control of tumour growth.
Publications listed on page 74
Research Groups | 23
Centrosomes, Microtubules and Cancer
www.cambridgecancer.org.uk/fannigergely
The work in our laboratory focuses on the centrosome, an
organelle best known for its role as a major microtubule
organising centre.
Group Leader
Fanni Gergely
Graduate Students
Monika Pütz
Joo-Hee Sir
Postdoctoral Scientists
Hani Ebrahimi†
Pavithra Lakshminarasimhan
Senior Scientific Officers
Nimesh Joseph*
Deborah Zyss†
Figure 1
CEP63-CEP152 appears as a
discrete ring in centrosomes.
Images on left show distribution of
CEP63 (red in merge, top panels)
and CEP152 (red in merge, bottom
panels) within centrosomes.
Centrioles are marked with the
distal end marker, centrin (green in
merge). The top panels depict two
centrosomes each containing two
centrin dots (corresponding to a pair
of centrioles) and one CEP63 ring.
Schematic on right shows position
of CEP63-CEP152 ring on mother
centrioles in relation to centrin.
*
joined in 2011
†
Emerging evidence, however, suggests that the
centrosome also acts as a communication hub
that spatially concentrates diverse signalling
pathways. While centrosome number and
function are strictly regulated within healthy
cells, tumours display a multitude of centrosome
abnormalities. How such anomalies contribute
to tumourigenesis is an important and as yet
unresolved question.
In most normal cells the centrosome is
composed of a pair of cylindrical structures,
the centrioles, which are embedded in an
electron-dense amorphous matrix, the
pericentriolar material. The latter provides the
site for microtubule nucleation and therefore
strongly influences microtubule numbers
and organisation throughout the cell cycle.
Proteomic studies of whole human centrosomes
suggest that the organelle contains up to 300
proteins, many with unknown function. Like
DNA, the centrosome duplicates in S-phase.
This process is tightly controlled, since abnormal
centrosome numbers cause mitotic spindle
defects, culminating in mitotic catastrophe or
loss of genome stability.
Tumours exhibit a wide variety of centrosome
abnormalities that range from numerical
and structural, to functional and positional
aberrations. It is not well understood how
centrin
CEP63
left in 2011
24 | Cambridge Research Institute Scientific Report 2011
Identification of a protein complex involved in
centriole formation
Centrosomes duplicate once and only once
per cell cycle. In brief, the process involves
the formation of a single procentriole next
to each parental centriole and its subsequent
elongation. While the morphological changes
that occur during centrosome duplication are
well documented, our understanding of the
molecular pathway responsible for the timely
assembly of one and only one procentriole
per parental centriole in each cell cycle is still
far from being complete. Our most recent
work reveals a new regulator of procentriole
merge
0.5µm
centrin
these abnormalities arise in cancer and how
they contribute to tumourigenesis. We have
two basic goals in the laboratory. First, we
want to address how centrosome biogenesis
and function are controlled in normal cells. In
addition, our studies will provide insight into
how deregulation of particular centrosomal
proteins affects cell growth, mitotic spindle
formation and genome stability. Second, we
want to identify the molecular mechanisms
and signals that govern centrosome positioning
and microtubule nucleation in response to
environmental cues. These goals aim to provide
insight into basic biological processes whose
de-regulation is implicated in the development
of cancer.
daughter
centriole
CEP152
centrin
pericentriolar
matrix
merge
CEP63CEP152
mother
centriole
Figure 2
CK1δ is required for centrosome
positioning in lymphocytes. (A)
A schematic model shows how
the T cell cytoskeleton becomes
re-organised upon activation
by an antigen-presenting cell.
Centrosome translocation to
the immunological synapse is
thought to rely on a pulling force
(purple arrows) at the synapse
that acts on long centrosomal
microtubules. (B) Top panel shows
an immunological synapse formed
between a Jurkat T cell and Raji B
cell pulsed with superantigen. Note
that the centrosome is located in
the centre of the immunological
synapse. Bottom panel reveals
an immunological synapse formed
between a CK1δ-depleted Jurkat
T cell and a Raji B cell. While the
immunological synapse appears
normal in CK1δ-depleted cells, the
centrosome does not translocate
to the synapse. Centrosome
and synapse are marked with
antibodies against CDK5RAP2
and CD3, respectively. The Raji
B cell is stained blue. Fluorescent
signals are overlaid onto phase
contrast microscopy images to
aid identification of individual
cells. (C) A schematic depiction
of microtubule organisation in
CK1δ-depleted T cells. We propose
that the failure of centrosome
translocation in these cells is due
to the absence of long centrosomal
microtubules.
A
Antigenpresenting cell
B
centrosome
synapse
T-cell
Control
B-cell
Immunological
synapse formation
T-cell
CK1δdepleted
Centrosome
translocation
T-cell
C
CK1δ-depleted
formation – the core centrosomal protein,
CEP63 (Sir et al., Nat Genet 2011; 43: 1147).
We generated cells that lack functional
CEP63. CEP63 mutant cells grew more
slowly than control cells and displayed a high
incidence of monopolar spindles as a result
of abortive centrosome duplication cycles.
We subsequently discovered that CEP63
formed a molecular complex with CEP152, an
evolutionarily highly conserved protein required
for initiating procentriole assembly. Using
super-resolution microscopy we were able to
visualise the sub-centrosomal localisation of the
CEP63-CEP152 complex. The two proteins
form a discrete ring-shaped structure at the
proximal end of parental centrioles, seemingly
occupying a space near the centriole walls, a
site implicated in both procentriole formation
and centriole cohesion (Figure 1). Our study
revealed that the role of CEP63 in centrosome
duplication is to enrich and organise CEP152 at
the centrosome. Inherited mutations in CEP63
and CEP152 lead to loss of genome stability and
severe neurodevelopmental defects in humans.
Our aims are now to establish the molecular
framework of CEP63-CEP152 function and
address whether de-regulation of this protein
complex occurs in cancer.
Centrosome positioning and symmetry
breaking events
Cell polarity plays important roles in regulating
cell division, migration, differentiation and
organogenesis. Impaired cell polarity is a
hallmark of cancers. The actin and microtubule
cytoskeletons are inherently polar structures
that are well suited to generate and/or
maintain cell polarity, in particular by assisting
the asymmetric distribution of subcellular
organelles and components. Centrosomes
nucleate symmetric arrays of microtubules, yet
controlling the position of centrosomes within
cells can generate asymmetric microtubule
organisation. How changes in cell polarity are
coupled to centrosome positioning is not well
defined. We have chosen T lymphocytes as
our model system to study this problem. T cell
activation by an antigen-presenting cell results
in the assembly of a specialised cell-cell junction,
the immunological synapse (Figure 2A). During
synapse formation, a symmetric microtubule
network is reorganised into an asymmetric one
as a result of rapid centrosome translocation
from the geometrical centre of the cell to the
immunological synapse. This reorganisation is
important for a normal immune response, as
it underlies targeted cell killing and cytokine
release by cytotoxic and helper T cells,
respectively. We have identified a member
of the serine-threonine casein kinase 1 family,
CK1δ, as a regulator of centrosome positioning
in human T lymphocytes (Figure 2B) (Zyss
et al., J Cell Biol 2011; 195: 781). Importantly,
CK1δ is not required for the assembly of
the immunological synapse, indicating that it
acts downstream of synapse formation. We
uncovered that CKIδ regulates microtubule
behaviour in T cells, and postulated that the
kinase could be involved in producing long
stable microtubules necessary for centrosome
translocation (Figure 2C). Our recent findings
indicate that CKI family members control
microtubule behaviour not only in lymphocytes
but also in epithelial cells, pointing to a more
universal role for these kinases in microtubulerelated processes.
Publications listed on page 74
Research Groups | 25
Magnetic Resonance Imaging and Spectroscopy (MRI
and MRS)
www.cambridgecancer.org.uk/johngriffiths
Magnetic resonance imaging and spectroscopy (MRI and MRS)
have many uses in cancer research. We use these methods, both
in the laboratory and in patients, to study basic cancer biology, to
improve non-invasive methods for tumour diagnosis and grading,
to personalise therapy to individual patients, and to develop
biomarkers for monitoring the action of anticancer drugs.
Group Leader
John Griffiths
Associate Scientist
Marion Stubbs
Graduate Students
Nicola Ainsworth
Leanne Bell
Anna Brown*
Sara Dietz
Shen-Han Lee
Tonci Sustic † (with M Narita)
Postdoctoral Scientist
Davina Honess
Principal Scientific Officers
Madhu Basetti
Dominick McIntyre
Senior Scientific Officer
Loreta Rodrigues
Staff Scientist
Mary McLean
Visiting Student
Daniel Lopez Martinez*†
Visiting Worker
Monika Golinska
*
joined in 2011
†
left in 2011
Tumour biology
Monika Golinska completed her PhD on
metabolic adaptation of cancer cells to a
non-functional HIF-1 pathway. HIF-1, which is
upregulated in many cancers and accelerates
their growth, is a cancer drug development
target. Monika showed that cancer cells in
which the expression of glycolytic enzymes
is downregulated because of the absence of
active HIF-1, can still maintain flux through
the glycolytic pathway by allosteric control
of phosphofructokinase-1. This suggests that
monitoring tumour glycolysis by FDG-PET
would not necessarily indicate whether drugs
are working in a patient. A paper was published
this year and another is in preparation.
In a collaborative project with Adrian Harris
(University of Oxford), Shen-Han Lee, a
PhD student, is studying the role of carbonic
anhydrase IX, an enzyme overexpressed in
many cancers, on tumour extracellular pH (pHe).
Using ISUCA, a pH sensitive MRS probe, ShenHan has shown that tumours that overexpress
carbonic anhydrase IX have significantly lower
pHe. His results suggest a mechanism whereby
tumours could maintain a zone of extracellular
fluid around themselves at a fixed, acidic pHe.
Metabolomics
Madhu Basetti leads several metabolomics
projects, including a study of cellular senescence
and malignant transformation in collaboration
with the Narita laboratory. With the Tavaré
group, Madhu has implemented a novel method
of metabolite-metabolite correlation analysis
that has demonstrated numerous unexpected
but statistically significant metabolic interactions,
many of which are altered by the induction of
senescence or malignant transformation. This
method gives us a unique way of eavesdropping
26 | Cambridge Research Institute Scientific Report 2011
on the complex metabolic mechanisms by
which cells maintain homeostasis. Figure 1
shows the effects of malignant transformation
on the correlations involving choline, one of 16
metabolites measured in the study.
Sara Dietz, a joint PhD student with Colin Watts
(Department of Neurosurgery) is characterising
the metabolome in stem-like cells derived from
human glioblastoma multiforme tissue, and in
cell lines produced by inducing differentiation in
these cells and then returning them to a stemlike phenotype. Sara has observed marked
metabolic differences between the stem-like cells
and the differentiated cells, most of which revert
to the stem-like metabolic phenotype when the
cells are returned to the stem-like state.
We performed a study with Vincent Zecchini
(Neal laboratory), on the metabolic effects
of beta-arrestin-1 (ARRB1). This scaffolding
protein modulates HIF1A-dependent
transcription, and we showed that it
shifts cellular metabolism from oxidative
phosphorylation to aerobic glycolysis. A paper
has been submitted. Another study with the
Neal laboratory on the androgen receptor in
prostate cancer showed that AR co-ordinately
regulated energy production and biosynthesis
at multiple levels, highlighting several metabolic
pathways as potential drug targets. A joint
paper has been published.
Preclinical MRI and MRS
Preclinical MRI and MRS studies are led by
Dominick McIntyre, together with Davina
Honess. Leanne Bell’s PhD project is on the
Tuveson laboratory’s KPC pancreatic tumour.
That model, like human pancreatic tumours,
responds poorly to gemcitabine, the current
standard of care for this cancer, probably because
C
10
Count
t-glucose
lactate
alanine
Choline
PC
GPC
PCr
Cr
atp+adp
glutamate
Glutamine
Glycine
valine
iso-leucine
leucine
Threonine
5
E1A/RAS transformed HDF
t-glucose
lactate
alanine
Choline
PC
GPC
PCr
Cr
atp+adp
glutamate
Glutamine
Glycine
valine
iso-leucine
leucine
Threonine
0
B
Control HDF
−0.5
0
Value
0.5
1
t-glucose
lactate
alanine
Choline
PC
GPC
PCr
Cr
atp+adp
glutamate
Glutamine
Glycine
valine
iso-leucine
leucine
Threonine
A
t-glucose
lactate
alanine
Choline
PC
GPC
PCr
Cr
atp+adp
glutamate
Glutamine
Glycine
valine
iso-leucine
leucine
Threonine
Figure 1
Metabolite-Metabolite Correlation
Analysis of the effect of malignant
transformation on correlations
between choline and other
metabolites in human diploid
fibroblasts (HDFs). (A) and (B):
heatmaps of molecular correlations
(all p<0.001) for normal HDFs
(n=53) and HDFs malignantly
transformed by transfection with
E1A and Ras (n=52). (C) and (D):
diagrams illustrating the major
correlations; red and blue arrows
indicate positive and negative
correlations respectively.
D
Phosphocreatine
Choline
Choline
Glutamate
Glycerophosphocholine
Isoleucine
Phosphocholine
Glutamate
Lactate
Phosphocholine
Valine
of the dense, collagenous tumour matrix. Leanne
has been developing ways to monitor this matrix
in different types of KPC tumours, using DCEMRI and magnetisation transfer MRI so as to
monitor drugs that break down the collagenous
matrix and enhance the action of gemcitabine.
She is confirming the MR measurements with
Second Harmonic Generation imaging of collagen
content of tumour sections with Lorraine Berry
(Microscopy Core) and studying the collagnenous
matrix ex vivo by HR-MAS, with Madhu Basetti.
Nicola Ainsworth, a clinical research fellow
(jointly supervised by Jonathan Gillard,
Department of Radiology, and in collaboration
with Susan Harden, Department of Oncology),
is studying cerebral metastasis of small cell lung
cancer (SCLC). About half of patients with
SCLC develop cerebral metastases, but since we
cannot predict which half, the current practice is
to give them all prophylactic cranial irradiation,
a therapy with significant long-term side
effects. Nicola’s project aims to develop MR
methods for detecting these metastases much
earlier, so that patients who would not benefit
can be spared irradiation therapy. Nicola has
developed a mouse model of brain metastasis
and is developing various MRI methods in mice
and in patients. She is also recruiting SCLC
patients into a study (CLUB-01), in which they
are imaged before and after prophylactic cranial
radiotherapy. An M Phil student, Anna Brown, is
working on optimizing texture analysis software
for early detection of these brain metastases and
prediction of their development.
The Griffiths (Dominick McIntyre and Davina
Honess) and Brindle (Dmitry Soloviev and
Creatine
ATP + ADP
Isoleucine
Leucine
David Lewis) laboratories will jointly participate
in QuIC-ConCePT, an EU-funded project
under the Innovative Medicines Initiative. The
aim is to qualify existing imaging methods for
use in anticancer drug trials, firstly in the KPC
pancreatic tumour model. Kathrin Heinzmann
will be the postdoc working on this project.
Clinical MRI and MRS
Mary McLean leads our work on tumours
in patients. We are collaborating with James
Brenton (CRI) and Evis Sala (Department
of Radiology) in an MRI and MRS study
(OVO3) on the response of ovarian cancer to
chemotherapy. Diffusion MRI was the best and
quickest marker of tumour response. Three
papers have been published and another is
in press. Another collaboration with Evis
Sala, Vincent Gnanapragasam (Department
of Surgery) and David Neal (CRI) uses DWI,
DCE-MRI and MRS for the prediction and
early detection of prostate cancer response
to androgen deprivation in advanced prostate
cancer. One paper is in print, one in press.
Sidhartha Nagala (Department of Head & Neck
Surgery) is taking a PhD supervised by Jonathan
Gillard (Department of Radiology) on the use
of MRS and DWI for the diagnosis of cancer
in follicular thyroid nodules and parotid lumps.
Tumour biopsies from these examinations will
be studied by 1H HRMAS NMR to correlate
metabolomic and clinical findings. Accurate
preoperative diagnosis, which is difficult for
these lesions, will enhance surgical planning as
well as reducing unnecessary operations.
Publications listed on page 75
Research Groups | 27
Pharmacology and Drug Development
www.cambridgecancer.org.uk/duncanjodrell
The aims of the Pharmacology and Drug Development Group
(PDDG) are to optimise the pre-clinical development and
science-led clinical application of novel therapies, including ‘first
into man’ (phase I) studies.
Group Leader
Duncan Jodrell
Clinical Fellows
Lucy Gossage
Jenny Harrington
Graduate Students
Sarah Eastmond (with D Tuveson)†
Ciorsdaidh Watts
Ruiling Xu*
Research Assistants
Tashinga Bapiro
Jo-Anne Bramhall
Research Associates
Aurelie Courtin
Ben-Fillippo Krippendorff
Yao Lin
Senior Research Associate
Frances Richards
Visiting Students
Tambudzai Shamu*
Aisha Tahira*†
Ramone Williams†
*
joined in 2011
†
left in 2011
Pre-clinical models are used to inform clinical
trial design for novel agents and combination
strategies. The PDDG is closely linked with
the Early Phase Clinical Trials Team (EPCTT)
based in the Cambridge Cancer Trials Centre, at
Addenbrooke’s Hospital.
In the laboratory, we tend to use model
systems representing pancreatic cancer, which
complements our clinical interests: Duncan
Jodrell is a member of the clinical team at
Addenbrooke’s who care for pancreatic cancer
patients, and is also the Deputy Director of
the Cambridge Pancreatic Cancer Centre
(http://www.cambridge-pcc.org). Pancreatic
cancer is also a major unmet clinical need and
a priority cancer for Cancer Research UK.
Through collaboration with David Tuveson
(CRI), we have access to the KPC GEM
(genetically engineered mouse) model. Using
our novel LC-MS assay, developed initially for
use in that model, we are assessing modulation
of gemcitabine delivery to tumour tissue in
various combination treatment regimes (Bapiro
et al., Cancer Chemother Pharmacol 2011; 68:
1243). In addition, we are learning more
about the metabolic pathways involved in the
processing of gemcitabine and studying how
newly discovered metabolites might impact
on its anti-tumour activity. Using the same
KPC models, we previously demonstrated
that the orally administered fluoropyrimidine
capecitabine is taken up into the tumours and
has activity comparable to gemcitabine. We
have now shown that capecitabine shows similar
efficacy to gemcitabine in the KPC model.
We are also assessing potential combination
strategies incorporating fluoropyrimidines, in
addition to studies with gemcitabine.
A clinical study assessing the pharmacokinetics
(PK) of capecitabine in patients following surgery
for pancreatic cancer has been completed and
the results will be published shortly. This study
28 | Cambridge Research Institute Scientific Report 2011
incorporated pharmacogenetic and functional
assessment of patients’ capecitabine metabolising
capacity, which will be linked to the PK data. In
addition to activating capecitabine, the enzyme
cytidine deaminase (CDA) is responsible for
the deactivation of gemcitabine and therefore
is of particular relevance to the treatment of
patients with pancreatic cancer. Together with
international collaborators, we intend to assess
whether the activity and/or genotype of CDA
can be used to guide therapy in patients.
A specific theme in our pre-clinical work is
the assessment of combination strategies.
We feel that current clinical trial design for
evaluating drug combinations may lead to missed
opportunities, unless pre-clinical data are used
to guide their design. We are taking two new
approaches to evaluating these data. We are
using mathematical models of the cell cycle,
receptor/ligand interactions and the spindle
assembly checkpoint to guide the pre-clinical
studies we perform. We are also using model
based approaches to evaluate pre-clinical
growth inhibition data and identify potentially
synergistic ‘dose’ ratios of compounds by
generating surfaces of interaction, as opposed to
simply trying to combine the maximum tolerable
doses of both agents when used as single agents
(Figure 1). We are generating such surfaces
using both cancer cell lines and human myeloid
(white cell) pre-cursor models to identify cell
lineage dependent differences in response, with
the aim of maximising the therapeutic index. In
collaboration with the MRC Trial Methodology
Hub (Adrian Mander and colleagues), we are
assessing novel adaptive Phase I trial designs that
will be informed by these pre-clinical studies.
We are currently utilising an adaptive design
in an ongoing Phase I trial (Whitehead et al.,
Statistics in Medicine 2012; In press).
An example of the application of these
combination approaches are studies using novel
Figure 1
Generating surfaces of interaction
for 2-drug combinations in a 96 well
plate format
Drug A
Drug B
%Inhibition
Single agents
Drug A
Drug B
Region of
synergy
Region of
antagonism
87
94
94
99
101
99
99
98
600
87
87
87
87
87
89
96
99
600
0
7
7
12
14
10
3
−2
400
66
77
75
84
85
83
88
78
400
66
67
67
66
66
72
89
98
400
0
9
8
18
19
11
−1
−20
200
25
54
52
57
55
59
55
54
200
25
27
25
25
25
37
75
96
200
0
27
27
32
31
22
−20
−42
100
17
50
54
50
47
45
45
43
100
17
20
18
17
17
30
72
96
100
0
30
36
32
29
15
−27
−53
60
13
38
40
47
38
43
47
42
60
13
16
14
13
13
27
71
96
60
0
22
27
34
25
16
−24
−54
30
12
34
37
36
38
47
59
61
30
12
14
12
12
12
26
70
96
30
0
20
25
25
27
21
−11
−35
10
1
10
8
8
9
56
84
91
10
1
4
1
1
1
16
67
95
10
0
6
7
7
9
40
17
−4
0
0
3
1
0
0
16
67
95
0
0
3
1
0
0
16
67
95
0
0
0
0
0
0
0
0
0
0
0.1
0.3
0.6
1
3
6
10
0
0.1
0.3
0.6
1
3
6
10
0
0.1
0.3
0.6
1
3
6
10
−
Drug A
Data
=
Drug B
600
Drug B
Drug B
Additivity Model
Drug A
Prediction
Aurora kinase inhibitors. The Aurora family of
serine/threonine protein kinases plays a critical
role in cell division, with key roles in the mitotic
spindle checkpoint. AK-A has been identified
as a cancer susceptibility gene, and elevated
expression levels of AK are detected in many
different types of cancer. In addition, AK-A
over-expression is associated with resistance
to taxanes. In cell culture models (pancreatic
and urothelial), we have demonstrated, using
a novel Aurora A specific inhibitor, regions
of both synergy and antagonism in growth
inhibition combination surfaces and we are now
proceeding to design in vivo experiments to test
these findings further.
In general, it is assumed that combinations of
agents have similar effects on normal and tumour
cells, but this is not always the case. An optimal
combination would lead to synergy in cancer cells
and antagonism in normal cells, reducing the toxic
side effects that often limit dosing. In studies
with normal myeloid precursors (CFU-GM), we
have demonstrated that the synergistic effects
of combining an AK-A inhibitor and paclitaxel
are not seen in non-malignant cells. This project
is also utilising a mathematical model of the
spindle assembly checkpoint to predict drug
effects, through collaboration with Bob Jackson
(Pharmacometrics Ltd). We ultimately intend to
extend our pre‑clinical findings into clinical trials
and a protocol is currently in the design phase.
Drug A
Data-Prediction
As a result of our collaboration with Steve Ley
and Rebecca Myers (Department of Chemistry)
and Fanni Gergely (CRI), novel, selective
allosteric inhibitors of the kinesin motor
protein HSET have been synthesised and we
are proceeding to the biological evaluation
of these compounds in cell line models. Our
collaboration with Gillian Murphy (CRI)
continues to investigate TNF-α converting
enzymes (TACE) as therapeutic targets. In
the evaluation of a novel antibody targeting
ADAM17, favourable pharmacokinetics and
encouraging pharmacodynamic read-outs have
been achieved and efficacy studies are ongoing.
In the last year, the EPCTT has completed a
collaborative combination Phase I trial, in addition
to the capecitabine PK study discussed above.
Currently, ten studies are recruiting patients; two
combination phase I trials, a further four single
agent phase I trials, three biomarker trials and a
PK study. We are continuing to explore novel
PET and MR approaches in our trials and have
recently collaborated with Kevin Brindle in a
successful Wellcome Trust application to support
the clinical development of hyperpolarised
13
C pyruvate based PD studies. Two further
protocols (one combination phase I and a PK
study in patients with impaired renal function)
are in the set-up phase for initiation in 2012.
Publications listed on page 75
Research Groups | 29
Computational Biology
www.cambridgecancer.org.uk/florianmarkowetz
The Markowetz lab develops algorithms and statistics to
leverage complex and heterogeneous data sources for
biomedical research. Our main research question is: How do
perturbations to cellular mechanisms shape phenotypes?
Visiting Worker
Eric Tsz Him Lai*†
Natural perturbations: Capturing the
heterogeneity of somatic alterations
Natural perturbations like copy number
alterations can promote cancer development,
which is a complex process involving the
accumulation of multiple mutations and genomic
aberrations. A consequence of these alterations
is the deregulation of cell signalling pathways
central to the control of cell growth, cell fate
and other important cellular functions. With
our partners at the CRI we aim to characterise
disruptions of signalling pathways in tumours
and to identify genomic alterations that drive
tumour evolution. Identifying drivers of cancer
requires dissecting tumour heterogeneity at all
levels.
joined in 2011
Population heterogeneity. At the population
level we dissect heterogeneity by statistical
stratification methods to define prognostic
subgroups. Prominent approaches for subtype
definition combine information from different
molecular levels, for example data on DNA
copy number changes with data on mRNA
expression changes. We have used such
integrated analyses to identify breast cancer
sub-types in the METABRIC collection (in
collaboration with Carlos Caldas and others;
Curtis et al., Nature 2012; In press). We
have also proposed a unified model that at
the same time fuses different data types,
finds informative features, and estimates the
number of subtypes in the data (Yuan et al.,
PLoS Comput Biol 2011; 7: e1002227). The
main strength of our model comes from the
fact that we assess for each patient whether
the different data agree on a subtype or
not. Competing methods combine the data
without checking for concordance of signals. In
breast and prostate cancer we have found that
concordance of signals has strong influence on
subtype definition and that our model allows
defining prognostic subtypes that would have
been missed otherwise.
Group Leader
Florian Markowetz
Graduate Students
Anne Trinh*
Xin Wang
Postdoctoral Scientists
Gökmen Altay (with D Neal)
Mauro Castro (with B Ponder)
Roland Schwarz
Yinyin Yuan
Visiting Student
Max Homilius*†
*
†
left in 2011
30 | Cambridge Research Institute Scientific Report 2011
Regulatory heterogeneity. To understand the
biology underlying cancer sub-types even
further, we have developed methods to unravel
regulatory heterogeneity between sub-types.
We have focussed on identifying regulatory
networks of copy-number driven gene
expression that reveal putative breast cancer
oncogenes (Yuan et al., IEEE TCBB 2011; Epub
20 July) and to elucidate aberration hotspots
mediating subtype-specific transcriptional
responses (Yuan et al., Bioinformatics 2011;
27: 2679). In collaboration with David Neal’s
group we have developed methods to identify
deregulation of cellular networks (Altay et al.,
BMC Bioinformatics 2011; 12: 296).
Intra-patient heterogeneity. Cancers are
not only heterogeneous on the population
level, but can evolve and change within a
single patient. To capture this intra-patient
heterogeneity and identify early driver events,
we have developed phylogenetic models
applicable to copy-number profiles based on
finite-state transducers (Schwarz et al., PLoS
ONE 2010; 5: e15788). We are currently
applying our methods to ovarian cancer in
collaboration with James Brenton’s lab.
Cellular heterogeneity. Solid tumours are
complex mixtures of cell types, which is rarely
taken into account when analyzing molecular
profiles of tumour samples, because deconvoluting high-dimensional cancer data
is almost impossible without knowing the
precise cellular composition of each sample.
Additionally, tissue architecture is generally
not reflected at all in molecular profiles. We
have addressed both problems by leveraging
a commonly available, but quantitatively
largely neglected, source of information:
H&E-stained images of tumor sections (Yuan
et al., submitted; Figure 1). We have shown
how histopathological images de-convolute
mixed signals in molecular data and identify
a
Supervised
classification
Cell types
and location
c
Cancer density
0
1
●
●
●●
● ●●
●●
●● ●
●●
●
●
●●
●
●
●●
●
● ●
●
●●●●
●
●● ●●●
● ●
●●● ●
●● ●
● ●●
●●
●
●● ●
● ●● ●
●
●●●● ● ●●●● ●
●
●●
●
●●
●
●
●●
●
● ● ● ●●
● ●●●
●●
●●
● ●
●● ●● ●● ●●●
●
●
● ●
●
●●● ●●
●
●
●
●●
●●●
● ●●
●
●
● ●●●●●
●●
●
●
●
●
●
●
●
●●●
●
● ● ●
● ●●●
●● ●
●
● ● ●●
●●
●●●●
●●
●● ● ●
●
● ●●
●● ●
●
●
●
●●●
● ● ●● ●
●
● ●●
●
●
●
●●
●●●●●
●●● ● ●
●
●●●●●●● ●
●●
●
●
●
●
●
●
●
●
●
●
●●● ● ●● ● ● ● ●
●●
●
● ● ●●● ●
●●
●●●● ●
●
● ● ●
●
●
●
●●
● ● ●● ●
●
●●
● ●
●●
●
●
● ●● ● ● ●
●●
●
●
●
●
●
●
●
●
●
●
●
●
●
●
●
●
●
str
om
al
b Original
Spatial
smoothing
●
cer
can
Figure 1
Quantitative analysis of tumour
heterogeneity. (A) In a stained
tumour image individual nuclei
are automatically classified into
different cell types, showing the
frequency of cell types and their
locations in the tumour sample.
(B) This information can be used
to highlight spatial patterns in the
tumour architecture, for example
here the distribution of cancer
cells in the sample. (C) A triangle
plot showing frequencies of cancer
cells, stromal cells and lymphocytes
(axes) in 323 samples (dots).
●
●
1
low
prognostic features of tumour architecture.
In a cohort of more than 300 breast cancers,
our approach led to profound insights that will
advance translational medicine. We developed
a novel algorithm to correct copy number data
for cellularity and an integrated pathologicalgenomic predictor for survival. Most
importantly, we found surprising prognostic
value in the spatial organization of the tumour
tissue architecture.
Experimental perturbations: RNA interference
(RNAi)
Experimental perturbations like RNAi are
key approaches at the forefront of functional
genomics. A goal that is becoming more and
more prominent in both experimental as well
as in computational research is to leverage
gene perturbation screens to the identification
of molecular interactions, cellular pathways
and regulatory mechanisms. Research focus is
shifting from understanding the phenotypes of
single proteins to understanding how proteins
fulfil their function, what other proteins they
interact with and where they act in a pathway.
Novel ideas on how to use perturbation
screens to uncover cellular wiring diagrams can
lead to a better understanding of how cellular
networks are deregulated in diseases like
cancer.
In our group we work on several projects to
analyse gene perturbation screens in terms
of pathways and cellular networks. We have
developed a methodology for the network
analysis of high-throughput RNAi screens
(Markowetz, PLoS Comp Biol 2010; 6: e1000655;
Wang et al., Bioinformatics 2011; 27: 879) and
work on methods for network reconstruction.
For example, in collaboration with Klaas Mulder
in Fiona Watt’s lab we combined siRNA-based
genetic screening and computational approaches
to map putative functional interactions among
332 chromatin modifiers in primary human
epidermal stem cells. Detailed analysis of
a significant sub-network revealed a high
degree of true genetic interactions among its
components, which physically associated with
distinct, yet functionally equivalent gene sets.
high
0
lymphocytes
0
1
The functional redundancy in gene expression
programs conferred by the epigenetic network
we have identified thus protects stem cells from
differentiation (Mulder et al., in revision).
A particular focus of the lab is on nested
effects models (NEMs), a statistical approach
that is specifically tailored to reconstruct
features of pathways from perturbation effects
in downstream reporters (Markowetz et al.,
Bioinformatics 2007; 23: i305). Based on NEMs,
we are developing an integrated experimental
and computational approach to identify new
branches in the NFκB pathway, a key pathway
involved in the immune response as well as in
cancer, working in collaboration with the group
of Thomas Meyer (Max-Planck Institute for
Infection Biology, Berlin).
The theory of NEMs has so far been mainly
limited to static snapshots of perturbation
effects. In a major conceptual improvement,
Xin Wang, a PhD student in the group, has
combined hidden Markov models with NEMs
to reconstruct rewiring events in pathway
topologies from time-series data derived
after silencing pathway components. We are
applying this method to gene expression timeseries in mouse embryonic stem cells to infer
changes in pathway activity in the early stages of
differentiation.
In the future, our lab plans to strengthen
its ties with our experimental collaborators
in order to approach pivotal questions in
biology and medicine by computationally
guided experimentation. Biological and clinical
questions motivate the development of novel
statistical algorithms, which guide the next
round of experiments.
Publications listed on page 76
Research Groups | 31
Proteases and the Tumour Microenvironment
www.cambridgecancer.org.uk/gillianmurphy
Group Leader
Gillian Murphy
Graduate Students
Pedro Correa De Sampaio
Chris Tape
Postdoctoral Scientists
Marton Fogarasi†
Yanchao Huang*
Hang Fai (Henry) Kwok *
Anthea Messent †
Magdalini Rapti*
Research Assistant
Elizabeth Shedden†
Visiting Student
Salvatore Santamaria†
Visiting Workers
Patricia Eisenach†
Magdalena Kielbas*†
Malcolm Lawson
Peter Stanley †
*
joined in 2011
†
left in 2011
Extracellular proteases are key players in the regulation of the
cellular environment, acting as major effectors of both cell-cell
and cell-extracellular matrix (ECM) interactions, essentially
as ‘signalling scissors’. Epithelial tumours evolve in a multistep
manner, involving both inflammatory and mesenchymal cells.
Although intrinsic factors drive malignant progression, the
microenvironment of neoplastic cells is a major feature of
tumourigenesis. Our premise – that proteases are integral
to the regulation of extrinsic effectors – is the basis for our
work and for our plans to dissect events at the cellular and
molecular level, as well as proceeding to complex tumour
models addressing tumour-stromal interactions. Based on our
findings we are developing and evaluating novel approaches to
understand the regulation of proteases in tumour systems.
Understanding the roles of proteases in
tumour biology
The successful development of tumours is
determined by the tissue environment in
which the ‘host’ participates in the induction,
selection and expansion of the neoplastic cells.
Malignant tumour cells recruit vasculature
and stroma through the production of
stimulatory factors. The locally activated host
environment (both cells and extracellular
matrix) in turn modifies the proliferative and
invasive behaviour of tumour cells. The nature
and function of the activating factors involved
and the subsequent effectors are important
areas of basic biological research in the field
of cancer studies. Extracellular proteases are
major effectors of both cell-cell and cell-ECM
interactions, modifying ECM integrity, growth
factor availability and the function of cell surface
signalling systems, with consequent effects
on cellular differentiation, proliferation and
apoptosis. Early data from screens of cancer
tissues have shown that different patterns
of protease elevation occur and that the
relationship of expression to tumour progression
and the contribution of individual cell types –
tumour cells, fibroblasts, endothelial cells and
inflammatory cells – requires detailed dissection.
A major aim of the drive to understand protease
32 | Cambridge Research Institute Scientific Report 2011
biology within a specific tumour environment
relates to the need to assess potential targets
within the interface between tumour cells and
the ‘host’ cells that may be appropriate for
therapeutic intervention. It is anticipated that
the understanding and the manipulation of
protease function will give clear leads as to the
critical stages in the breakdown of the normal
tissue-cell ‘society’ that occurs in neoplasia.
Within this remit our research is focussed on cell
surface associated forms of the zinc-dependent
proteases, notably the membrane type matrix
metalloproteinase-1 (MMP14) and members
of the disintegrin‑type metalloproteinases
(ADAM). We aim to elucidate how these
metalloproteases function in the regulation of
extrinsic effectors at the cellular and molecular
level, as well as proceeding to complex tumour
models addressing tumour-stromal interactions.
The fundamental data accrued will drive the
development of novel reagents for disease
therapy and diagnosis.
Membrane associated metalloproteases
The membrane type 1 matrix metalloproteinase
MT1 MMP plays a major role in tumourigenesis,
including tumour cell migration, aspects of
stromal cell function and angiogenesis. As a
A
VEGFR-2
MT1-MMP
AntipY416-Src
AntiVEGFR-2
AntiMT1-MMP
+ pcDNA3.1
+ Src
B
+ MT1-MMP
+ Src
Figure 1
MT1-MMP forms a ternary complex
with phospho-Src-Y416 and VEGFR-2
at the cell surface of MCF-7 cells
and modulates the subcellular
localization of VEGFR-2. (A) Model
of the MT1-MMP induced pathway.
The ternary MT1-MMP–pSrc-Y416–
VEGFR-2 complex was found to be
required for the MT1-MMP induced
up-regulation of VEGF-A expression
by activating the PI3Kinase–
Akt–mTOR pathway. (B) MCF-7
cells were transiently transfected
with either Src (upper panel) or
Src and MT1-WT (lower panel)
and the localization of pSrc-Y416,
VEGFR-2 and MT1-MMP was
observed by confocal microscopy.
The intracellular staining pattern
of VEGFR-2 in the absence of
MT1-MMP is shown by an empty
arrowhead. In MT1-MMP expressing
cells co-localisation is indicated by
arrows (Eisenach et al., J Cell Sci
2010; 123: 4182)
Src
PI3K
P
mTOR
P
Akt
P
P
VEGF-A
Merge
potential key target for therapeutic approaches
to cancer we are addressing its involvement
in intracellular signalling with a focus on the
role of its different domains in important
interactions. We have elucidated novel roles
for the MT1 MMP cytoplasmic domain in its
regulation during cellular trafficking and have
identified several intracellular and extracellular
interaction partners (Figure 1).
We are also collaboratively developing novel
inhibitors of MT1 MMP. In particular, the
characterisation of scFv antibodies that we have
isolated is being used to address the question
of the importance of exosite interactions in the
collagenolytic capacity of MT1 MMP. The effect
of enzyme inhibition in cell model systems,
such as the mini tumour (see below) have
demonstrated the key role of this enzyme.
The disintegrin metalloproteinases are also
regulators of cellular signalling and we are
studying ADAM17 in this respect. Biochemical
studies have focussed on the structure-function
relationships of ADAM17 and projects are in
progress on the role of different ADAM domains
in the proteolytic ‘shedding’ of cell surface
proteins. We are particularly interested in the
generation of soluble EGF receptor ligands which
are key drivers of a number of different tumour
types. The significance of ADAM activity in cell
models is being investigated using shRNA and
siRNA techniques and novel antibody tools that
we have recently developed. ADAM regulation
by G-protein coupled receptors and redox
mechanisms are being investigated. The roles
of ADAM10 and ADAM17 in the development
of ovarian and gut cancers are being evaluated
collaboratively using gene ablation or overexpression studies in animal models.
3D in vitro models
In order to carry out molecular studies and
inhibitor screens on the complexity of cells
within tumour tissue, we have set up several
more complex 3D models of interacting cells in
collagen gels. In particular we have developed
a novel in vitro “mini-tumour” angiogenesis
model, by co-culturing cancer cell lines with
primary human endothelial cells and fibroblasts.
All the cell types are in direct contact and in a
three dimensional system, in which pre-capillary
sprout formation can be easily quantified. This
spheroid model has been shown to promote
the development of pre-capillary sprouts after
36h, under the influence of the tumour cells and
independent of external growth factors. Sprout
formation was shown to respond to known
anti-angiogenic compounds and growth factor
receptor inhibitors similar to observations made
in clinical trials. Our major goal is to evaluate
metalloproteinase function in individual cell types
and the outcomes of their abrogation. The
use of function blocking antibodies and siRNA
or shRNA for our protease targets has been
evaluated. We can specifically target fibroblasts
prior to their inclusion in the model and
examine their specific role in supporting precapillary sprout formation. We have shown that
fibroblast MT1 MMP and ADAM10 expression
are key to angiogenic sprout formation in the
model and we are currently investigating their
roles in the mechanisms of fibroblast regulation
of endothelial cell behaviour.
Publications listed on page 76
Research Groups | 33
Genomic Imprinting and Cancer
www.cambridgecancer.org.uk/adelemurrell
DNA methylation, post translational histone modifications,
together with chromatin structure, underpin the epigenetic
organization of the genome which responds to the
environment and changes during tissue differentiation, cell
cycle, cell death, wound healing and neoplasia.
Group Leader
Adele Murrell
Graduate Students
Abdullah Al-Jeffery *
Malwina Niemczyk
Postdoctoral Scientists
Yoko Ito
Lovorka Stojic
Santiago Uribe-Lewis
Kathryn Woodfine†
Visiting Workers
Martin Bachman
Marjolein Droog*†
Paola Mirrar *†
Shani Mulholland†
Liang Wu
*
joined in 2011
†
left in 2011
Genomic imprinting is a fascinating epigenetic
process that marks the gametic origin of a
subset of genes and results in the expression
of one parental allele and the reciprocal
silencing of its homologue. Imprinted genes
are exceptionally stable in their maintenance of
DNA methylation (Woodfine et al., Epigenetics
Chromatin 2011; 4: 1), despite being expressed
in a tissue specific manner and resist DNA
methylation reprogramming during normal
development. This makes them tractable
markers for detecting abnormal epigenetic
reprogramming in cancer.
Imprinted genes are well characterised in terms
of their epigenetic marks and expression profiles
and are therefore an excellent model system in
which to study the impact of DNA methylation
and chromatin changes in cancer. Aberrant
imprinting of the insulin-like growth factor 2
(IGF2) gene locus is part of the aetiology of
congenital growth disorders such as Beckwith
Wiedemann syndrome (BWS, OMIM#130650),
as well as various human cancers including
Wilms’ tumour, rhabdomyosarcoma,
hepatoblastoma, colorectal and breast
carcinomas (Cooper et al., Eur J Hum Genet
2009; 13: 1025; Murrell, ScientificWorldJournal
2006; 6: 1888). Imprinting of IGF2 and H19 are
regulated by an insulator element upstream of
H19. The zinc finger protein CTCF binds to
the insulator and mediates its function, such
that when CTCF is bound on the unmethylated
maternal allele, the maternal copy of IGF2
cannot access enhancers downstream of the
H19 gene (Bell and Felsenfeld, Nature 2000;
405: 482). Methylation at the insulator sequence
on the paternal allele prevents CTCF binding,
inactivates the insulator function and enables the
IGF2 gene to access the enhancers.
We have previously shown that in mice
chromatin looping is mediated by CTCF binding
34 | Cambridge Research Institute Scientific Report 2011
at the insulator (Murrell et al., Nat Genet 2004;
36: 889), and have hypothesised that DNA
methylation of the insulator sequence would
result in differential loops on the maternal and
paternal allele. Subsequently, genome-wide
studies have shown that cohesin complexes
co-localise onto the same DNA sequences as
CTCF (Wendt et al., Nature 2008; 451: 796).
We therefore speculated that cohesin may
have a function in holding chromatin loops
together by connecting two DNA molecules
in cis, in an analogous manner to its role in
holding two sister chromatids together. We
found that CTCF sites upstream, within
and downstream of the locus interacted to
form looping domains. Cohesin depletion
significantly reduced the interaction frequencies
between CTCF binding sites suggesting that
cohesin is required for stabilisation of chromatin
loops. Interestingly when chromatin looping
conformation was destabilised IGF2 expression
was up-regulated and biallelically expressed,
despite methylation at the insulator sequence
not changing, indicating that chromatin
conformation changes can override epigenetic
imprinted information (Nativio et al., PLoS Genet
2009; 5: e1000739; Nativio et al., Hum Mol
Genet 2011; 20: 1363).
Others have confirmed that cancer cells have
changes in chromatin conformation at the
IGF2-H19 locus (Vu et al., Hum Mol Genet
2010; 19: 901). We previously showed a
disconnection between DNA methylation at
the IGF2 gene (the DMR0 region) and the H19
insulator in Wilms’ tumours (Murrell et al., PLoS
ONE 2008; 3: e1849), colorectal and breast
cancer (Ito et al., Hum Mol Genet 2008; 17:
2366). In colorectal cancer hypomethylation of
the IGF2 DMR0 is prevalent as an early event
and may potentially even be indicative of cancer
(Ito et al., Hum Mol Genet 2008; 17: 2633;
Ibrahim et al., Gut 2011; 60: 499).
Figure 1
Global reduction of
5-hydroxymethylcytidine (5hmC) in colon cancer associated
with rapid cell proliferation. (A)
Cycle of covalent modifications to
cytidine at the 5 C position. DNA
methyltransferases add a methyl
group to form 5-mC which can be
oxidised by TET or ELP enzymes to
form 5-hmC. Hydroxymethylated
cytidine may be further oxidised to
formyl and carboxyl cytidine (5-fC
and 5-cC) and finally repaired into
an unmethylated cytidine. 5-mC
and 5-hmC are also deaminated
and replaced into unmethylated
cytidine by DNA repair. (B) Global
5hmC levels measured on total
DNA extracted from colon samples
at sites away from a tumour
(normal away, NA), adjacent to
the tumour (normal close, NC),
adenoma (Ad) and tumour (T) in
two colorectal cancer patients. The
left-hand panel is a DNA stain
indicating DNA loading, the middle
panel is immunoblot probed with
an antibody for hydroxymethylated
DNA (hmC), and the right-hand
panel is the same blot probed with
an antibody to methylated DNA
(mC). (C) Spatial distribution of
5hmC in normal mouse intestine
– 5hmC is strongly present in
terminally differentiated epithelia
(long arrow) and weakly present
in dividing transit amplifying cells
(short arrow).
A
B
C
DNA repair
C
SAM
DNMTs
cC
SAH
fC
Deamination
and
Repair
hmC
mC
aKG,
Fe++
Methionine
ELPs
(radical SAM?)
SAM
Succinate
TETs
(oxidative
de-methylation)
Methylated cytosines (5mC) have recently
been shown to be a substrate for hydroxylases
that convert them to 5-hydroxymethylcytosine
(5hmC) (Tahiliani et al., Science 2009; 324: 930;
Ito et al., Science 2011: 333: 1300) and it is widely
postulated that 5hmC may be an intermediate
molecule during DNA demethylation. Since
global hypomethylation is a feature of several
cancers, we decided to look at 5hmC levels in a
set of well defined colorectal cancers consisting
of matched normal tissue hypoplastic polyps,
adenomas and also tumours (Figure 1). We
found that 5hmC levels are strikingly reduced
at early stage of carcinogenesis such as in
adenomas, but also in some hypoplastic polyps.
Our results further indicate that genes marked
by 5hmC are actively transcribed and protected
from DNA methylation changes in tumours.
It is now beginning to look as if 5hmC plays a
tissue specific role in gene regulation rather
than simply being an intermediate of DNA
demethylation and we are now actively looking
for the function and associated mechanism of
this newest epigenetic 5hmC mark.
Publications listed on page 76
Research Groups | 35
Mechanisms of Cellular Senescence
www.cambridgecancer.org.uk/masashinarita
Visiting Worker
Kevin Cheng
Cellular senescence is a state of stable cell cycle arrest with
active metabolism. Similar to apoptosis, senescence can be a
failsafe program against a variety of cellular insults. In contrast
to apoptosis, in which cytotoxic signals converge to a common
mechanism, senescence is typically a delayed stress response
involving multiple effector mechanisms. These effector
mechanisms include epigenetic regulation, the DNA damage
response, the senescence-associated secretion phenotype
(SASP) and autophagy. The relative contribution of these
effectors varies depending on the trigger and cell type, and it
is possible that the combination and balance of these effectors
determines the quality of the senescence phenotype. Thus, to
understand the senescence program, it is important to identify
new effector mechanisms and examine how they associate
with each other, and also to identify which effector mechanisms
could be potential targets for cancer therapy.
joined in 2011
Genome-wide analysis of heterochromatin
components in SAHF
Certain types of cells undergo distinct alterations
in chromatin structure during senescence, called
senescence-associated heterochromatic foci
(SAHF). SAHF have been widely used as a
marker of senescence, and more importantly,
several new components of senescence
machinery have been successfully identified using
SAHF as a readout. Thus, it is important to
understand SAHF structure in more detail and
how SAHF are actually formed. Interestingly,
we have shown that SAHF are indistinguishable
from the inactive X (Xi) chromosome, one
of the best studied heterochromatin models,
and other groups recently suggested that
each individual SAHF might represent each
chromosome territory. In contrast to Xi,
SAHF formation can be dynamically regulated
in normal human diploid fibroblasts (HDFs),
thus providing a unique tool to study not only
senescence, but also chromatin biology. To
further characterise SAHF in detail, we are
currently investigating a dynamic redistribution
of the specific histone modifications and their
adaptor proteins using confocal microscopy.
Group Leader
Masashi Narita
Graduate Students
Tamir Chandra†
Tonci Sustic † (with J Griffiths)
Postdoctoral Scientists
Matthew Hoare
Kristina Kirschner
Masako Narita
Mahito Sadaie
Rafik Salama
Andrew Young
*
†
left in 2011
36 | Cambridge Research Institute Scientific Report 2011
In addition, we are currently analysing the
genome-wide redistribution of these chromatin
components by chromatin-IP coupled with deep
sequencing (ChIP-seq).
TOR-autophagy spatial coupling compartment,
TASCC
Oncogene induced senescence (OIS) is a
very dynamic process where cells typically
undergo an initial burst of cell proliferation
(‘mitotic phase’), followed by the induction
of pro-senescent factors (‘transition phase’).
Eventually, the senescent phenotype dominates
(‘senescence phase’). During the transition
phase, oncogenic and pro‑senescence activities
work against each other, and senescence usually
prevails in normal cells. How cells can achieve
such a drastic phenotypic remodelling is unclear.
A new area of interest in my group is in another
layer of gene expression control, namely protein
metabolism, during senescence. We reason that
global epigenetic alteration should be coupled
with efficient protein turnover as a part of the
execution of epigenetic ‘blue prints’, in such an
emergent context. Consistent with this idea,
we have identified that autophagy, a bulk protein
Figure 1
The TASCC in Ras-induced
senescence (RIS) IMR90 cells.
Confocal images of indirect
immunofluorescence for mTOR
and p62 (an autophagy marker)
or LAMP2 (a lysosomal protein) in
the cells shown. From Narita et al.,
Science 2011; 332; 966
Growing
mTOR / p62
RIS
mTOR / p62
degradation program, facilitates synthesis of
IL6/8, which are central components of SASP
(Young et al., Genes Dev 2009; 23: 798).
We have extended this study to show
that mTOR and autophagy cooperatively
facilitate SASP through forming a new cellular
compartment, the TOR-autophagy spatial
coupling compartment (TASCC), which
provides a local environment enriched for
amino acids and mRNA translation machinery
(Figure 1) (Narita et al., Science 2011; 332: 966).
Autophagic degradation and mRNA translation
are regulated in opposite directions by mTOR,
the central regulator of protein synthesis.
Using immunofluorescence, we identified
well‑demarcated cytoplasmic compartments,
which are enriched for both autolysosomes (the
end stage of autophagy) and mTOR. The TASCC
invariably localises in the vicinity of the rER-Golgi
apparatus, where lysosomal/membrane and
secretory proteins are synthesised and processed.
Thus, it is conceivable that the concentrated
localisation of mTOR on autolysosomes facilitates
localised mRNA translation, including lysosomal
proteins (thus reinforcing TASCC formation) as
well as SASP/SMS components. Notably, since
mTOR inhibits the initial stage of autophagosome
formation, the compartmentalisation of mTOR
with autolysosomes permits autophagosome
formation outside the TASCC. Therefore, the
TASCC would allow a simultaneous activation of
anabolic (by mTOR) and catabolic (by autophagy)
processes.
It has recently been shown that mTOR can be
recruited to the lysosomal surface in response
to amino acids, in a Rag GTPase‑dependent
RIS
mTOR / LAMP2
manner, to become activated by Rheb.
Consistent with this, we showed that
autolysosome-derived amino acids are required
for mTOR recruitment to the TASCC. In
addition, a dominant negative mutant of RagB
inhibited mTOR recruitment to the TASCC,
resulting in decreased synthesis of IL6 and IL8
during RIS, indicating a functional relevance of
the TASCC.
Identification of senescence-associated p53
function
A tumour suppressive transcription factor,
p53, plays a critical role in many stress
responsive phenotypes, including DNA damage
checkpoints, apoptosis, and senescence.
Although ample data have supported a
role for p53 in senescence, the precise
mechanism is not clear. To address this issue,
we are currently using HDFs, where we can
induce different phenotypes depending on
environmental stimuli or other conditions. By
adding either retroviral- or lentiviral-mediated
stable RNAi to HDFs, we are comparing
the impact of p53 knockdown on the gene
expression profile in each condition, which
represents a phenotype specific p53 function.
We have finished the array experiments, and
are now attempting to build a comprehensive
picture of p53’s functions. So far, in a
primary analysis, we have identified several
genes whose products are up-regulated in a
p53‑dependent manner during senescence,
but not in the other stress responsive contexts
(e.g. apoptosis). We are currently undertaking
functional verification of several candidate
genes.
Publications listed on page 77
Research Groups | 37
Prostate Research
www.cambridgecancer.org.uk/davidneal
We are a translational research group with a focus on
castration independent prostate cancer, and have strong clinical
and surgical links.
Group Leader
David Neal
Associate Scientist
Lee Fryer
Bioinformatician
Gökmen Altay (with F Markowetz)
Clinical Fellows
Naomi Sharma†
Karan Wadhwa
Clinical Lecturer
Greg Shaw
Clinician Scientist
Maxine Tran
Graduate Students
Ajoeb Baridi (with J Stingl)
Hélène Bon
Joana Borlido
Samantha Cheung †
Satoshi Hori*
Sarah Jurmeister *
Roheet Rao
Jessica Robinson (with J Carroll)
Ben Thomas*
Postdoctoral Scientists
Mohammad Asim
Antonio Ramos-Montoya
Helen Ross-Adams
Vincent Zecchini
Principal Scientific Officer
Hayley Whitaker
Research Assistant
Jonathan Kay *
Scientific Officers
Helen Scott
Senior Clinical Fellow
Lui Shiong
*
joined in 2011
†
left in 2011
Our clinical practice informs and underpins the
research. We have the largest NHS practice
in robotic prostatectomy: biorepositories from
this, and from ProtecT (the largest ever surgical
randomised controlled trial in prostate cancer),
have led to important collaborative research
with Doug Easton at the Strangeways Research
Laboratory and Ros Eeles at the Institute of
Cancer Research (ICR) (Al Olama et al., Nat
Genet 2009; 41: 1058; Eeles et al., Nat Genet
2009; 41: 1116). Further papers have been
published over the past year (Stacey et al., Nat
Genet 2011; 43: 1098; Schumacher et al., Hum
Mol Genet 2011; 20: 3867; Pashayan et al., Br J
Cancer 2011; 104: 1656; Kote-Jarai et al., Hum
Genet 2011a; 129: 687; Kote-Jarai et al., Nat
Genet 2011b; 43: 785). Our application to carry
out in-depth next generation sequencing of
prostate cancers has been funded by Cancer
Research UK.
We have been joined by Lee Fryer, Mohammad
Asim and Gökmen Altay, and Stephen
Connolly has joined the clinical academic
department to establish joint research in
renal cancer with Tim Eisen (Department of
Oncology).
Castration-independent prostate cancer
Androgen receptor (AR) signalling is
maintained in most men with castrationindependent prostate cancer and new
management and therapeutic approaches
are needed. Our goals are to identify and
characterise markers that better predict
progression, and to identify signalling pathways
that lead to more effective treatments. AR
remains the primary target for treatment
and the rationale remains strong for better
targeting of this pathway and to uncover
biomarkers. We are aiming to work with
human tissue wherever possible (Figure 1),
but our portfolio now includes pre-clinical
in vivo models, which will give us better
information on how individual genes function
throughout tumour development. Examples
38 | Cambridge Research Institute Scientific Report 2011
include p53/pRb or PTEN prostate specific
knockouts, which express the luciferase gene in
tumour cells and makes them traceable though
bioluminescence imaging (BLI).
Main discoveries
Our paper on ChIP sequencing for the
androgen receptor coupled with ChIP for Pol
II has been published (Massie et al., EMBO J
2011; 30: 2719). This research has led to a
new understanding about how the AR binds
to the genome and discoveries about how this
influences major metabolic signalling pathways.
It is now leading to studies on inhibition
of metabolism in man, which are being
underpinned by metabolomic and biological
endpoints (Barrett et al., Magn Reson Med
2011; Epub 29 Aug; McLean et al., Magn Reson
Med 2011; 64: 914). We have two papers
under review at present: one is on studies of
ChIP sequencing in human prostate cancer,
and the other on the role of beta-arrestin in
prostate cancer.
1. ChIP sequencing studies (Massie et al.,
EMBO J 2011; 30: 2719)
We have comprehensively mapped AR binding
sites in two models of prostate cancer using
ChIP-seq, and mapped transcriptionally active
targets using ChIP-seq for phosphorylated
RNA polymerase II, combined with expression
profiling. This approach identified thousands
of novel targets, defined distinct characteristics
of transcriptionally active AR binding sites and
identified signalling pathways directly regulated
by the AR. Amongst these, we identified
calcium/calmodulin kinase kinase 2 (CAMKK2)
as over-expressed in castrate resistant prostate
cancer and as being functionally important for
proliferation. Our data provide new direct
links between the AR and signalling pathways
and offer the potential for novel therapeutic
interventions. We are now expanding these
studies into human material and have discovered
several novel binding sites that appear to be
functional.
Figure 1
We have developed a novel
sampling method to provide fresh
prostate tissue for use in research.
Following radical prostatectomy the
prostate is sliced in a way that does
not compromise patient diagnosis.
Cores of tissue are taken throughout
the fresh tissue slice and snap
frozen. These cores are ‘mapped’ to
allow easy identification of tumour
containing cores and benign controls
for many of our studies.
Visiting Workers
Nejla Altay *†
Marie Corcoran
Rhian Holvey
Ajay Joseph†
Naveen Kachroo
Alastair Lamb
Charlie Massie
Ian Mills
Tania Murphy †
2. Studies on MSMB (PSP94)
Our recent collaborative genome-wide
association studies have shown an association of
a SNP two base pairs upstream of the 5′ UTR
of the microseminoprotein-beta (MSMB) gene
with an increased risk of developing prostate
cancer (Eeles et al., Nat Genet 2009; 41: 1116).
MSMB expression is high in normal and benign
prostate tissue and lowered or lost in prostate
cancer, suggesting that it might be a useful tissue
biomarker for prostate cancer diagnosis. We
have developed an ELISA, which is now being
tested on over 1,500 men (Whitaker et al.,
PLoS ONE 2010; 5: e13363) to determine the
sensitivity and specificity of this approach. We
have also published two other papers on novel
biomarkers (Morgan et al., Clin Cancer Res 2011;
17: 1090; Gudmundsson et al., Sci Transl Med
2011; 2: 62ra92).
3. Studies on HES6
We have now completed our work on HES6,
which is a transcription co-factor best known
for its role in fate decisions of certain stem
cell lineages. Its expression is increased by
c-Myc and the AR, and this creates an altered
transcriptional environment where prostate
cancer cell division and growth is maintained in
the presence of an active AR but in the absence
of ligand binding by dihydrotestosterone
(DHT). We have shown that Hes6 is able, in
isolation, to drive cell growth in an androgen
deprived/castrate setting, and that this
maintained proliferation occurs in the context
of a transcriptionally active AR. We have also
shown that cell cycle and metabolic networks
are activated including up-regulation of E2F
family members, CDC2, UBE2C, CDC20,
Aurora kinases, PLK1, Cyclins, AMACR, GDF15
and LDHA. We have shown by ChIPseq the
cooperation between Hes6, E2F1 and the AR to
maintain G1/S transition and cell proliferation.
4. Studies on beta-arrestin1 (ARRB1: Borlido
et al., Traffic 2009; 10: 1209)
ARRB1 plays a role in cancer progression
and some tumours show elevated levels
in the nucleus where it may regulate gene
expression via epigenetic mechanisms. We
aim to determine the potential role played by
ARRB1 in prostate cancer and to identify novel
biomarkers and therapeutic targets. Prostate
cancer displays elevated levels of ARRB1 that
correlate with stage and aggressiveness, it is also
present in the nucleus in high-grade cancer. We
have identified several genes whose expression
is differentially regulated by ARRB1 and they
are involved in processes such as the cell cycle,
cell motility and metabolism. Using ChIP
sequencing, we have identified several binding
sites for endogenous ARRB1, which reside
mainly in enhancers or proximal promoters and
include genes involved in the unfolded protein
response and autophagy. A paper has been
submitted.
Publications listed on page 77
Research Groups | 39
Regulatory Systems Biology
www.cambridgecancer.org.uk/duncanodom
Group Leader
Duncan Odom
Bioinformatician
Margus Lukk
Graduate Students
Sarah Leigh-Brown†
Caryn Ross-Innes† (with J Carroll)
Dominic Schmidt † (with J Carroll)
Bianca Schmitt
Michelle Ward
Postdoctoral Scientists
Gordon Brown
Claudia Kutter
Klara Stefflova
Diego Villar Lozano*
Michael Wilson
Principal Scientific Officer
Sarah Aldridge*
Scientific Officer
Stephen Watt
Visiting Workers
Benoit Ballester
Paul Flicek
Michaela Frye
Angela Goncalves*
Aileen Marshall
Elisabete Nascimento†
*
joined in 2011
†
left in 2011
The functional elements that control gene expression that are
used to create a diversity of tissues remain poorly understood.
The ultimate aim of classical genetics and modern genomics
is to understand the molecular details of how the genome is
deployed transcriptionally to create a diversity of tissues and
species. This understanding has profound importance for
cancer research, as a major hallmark of tumour progression is
the occurrence of new genetic mutations and their resulting
perturbation of gene expression programs. Using liver and
liver cancer as model systems, we research the regulation and
evolution of all forms of transcription that occur in mammals.
The control and evolution of cellular gene
expression
The proteins that control DNA, known
as transcription factors, bind to it in a
combinatorial manner in yeast and bacteria, and
my early work showed that this combinatorial
binding occurs in mammalian tissues as
well. Master regulators in primary human
hepatocytes form a highly interconnected core
circuitry that frequently bind promoter regions
in clusters, particularly at highly regulated and
transcribed genes (Odom et al., Mol Syst Biol
2006; 2: 2006.0017). More surprisingly, we have
recently found that transcriptional regulation
can vary much more rapidly and widely than
previously appreciated among homologous
tissues from many mammals (Schmidt et al.,
Science 2010; 328: 1036; Odom et al., Nat
Genet 2007; 39: 730). The experiments in our
laboratory allowed the identification of specific
genetic architectures that appear to preserve
a small handful of transcription factor binding
events across large evolutionary timescales
(>300 million years) (Schmidt et al., Science
2010; 328: 1036).
In asking why rapid variation occurs among
most transcription factor binding events, we
realised that a number of causative factors
could contribute. These possible causes may
be the result of variability of genetic sequences,
the types and number of marks left in the
histone proteins that package DNA (commonly
thought of as an epigenetic code), or even diet
or environmental differences between different
40 | Cambridge Research Institute Scientific Report 2011
species. In order to isolate a single one of these
variables, we used a previously created mouse
model of Down’s syndrome that carries a
virtually complete copy of a human chromosome
(O’Doherty et al., Science 2005; 309: 2033).
By exploiting this aneuploid mouse strain, a
unique and powerful genetic tool designed for
an entirely different purpose, my laboratory
was able to determine that genetic sequence
dominates all others in directing transcription
(Wilson et al., Science 2008; 322: 434).
The origin, regulation, and evolution of
noncoding RNA transcription
We have been using similar comparative
functional genomics approaches to look at the
regions of the genome that are transcribed, but
which do not code for proteins. These regions
are known as non-coding RNAs, and range
from well-characterised species like tRNAs
and rRNAs to newer categories of regulatory
nucleic acids like microRNAs, piRNAs, and
endogenously expressed RNAi. We recently
published results describing previously
unseen functional conservation in tRNA gene
transcription driven by RNA polymerase III, that
only becomes apparent after analysis of data
from multiple mammalian species (Figure 1).
The complex interplay of CTCF, cohesion, and
repetitive sequences in the genome
The CTCF protein is a genomic anchor that
appears to have roles in regulating mitosis and
meiosis, and in insulating chromatin and gene
expression across the genome. Many of these
RNA polymerase III ChIP-seq
Figure 1
RNA polymerase III regulation of
tRNA loci in six mammals.
110
12
5 kb
Mmu
110
95
Cfa
8 0
7
Lineage divergence in MY
_
382
44
6
4
23
0
3
1240
2
32
0
116
220
6
2
168
1
1
1
0
0
80
18
12
17 3
5
Rno
9
7
14
25
93
Hsa
Mml
180
Thr
A
AmnSINE
expand
Few tRNAs are bound by pol III
in all placental mammals
Pro Val Leu
ur-Mammal
ur-Placental
Hsap
MamRep
expand
Mmul
B2 expand
ur-Rodent
Mmus
B2 expand
Rnor
B2 expand
150
100
Lys expand
Cfam
MIR expand
Mdom
50
0
Time (million years)
CC
AT
G
T
A
C
T
C
CT
A
AA
T
Zn
C
C
T
T
T
TGC
C
GT
G
C
A
G
3′
Mmul
Mmus
7
8
Zn
Zn
10
Zn
Zn
2
Rnor
11
Zn
Zn
1
A
CT
T T CCG
C
9
Zn
Zn
G
G
A GCA A T A
AA
A
G
C
6
5
4
3
G
G
C
G
CA
T
34
GT
30
AT
chr9: 20,356,303 - 20,356,647
Hsap
C CT TG
GC
G
25
5′
T
A
20
A
T
G
10
B2 transposon
sequence
T
CT A
G
C
TG
C
5
0
15
2
bits
B
1
Figure 2
CTCF binding evolution across
mammals reveals new mechanisms
of genome evolution, driven by
repetitive elements.
982
Zn
Zn
CTCF
Cfam
Mdom
Mammal
Conservation
CTCF motif
disrupted in Mouse and Rat
AmnSINE1
functions are mediated by the cohesin complex in
mammalian cells. We have discovered how the
cohesin complex can co-regulate gene expression
with tissue-specific transcription factors in the
absence of its canonical partner CTCF (Schmidt
et al., Genome Res 2010; 20: 578). More recently,
we have made the surprising discovery that most
lineage-specific CTCF binding is not born in the
same way as other, tissue-specific transcription
factors, but appears in the genome via carriage
within repetitive elements that are active in a
species-specific manner in mammals (Schmidt et
al., Cell 2012; 148: 335)(Figure 2). Collectively we
found that these newborn CTCF binding events
are as functionally active as ancient ones found
in six or more mammals, and that these ancient
binding events show fossilized remains of the
prior repeat expansions that gave birth to them.
Publications listed on page 79
Research Groups | 41
Polygenic Predisposition to Breast Cancer
www.cambridgecancer.org.uk/bruceponder
Our research focusses on inherited susceptibility to breast
and other common cancers. Our overall aims are to identify
the genes involved and their mechanisms, so as (1) to define
high‑risk groups within the population, and (2) ultimately, to
devise strategies for prevention based on the mechanisms of
risk.
Group Leader
Bruce Ponder
Associate Scientist
Kerstin Meyer
Graduate Students
Michael Fletcher
Esther Musgrave-Brown (with
C Lichtenstein, Population
Genetic Technologies)
Postdoctoral Scientists
Mauro Castro (with F Markowetz)
Ana-Teresa Maia†
Scientific Officers
Martin O’Reilly
Radhika Prathalingam
Visiting Worker
Ramsay Bowden*†
To date our work has been following up the
results of the genome-wide association studies
in breast cancer that we and our colleagues
initiated. The chance of an individual developing
breast cancer is roughly two-fold greater if that
individual has a close relative with breast cancer.
Twin studies indicate that this risk is largely
genetic. The genes that confer this risk have
been sought either by genetic linkage mapping
in multiple-case families, or by genome-wide
association studies (GWAS). The former have
identified rare but higher risk alleles such as
those of BRCA1 and BRCA2, while GWAS have
identified common variants that each carry only a
small risk of cancer. BRCA1 and 2 explain about
15–20% of the estimated total genetic risk of
breast cancer, and loci identified through GWAS
a further 10%.
One question is how to find the genes that
account for the ‘missing’ 70% or so of heritability.
Larger and more powerful GWAS will find
some, while genome resequencing will identify an
unknown contribution from rare genetic variants.
Pending these studies, we are exploring other
approaches in breast and in lung cancer.
*
joined in 2011
†
left in 2011
In breast cancer, Ana-Teresa Maia is making a
catalogue of genes in which there are common
variants that cause different levels of expression
of the two alleles in a heterozygous individual.
Since many of the genetic variants so far identified
in GWAS studies in general appear to have an
effect through altered gene regulation, the subset
of genes that show differential allelic expression
(DAE) should be enriched for genes involved in
susceptibility. If correct, this information would
allow prioritisation of genes for further study
from the very large numbers of loci that show
borderline levels of significance in existing GWAS.
We are now testing this hypothesis. We also
42 | Cambridge Research Institute Scientific Report 2011
have preliminary data to show that differences in
expression level of the remaining wild-type allele
influence the penetrance of BRCA2 mutations in
heterozygous carriers.
The common genetic variants identified through
GWAS each have very small effects. They can
be thought of as causing small perturbations of
regulatory gene networks within the cell – the
combined effect of many variants produces a
greater perturbation that leads to disease. A
regulatory variant in the fibroblast growth
factor receptor 2 (FGFR2) gene is the common
variant with the greatest effect on breast cancer
susceptibility. We are using a systems biology
approach to understand the function of this
predisposing gene. We have treated the wellstudied oestrogen dependent cell line MCF-7
with oestrogen and FGF10, an activator of the
FGFR signalling pathway. Microarray analysis
has identified downstream target genes that are
differentially regulated. We are now building
a network of genes that are co-ordinately upor down-regulated after FGFR and oestrogen
receptor signalling in a time-dependent manner.
Highly connected genes, forming a hub in the
network, are likely to be master regulators and
might themselves act as predisposing genes.
These hubs may also be promising therapeutic
targets that could be modulated in order to
correct a de-regulated network. As part of
this analysis Mauro Castro has developed a
software package, RedeR, that allows both visual
representation and analysis of nested network
structures (manuscript submitted), harnessing
the advantages of the R and Bioconductor
bioinformatics tools (Figure 1).
The list of loci associated with breast cancer has
been growing, and over 20 are now published.
We have focussed our functional analysis on




























  














 








 






















 











 








 
























 





 














  






 














 











 














 


 


 


 

 











   












 









 






 

























 









  













 






 










































 











 





















Figure 1
Protein-protein interaction subnetworks at different times after
oestrogen stimulation in MCF-7 cells
are shown. Node colouring depicts
differential expression as log2 foldchange (logFC).















genomic regions that confer risk to multiple
different types of cancer, such as regions on
chromosome 8q24 and 11q13. We have
proposed a mechanism for predisposition at the
8q24 locus: the risk allele changes binding by the
transcription factor YYI, which alters expression
of the oncogene PVT1 (Meyer et al., PLoS
Genetics 2011; 7: e1002165). In collaboration with
the Strangeways Research Laboratory and the
Queensland Institute for Medical Research we
have also investigated a locus for susceptibility to
breast cancer at 11q13. By a combination of fine
mapping, analysis of chromosome architecture,
transcription assays and biochemical analysis of
protein-DNA interactions, we have identified a
SNP that abolishes binding by the transcription
factor ELK4 and affects activity of the enhancer
element in which the SNP lies. Chromatin
conformation capture suggests that the target
gene is cyclin D1.
question about whether there are individual
differences in the response of airway cells to
cigarette smoke injury that correlate with lung
cancer. The read out of injury response will be
patterns of mRNA and miRNA expression. If
differences are found these may provide useful
markers of risk; if the patterns of expression
that are associated with risk can be resolved
into networks that indicate mechanism, this may
provide possible targets for mechanism-based
prevention.
Publications listed on page 79
In lung cancer, less than 5% of the estimated
genetic variance of risk is explained by the DNA
sequence variants so far identified. We are
taking two approaches to search for this ‘missing
heritability’. In one approach we will attempt
to use novel assays of DNA repair capacity to
ask whether differences in DNA repair affect
individual risk of lung cancer in smokers. In the
second approach we will ask a more open-ended
Research Groups | 43
Molecular and Computational Diagnostics
www.cambridgecancer.org.uk/nitzanrosenfeld
Advances in cancer genomics and molecular technologies are
opening new possibilities for diagnostics. We are applying
these to develop diagnostic tools that use cell-free circulating
nucleic acids.
Group Leader
Nitzan Rosenfeld
Graduate Student
Muhammed Murtaza*
Postdoctoral Scientists
Tim Forshew
Dana Tsui
Senior Scientific Officer
Davina Gale
Rational clinical decisions on the management
and treatment of cancer rely on accurate
diagnostic information. Molecular analysis
of tumour samples has been used to predict
prognosis or response to treatment, but should
be complemented by non-invasive methods for
monitoring disease progression or dynamics.
Circulating DNA in plasma and serum include
tumour-specific sequences that are a promising
source of diagnostic information.
The mechanisms through which tumour DNA
reaches blood circulation are unclear, although
fragmentation patterns of DNA in the plasma
of cancer patients suggest it may originate from
cell death. Overall levels of circulating DNA
are higher in cancer patients compared with
healthy controls, but these differences are not
consistent enough for robust diagnostic tools.
The maturation of genomic technologies allows
circulating tumour-specific DNA to be used as
personalised biomarkers (Figure 1).
Circulating tumour DNA (ctDNA) can be
measured by tying together genomic and
molecular techniques. First, tumour-specific
somatic alterations must be identified on a
case-by-case basis. Second, sequence-specific
molecular assays must be designed that can
precisely detect and measure tumour-specific
sequences in the background of circulating
genomic DNA. Finally, these assays must be
applied to body fluid samples such as blood
plasma that have been carefully collected and
processed to extract circulating DNA.
*
joined in 2011
†
left in 2011
Circulating tumour DNA may be useful for
identifying the presence of cancer mutations,
for detecting systemic or residual tumour
burden, or for non-invasive monitoring of
tumour changes. Preliminary studies suggest
that ctDNA compares favourably to imaging
or to currently used protein markers. Our
goal is to translate this potential into diagnostic
44 | Cambridge Research Institute Scientific Report 2011
applications, by integrating new quantification
methods and computational insights with clinical
research.
Measurement and noise in molecular biology
Quantitative measurements in molecular biology
are challenging; objects of study are highly
sensitive biochemical systems and repeated
sampling is limited since living organisms are
highly variable and dynamic. Reliability depends
on our ability to take into account biological
variation, measurement noise and biases.
In earlier studies (at the Weizmann Institute of
Science), time-lapse microscopy and fluorescent
reporter fusions were used to study gene
regulation circuits. These studies demonstrate
one approach to overcoming biological variation,
by performing measurements in individual living
cells.
Medical diagnostics poses different challenges.
Clinical samples are often limited and
heterogeneous, and can vary in collection
conditions or contain a mixture of tumour
and other material. Molecular quantification
methods introduce additional noise and bias.
We need to understand these effects and
consider their impact on the design of diagnostic
tests.
We are studying collection and processing
protocols for peripheral blood samples, to
optimise these for measurement of ctDNA
and adapt them for simplified clinical use.
We quantify DNA using parallel or ‘digital’
PCR, arguably the most accurate method
for quantification of nucleic acid sequences.
Template molecules are distributed into multiple
independent reactions, reducing background
interference. Quantification is obtained through
counting of positive amplifications, and does not
rely on calibration standards or curves.
Figure 1
Workflow for studies on circulating
tumour-specific DNA. DNA
obtained from a patient’s tumour
or biopsy sample is used to identify
tumour-specific genomic alterations.
Assays are designed to specifically
measure these tumour-specific
DNA sequences. Assays are
validated using tumour DNA as
positive control and DNA from other
subjects (and normal) as negative
controls. The assays are used to
measure ctDNA levels in blood
samples from the same patient.
These data are compared to
clinical information to study ctDNA
dynamics and diagnostic potential.
!"#$%&'
(")%#*'
+,-'.&/0'120/2&'3"0(4%'
56#/&0"47'
120/2&<3(%$9=$'
+,-'"4*%&")/#3'
120/2&<3(%$9=$'
0/4%$24"&'"33"B'
!49#9$"4'
./44/?<2('
8%&9"4':4//;'
3"0(4%3'
,%?'
0/4%$24"&'
;9"@#/3)$'
*//43A'
>2"#)=$")/#'
/.'$*+,-'
Diagnostic algorithms
A major challenge in designing diagnostic
tests is in defining categories that are clinically
informative and can also be robustly identified.
Tumours can be classified, for example, as
positive or negative for hormone receptors,
indicating the suitability of hormonal treatment.
To be effective, diagnostic algorithms need to
take into account both measurement limitations
and clinical considerations.
Non-invasive diagnostics using ctDNA
The study of ctDNA requires carefully collected
samples from clinical studies that include tumour
or biopsy material and matched collections
of blood samples. It is possible to accurately
measure ctDNA in these samples using the
methods that we are developing and utilising.
These data must then be compared to clinical
follow-up data to identify associations and
potential diagnostic roles of ctDNA (Figure 1).
In previous projects (at Rosetta Genomics Ltd.),
microRNA expression levels were used to
classify tumour histological types and sites
of origin. Classification was based on strong
biomarkers and intuitive, ‘logical’ decision
criteria. The robust design of these algorithms
enabled their rapid translation into clinical
tests. This practical approach to molecular
classification is likely to be effective in translating
other types of diagnostic assays into clinical
practice.
We work in close collaboration with clinical
groups to study the dynamics and utility of
ctDNA in solid malignancies, with the aim
of developing findings into robust diagnostic
assays. In 2011, we developed novel diagnostic
tools based on targeted deep sequencing. In
collaboration with the Brenton and Caldas
groups at the CRI, we demonstrated the
effectiveness of ctDNA as biomarkers for
advanced ovarian and breast cancers, as well as
the potential of this approach for non-invasive
monitoring of clonal changes in tumours.
We use state of the art genomic tools to
identify somatic changes in the DNA from a
tumour or biopsy sample, and design tumourspecific molecular assays. The complex analysis
of tumour material shifts the burden of proof
and makes the measurement of ctDNA in blood
samples direct and unequivocal. We believe
that these personalised biomarkers will prove to
be highly informative and clinically effective.
Research Groups | 45
Mammary Stem Cell Biology
www.cambridgecancer.org.uk/johnstingl
Group Leader
John Stingl
Graduate Students
Ajoeb Baridi (with D Neal)
Rajshekhar Giraddi
Michael Prater
Siru Virtanen (with J Brenton)
Postdoctoral Scientists
Alasdair Russell
Amel Saadi
Mona Shehata
Visiting Students
John Chen*†
Katerina Georgopoulou*
*
joined in 2011
†
left in 2011
My laboratory is interested in identifying and characterising
the cells that make up the normal mammary epithelium, and
how these cells relate to those present in different types
of human breast tumours. We are particularly interested
in studying mammary stem and progenitor cells since we
hypothesise that these cells are the initial targets for malignant
transformation. The laboratory also has an interest in
characterising the cellular hierarchies present in human serous
ovarian tumours and in normal and malignant human prostate
tumours.
My laboratory currently has five main research
themes:
1. Characterisation of normal mammary
epithelial stem and progenitor cells
Mammary stem and progenitor cells are
perceived to be the cell of origin of breast
tumours since only these cells have the
replicative capacity that allow the multiple
mutations required for tumour progression
to accumulate. My previous research
demonstrated that functionally distinct
mammary cells can be purified and detected
via the use of flow cytometry and functional
assays (Figure 1). We have recently identified
two novel types of progenitor cells within the
human mammary epithelium and are currently
determining their properties and how they
relate to breast tumours.
2. Determining the cell of origin in breast
cancer
Breast cancer is a heterogeneous disease with
approximately five molecular subtypes and 18
histological subtypes identified. Our laboratory
is interested in elucidating the mechanisms
that account for this heterogeneity. One
possible mechanism is that different types of
breast cancers initiate in, and are propagated
by, different types of mammary cells. To test
this directly, we are conducting experiments in
which we are introducing common oncogenic
mutations into different cellular backgrounds to
see if events preferentially exert their effects
in specific types of cells. In addition, we are
46 | Cambridge Research Institute Scientific Report 2011
particularly interested in finding out if loss of
common tumour suppressor genes can impart
some properties of stem cells to committed
progenitor and differentiated cells.
3. Identification of the molecular mechanisms
that regulate stem cell self-renewal
Self-renewal is perceived to be a defining
property of stem cells. Cellular pathways that
regulate stem cell self-renewal are considered
to be good targets for therapeutic intervention
since tumours should eventually exhaust their
proliferative capacity in the absence of these
pathways. We have identified a number of
candidate genes that are differentially expressed
between stem cells that are undergoing
symmetric cell division vs. asymmetric cell
division, and we are currently evaluating the
influence of these genes on mammary stem cell
function.
4. Characterisation of human ovarian cancer
stem cells (collaborative project with James
Brenton, CRI)
Serous ovarian cancer is an aggressive disease
that initially responds to chemotherapy, but
approximately 70% of patients will relapse
and become resistant to therapy. It is our
hypothesis that this resistance is mediated by
the emergence of a sub-population of ovarian
cancer stem cells. We are currently evaluating
the proliferative potential of phenotypically
distinct subsets of ovarian tumour cells in
order to identify the putative cancer stem cells.
Future research includes tracking experiments
104
LP
NCL
13.7
103
EpCAM
Figure 1
Flow cytometry dot plot
demonstrating the distribution of
epithelial cell adhesion molecule
(EpCAM) and alpha 6 integrin
(CD49f ) among freshly dissociated
human mammary epithelial cells.
The differentiated luminal (NCL),
luminal progenitor (LP) and basal
cell populations are indicated. The
LP population generates colonies
of pure luminal cells, whereas the
basal cell population is enriched
in bipotent progenitors, which
generate mixed lineage colonies in
vitro. Re-plating of these mixed
colonies demonstrates that they
are precursors to myoepithelialrestricted progenitors. Scale bars
are 1mm.
102
7.82
101
Basal
100
100
101
102
103
104
CD49f
to follow the fate of individual clones during
chemotherapy and gene expression profiling of
these cells.
5. Characterisation of the normal and
malignant prostate epithelial cell hierarchy
(collaborative project with David Neal, CRI)
We are applying our expertise in the
characterisation of the mammary epithelial
cell hierarchy to the human prostate epithelial
cell hierarchy. We are particularly interested
in characterising progenitor cells within the
human prostate and their developmental
relationships.
Publications listed on page 80
Research Groups | 47
Computational Biology and Statistics
www.cambridgecancer.org.uk/simontavare
Our work has continued its focus on three main areas:
Statistical methods for the analysis of next‑generation
sequencing data, evolutionary approaches to cancer and
methods for the analysis of genomics data.
Group Leader
Simon Tavaré
Associate Scientist
Andrew Lynch
Bioinformatician
Nick Shannon*
Graduate Students
Daniel Andrews
Jonathan Cairns
Angelina Chattopadhyay Mukherjee*
Michael Smith
Andrea Sottoriva
Alex Tunnicliffe*
Julie Woolford†
Postdoctoral Scientists
Benilton Carvalho
Edward Morrissey *
Shamith Samarajiwa
Anestis Touloumis* (with J Marioni,
EBI)
Ernest Turro*
Senior Scientific Officer
Inmaculada Spiteri
Visiting Student
Rishi Kanungo*†
Visiting Workers
Audrey Qiuyan Fu†
John Marioni*
Nick Marko*
*
joined in 2011
†
left in 2011
Illumina BeadArray technologies continue to
be an important tool in cancer studies (such
as METABRIC) and we, in collaboration with
Mark Dunning and Matt Eldridge (Bioinformatics
Core), continue to update and support the
beadarray Bioconductor package in order to
facilitate transparent and flexible statistical
analyses of full bead-level data. In addition, we
continue to provide independent annotations of
the expression BeadArray platforms.
A major new focus in the group is the
International Cancer Genome Consortium
project on oesophageal adenocarcinoma. This
is led by Dr Rebecca Fitzgerald (Hutchison/
MRC Research Centre), in collaboration with
the Bioinformatics Core. This project is funded
by Cancer Research UK, and has just completed
its pilot phase of sequencing some 30 tumournormal pairs. The full project will sequence 500
pairs, and should provide interesting and medically
relevant information about the aberrations that
occur in the genomes of this tumour. The pilot
study has already highlighted the complexities
of large-scale sequencing projects, particularly
with respect to the identification of structural
variants, SNPs and SNVs, and the computational
infrastructure required to support such projects.
We have continued our research in the area of
evolutionary methods in cancer biology, focussing
in particular on spatial stochastic models for the
evolution of colorectal cancer. Such models
can be used, inter alia, to study the cancer
stem cell (CSC) hypothesis by comparing the
dynamics of molecular markers in a CSC-driven
tumour with that of a non-hierarchical growth
model (see Figure 1). This approach allows
us to estimate the CSC fraction in a tumour,
and to predict the effects of treatment. We
have used high-throughput 454 sequencing to
generate large amounts of data on heterogeneity
in colon cancer. We have also continued
our development of approximate Bayesian
48 | Cambridge Research Institute Scientific Report 2011
computation (ABC) for inference in agent-based
models such as those used for colon cancer.
We have a number of other ongoing
collaborations within the CRI, in particular with
the Narita, Neal and Winton labs. We have
also been collaborating with Dr Colin Watts’
lab in Clinical Neurosciences in a study of
intra‑tumour heterogeneity in glioblastoma.
We have continued our collaboration with the
Caldas laboratory on the statistical analysis
of the METABRIC project that has assayed
germline and somatic copy number variants,
and their impact on expression variation, in
some 2,000 breast tumours using high-density
microarrays.
The lab has several new recruits this year.
Drs Edward Morrissey (Warwick) and Ernest
Turro (Imperial) completed their PhDs in 2011
and joined us as postdocs. Ed is working with
the Winton lab, and Ernest’s expertise is in the
analysis of RNA-seq experiments. Dr Anestis
Touloumis completed his Statistics PhD at the
University of Florida and has begun a postdoc,
shared with Dr John Marioni at the EBI.
Dr Nick Shannon joined the group as part of
the International Cancer Genome Consortium
project and Dr Nick Marko is a visiting fellow
supported by an American Association
of Neurological Surgeons Van Wagenen
Fellowship. Alex Tunnicliffe and Angelina
Mukherjee began their PhDs in October.
Dr Audrey Fu, a statistician who worked
closely with Professor Sarah Bray (PDN) and
Dr Steve Russell (Genetics) on the downstream
transcriptional effects of Notch in Drosophila,
completed her postdoc, and is now in Matthew
Stephens’ group in the Statistics Department at
the University of Chicago.
Publications listed on page 80
Figure 1
3D simulation of a cellular Potts
model of a tumor, showing
heterogeneity and spatial structure.
Regions of different colour
correspond to different mutations
present in the tumor.
Research Groups | 49
Tumour Modelling and Experimental Medicine
www.cambridgecancer.org.uk/davidtuveson
Our laboratory conducts basic and applied research
in pancreatic ductal adenocarcinoma (PDA) to aid the
development of efficacious therapies and accurate diagnostics
for this highly fatal disease.
Group Leader
David Tuveson
Clinical Fellows
Natalie Cook †
Shivan Sivakumar
Clinician Scientist
Athena Matakidou
Graduate Students
Meredith Caldwell†
Derek Chan*
Sarah Eastmond† (with D Jodrell)
Timothy Humpton
Michael Jacobetz †
Postdoctoral Scientists
David Allard†
Danielle Engle*
Christine Feig
Kristopher Frese
Siong-Seng Liau
Albrecht Neesse*
Pedro Perez-Mancera
Daniele Perna
Hervé Tiriac*
Principal Scientific Officer
Frances Connor
Scientific Officers
Nicola Brindle†
Aarthi Gopinathan
Lisa Young
Senior Scientific Officers
Judit España*
Paul Mackin
Visiting Workers
Anne Kultti*†
Carla Martins
Tomoaki Nakagawa*
Ashley Nicholls†
*
joined in 2011
†
left in 2011
We have produced genetically engineered
mouse models (GEMMs) that accurately mimic
human PDA, and used them to characterise
the fundamental molecular, cellular and
pathophysiological principles of PDA. These
GEMMs have enabled us to establish a
preclinical therapeutics effort for the systematic
evaluation of traditional and experimental
agents. Specifically, we have determined that
KrasG12D induces the Nrf2 transcription factor
to promote tumour initiation. Furthermore,
we found that Craf and Cathepsin B play
important roles in KrasG12D driven epithelial
cell proliferation and transformation, providing
direct targets to pursue for therapeutic
strategies. Pre-clinically, we have determined
that the deficient vasculature of PDA tumours
can be destabilised by blocking the enzyme
gamma secretase, and in combination with
gemcitabine this promotes hypoxic necrosis
and prolongation of survival. We established
a pancreatic cancer clinical research team to
enable the translation of this observation, and
currently have multiple trials open related to our
pre-clinical work.
Kras induces Nrf2
Oncogenic Ras signalling has variably been
associated with proliferation and senescence,
two opposing outcomes that may shape the
tropism of carcinogenesis. Since KrasG12D
expression promoted the proliferation of the
pancreatic ductal lineage in vivo, we investigated
the pathways promoting this increased cellular
fitness. Although the ectopic overexpression
of oncogenic Ras in primary fibroblasts
was previously shown to cause increases in
intracellular reactive oxygen species (ROS)
and cell cycle arrest due to OIS, we showed
that endogenous KrasG12D expression directly
lowers ROS and promotes cellular proliferation,
providing a potential explanation for the
differing observations between prior work
and our own. A proteomic and cell biological
50 | Cambridge Research Institute Scientific Report 2011
approach revealed that KrasG12D induced a
lowering of ROS due to the increased mRNA
and protein expression of the Nrf2 (Nfe2l2)
transcription factor. Nrf2 coordinates the
transcription of many genes including those
primarily responsible for the detoxification
of ROS by modulating the levels of active
glutathione and thioredoxin. Nrf2 normally has
a short protein half-life of less than five minutes
due to its association with the Keap1 repressor
protein. Various cellular stressors that lead to
the modification of the Nrf2 or Keap1 proteins
can prolong the half-life of Nrf2, and somatic
cancer associated mutations in either Keap1 or
Nrf2 that interfere with Keap1 binding to Nrf2
and thereby stabilize Nrf2 have been reported.
We found that Nrf2 half-life was not prolonged
in KrasG12D -expressing cells, and also observed
that mutations in Nrf2 and Keap1 were rare
in human pancreatic cancer. Differences
between ectopic and endogenous oncogenic
Kras expression included the activation of the
ROS generating NADPH oxidase 1 (NOX-1)
by ectopic Kras. Furthermore, ectopic Ras only
transiently induced Nrf2 expression whereas
this was stably sustained by endogenous
KrasG12D.
The MEK inhibitor U0126 was used to show
that the MAP kinase Ras effector pathway was
responsible for Nrf2 mRNA induction and
decreased ROS levels following expression
of KrasG12D. The downstream mediator of
this finding was the AP1 family member Jun:
following KrasG12D expression, Jun protein
levels increase. Indeed, this was supported by
a previous ENCODE study showing that Jun
bound to the human NRF2 promoter.
Nrf2 nullizygous mice demonstrated that
Nrf2 was required for endogenous KrasG12D to
promote proliferation in preneoplastic lung and
pancreatic epithelial cells in vivo. Furthermore,
the genetic silencing of Nrf2 in cell culture or
Figure 1
Oncogenic signalling that leads to
increased Nrf2 gene expression
is an alternative mechanism to
activate Nrf2 and thereby promote
tumourigenesis.
K-Ras
K-RasG12D
B-Raf
B-RafV600E
MAP kinase cascade that are essential for
tumourigenesis, we selectively and conditionally
deleted either Craf or Braf while simultaneously
activating KrasG12D expression in lung epithelial
cells. Despite the prominent role Braf plays
in activating MEK, we found that Craf, but not
Braf, was required for the proliferation and
transformation of lung epithelial cells in response
to KrasG12D.
MEK
Deregulated Myc
ERK
Jun
Myc
Nrf2
tumourigenesis
ROS
GSH/GSSG
Antioxidant
response
in vivo promoted cellular senescence in the
context of KrasG12D expression, suggesting
that KrasG12D expressing cells are dependent
upon Nrf2 during the earliest stages of cellular
transformation such as tumour initiation
reinforcing our prior findings in fibroblasts.
Interestingly, these effects could be mimicked
pharmacologically by suppressing ROS
metabolism with an inhibitor of glutathione
synthesis, buthionine sulfoxime. Also, the loss
of Nrf2 could be compensated for by treating
cells with N-acetyl cysteine, an anti-oxidant
that acts as a glutathione mimetic to restore
the intracellular reducing environment. This
work raises the concern that anti-oxidants
could have tumour promoting properties in
certain contexts, a concept we will pursue in
future studies. Lastly, our findings extended
to an endogenous oncogenic BRAF V619E allele
(human V600E) and the nearly physiological
Rosa26 MycERT/ERT allele, suggesting a mechanism
of general importance in two oncogenic
pathways (Figure 1).
Preclinical and clinical translation
Notch pathway antagonism has been shown to
prevent pancreatic pre-neoplasia progression
in mouse models, but potential benefits in
the setting of an established PDA tumour
have not been established. We found that
gamma secretase inhibitors (GSI) inhibited
intra-tumoural Notch signalling in our mouse
GEMMs. While GSI alone failed to extend
the lifespan of KPC mice, the combination
with the chemotherapeutic gemcitabine
prolonged survival. Combination treatment
killed tumour endothelial cells and synergistically
promoted widespread hypoxic necrosis. These
results indicate that the pauci-vascular nature
of PDA can be exploited as a therapeutic
vulnerability, and the dual targeting of the
tumour endothelium and neoplastic cells
by gamma secretase inhibition constitutes a
rationale for clinical translation. We designed an
investigational Phase 1/2 clinical trial to evaluate
whether these observations would translate
to patients, and have already enrolled several
patients. This trial is directed by Prof. Duncan
Jodrell at the University of Cambridge and the
CRI.
Publications listed on page 81
Pathways governing Kras oncogenesis: Craf
and cathepsin B
Our work with Nrf2 suggested that the MAP
kinase cascade was essential to activate this
detoxification programme and stimulate
proliferation and neoplasia. To further
understand the molecular aspects of the
Research Groups | 51
Keratinocytes in Normal Tissue and in Tumours
www.cambridgecancer.org.uk/fionawatt
Group Leader
Fiona Watt
Clinical Fellows
Sven Quist
Stephen Goldie†
Graduate Students
Esther Arwert †
Carles Escriu
Grace Kaushal
Paweł Schweiger
Christine Weber
Postdoctoral Scientists
Sara Cipolat
Giacomo Donati
Ryan Fiehler
Hironobu Fujiwara
Esther Hoste
Alex Kuznetsov
Ajay Mishra
Klaas Mulder
Ken Natsuga
Alexander Schreiner
Principal Scientific Officers
Simon Broad
Paul Newman
Visiting Worker
Nathan Benaich
*
joined in 2011
†
left in 2011
The epidermis consists of a multilayered epithelium, the
interfollicular epidermis, and associated hair follicles, sweat
glands and sebaceous glands. All of the different lineages within
the epidermis are maintained through proliferation of stem
cells and differentiation of their progeny (Watt and Jensen,
EMBO Mol Med 2009; 1: 260). By investigating how stem cell
renewal and differentiation are controlled in normal tissue, we
hope to identify new approaches to preventing and controlling
tumours of the epidermis and other stratified squamous
epithelia (Watt and Driskell, Phil Trans Roy Soc B 2010; 365:
155).
Stem cell renewal and lineage selection
One of our ongoing interests is in how stem
cell behaviour is regulated by extrinsic signals
from the local microenvironment, or niche.
Two different and complementary approaches
can be taken to investigate this: observing
stem cells in vivo and recreating the niche in
vitro. Stem cell behaviour in vivo is a composite
response to all niche signals, whereas in vitro
it is possible to parse out the response to
individual signals.
For our in vitro studies we have developed, in
collaboration with Wilhelm Huck (Department
of Chemistry, University of Cambridge and
Radboud University, Nijmegen), micropatterned
extracellular matrix (ECM)-coated glass
substrates that selectively capture single
human epidermal stem cells. The substrates
are amenable to microscopic analysis of living
cells, allowing us to perform FRET and image
cytoskeletal dynamics. In addition, we can
perform single cell gene expression profiling
of cells on these substrates (Gautrot et al.,
Biomaterials 2010; 31: 5030; Connelly et al.,
Nat Cell Biol 2010; 12: 711). We found that
when spreading is restricted on small circular
islands, cells exit the stem cell compartment
and differentiate. The state of assembly of
the actin cytoskeleton regulates differentiation
by controlling serum response factor (SRF)
transcriptional activity. Our studies have also
established a role for extrinsic physical cues
in the regulation of chromatin remodelling
52 | Cambridge Research Institute Scientific Report 2011
(Connelly et al., PLoS ONE 2011; 6: e27259).
We are now investigating how stem cells
respond to differences in substrate stiffness
and topology, and whether environmental
responsiveness is altered in cells from squamous
cell carcinomas.
Complementing the in vitro studies, we
have continued to investigate the stem cell
compartment in vivo, using genetically modified
mice. One of the key pathways that regulates
epidermal stem cells is the Wnt pathway. We
have found that activation of β-catenin in
epidermal stem cells causes reprogramming
of the underlying dermis to a neonatal state,
characterised by remodelling of the extracellular
matrix and stimulation of fibroblast proliferation
(Collins et al., Development 2011; 138: 5189). We
are now investigating whether the changes we
have observed are also features of the stroma of
epithelial tumours in which the Wnt pathway is
activated.
While it is well established that stem cell
behaviour is regulated by signals from the
niche, it is only now becoming clear that stem
cells can provide a niche for neighbouring
cells. We have observed this in skin, through
studies of an extracellular matrix protein called
nephronectin (Fujiwara et al., Cell 2011; 144:
577). Nephronectin is a Wnt target gene that
is specifically expressed by stem cells in the
hair follicle bulge. Deposition of nephronectin
stimulates neighbouring mesenchymal cells
Figure 1
Whole mount of mouse skin showing
arrector pili muscles attaching to
hair follicles. Photograph courtesy of
Heather Zecchini (Light Microscopy
core facility).
αSMA/SM22α/DAPI
to differentiate into smooth muscle cells and
determines the anchorage of the arrector
pili muscle to the bulge (Figure 1). It will
be interesting to determine whether other
extracellular matrix proteins deposited by
epidermal stem cells regulate the behaviour of
dermal cell subpopulations.
Stem cells, differentiated cells and tumour
formation
Interactions between epidermal cells,
stromal cells and bone marrow derived cells
profoundly influence normal differentiation
and tumour formation. We are using a variety
of approaches to study these interactions.
One line of investigation is based on the
observation that whereas integrin expression
is normally confined to the basal epidermal
layer, in many squamous cell carcinomas
expression extends to the suprabasal cell
layers. Suprabasal integrin expression results
in upregulation of Erk mitogen-activated
protein kinase (MAPK) signalling and we have
modelled this by expressing an activated MAPK
kinase 1 (MEK1) transgene in the suprabasal,
non-dividing, differentiated epidermal cell
layers (InvEE transgenics). We have found
that wounding induces benign skin tumours
in InvEE mice (Arwert et al., PNAS 2010; 107:
19903). Differentiating, non-dividing cells
that express MEK1 stimulate adjacent cells
to divide and become incorporated into the
tumour. Tumour formation is associated with
epidermal expression of IL1α and blockade
of IL1α delays tumour formation and reduces
tumour incidence. Depletion of γδ T cells and
macrophages also reduces tumour formation.
Our results are quite unexpected, because they
show that differentiated epidermal cells can
trigger tumorigenesis without re-acquiring the
ability to divide.
As we have continued to examine wound
induced tumour formation in InvEE transgenic
mice we have found that CD26 (dipeptidyl
peptidase-4) is upregulated in keratinocytes
expressing mutant MEK1 and in the
epithelial compartment of InvEE tumours,
where it accumulates at cell-cell borders.
CD26 expression is increased in dermal
fibroblasts following skin wounding but is
down‑regulated in tumour stroma. CD26
activity in keratinocytes is stimulated by
calcium-induced intercellular adhesion. IL-1α
treatment of dermal fibroblasts stimulates
CD26 activity, suggesting that epidermal IL-1α
release may contribute to the upregulation
of CD26 expression in wounded dermis.
Pharmacological blockade of CD26 reduces
the growth of InvEE tumours (Arwert et al.,
Oncogene 2011; Epub 18 July).
Our current goals are to elucidate further
changes in InvEE dermal fibroblasts that are
associated with tumour formation, and to
compare the InvEE tumour stroma with that of
human squamous cell carcinomas.
Publications listed on page 81
Research Groups | 53
Cancer and Intestinal Stem Cells
www.cambridgecancer.org.uk/dougwinton
We address how stem cell biology is exploited to maintain
intestinal cancers by developing new functional approaches to
assaying stem cells in situ. After validation in normal intestine,
these end-points are applied to assess stem-like cells in cancers
where they can be used to determine the efficacy of therapies.
Group Leader
Doug Winton
Graduate Students
Simon Buczacki
Christopher Hurley
Sarah Kozar
Hinal Tanna*
Postdoctoral Scientists
Nikki March†
Anna Nicholson*
Principal Scientific Officer
Richard Kemp
Scientific Officer
Carol Houghton
Renewing tissues and many cancers are
maintained by a small number of long-lived stem
cells. Most models of stem cell organisation
take account of their longevity and the fact that
they self-renew, and also assume that they are
stable populations carrying unique identifying
characteristics. For decades the assays used to
test different cell populations for their ‘stemness’
have appeared consistent with such deterministic
models. These assays commonly challenge the
ability of cells, separated into discrete populations
based on the expression of cell surface antigens,
to undergo growth when cultured or engrafted.
Cells that are able to support long-term growth
are viewed as being synonymous with stem cells.
However, this interpretation of stem cell
organisation now seems too simplistic. For
example: cell fate is likely determined by
small changes in the expression of regulatory
transcription factors in the context of
transcriptional networks; the cell surface
signatures of stem cells may not be as stable
over time as previously thought; the success of
stem cell engraftment may be partly determined
by properties of the recipient rather than the
transplanted cells (Chang et al., Nature 2008;
453: 544; Quintana et al., Nature 2008; 456:
593). Stem cell biology may be driven by
stochastic switching between different states in
response to variations in the balance of signals
coming from complex transcriptional networks.
In accordance with this view we have recently
demonstrated, by following the dynamics of
clonal growth in situ, that intestinal stem cell
turnover is a constant and rapid stochastic
process that follows a pattern of neutral drift
(Lopez-Garcia et al., Science 2010; 330: 822).
*
joined in 2011
†
left in 2011
Given the above our approach is pragmatic: to
identify novel ways of assaying stem cells in situ
with respect to the functional end-points that
are integral to their biology.
54 | Cambridge Research Institute Scientific Report 2011
What is the multi-potentiality of stem-like cells
in intestinal cancers?
Our long-term objective is to determine the
repertoire of differentiation options available
to cancer stem cells, how this differs from
normal stem cells, and thereby identify unique
opportunities for therapies. To measure
potentiality we are exploiting the known
differences between cell types in the timing of
DNA replication during the cell cycle. Genes
associated with maintaining pluripotency
are replicated early in S-phase, while those
associated with neural lineages are replicated
late in S-phase (Azuara et al., Nat Cell Biol 2006;
8: 532). The pattern of replication timing for
key transcription factors has been described
as a barcode of potentiality, indicative of the
accessibility of the chromatin for subsequent
expression.
We are attempting to devise such a barcode
for intestinal stem cells to identify changes in
potentiality during carcinogenesis. S-phase cells
can be isolated and sorted by DNA content
into four fractions. Immunoprecipitation
for BrdU allows newly synthesised DNA
to be analysed. To date we have shown
reproducible differences in replication timing
between different loci. For example, the neural
transcription factor Mash1 is replicated late,
while the transcription factor Ngn3, expressed
in the intestine, is replicated early. Currently,
the amount of material obtained on pull-down
is restrictive. We aim to increase genomic
coverage by amplification to generate a
comprehensive characterisation of replication
timing. The effect of deleting the APC
tumour suppressor gene on replication timing
patterns is also being determined — deleting
this gene also results in dramatic changes in
cell type (loss of secretory cell lineages) and
differentiation.
Figure 1
Quiescent cells (yellow) are
visualised in an intestinal tumour
due to retained expression of
H2BYFP fluorescent protein.
Role of quiescent stem cells
Label retaining cells, identified by their ability
to sequester and retain label, have long been
thought to be synonymous with quiescent stem
cells. Using inducible expression of nuclearlocalised fluorescent protein (Histone H2B-YFP)
we have identified a population of crypt-base
cells that appear to divide either very slowly or
to be quiescent. Conventional views of stem cell
organisation would place these cells as potential
long-lived cells acting at the apex of a proliferative
hierarchy. However, such an interpretation is
not compatible with the dynamics that we have
documented: rapid stem cell turnover with
neutral drift. It now appears that these cells are
committed to become secretory Paneth cells
and do not normally contribute to stem cell
maintenance. However, they can do so following
injury illustrating that they can be recalled to
the stem cell compartment. Importantly similar
quiescent secretory cells are found in tumours
and are also clonogenic under regenerative
conditions (Figure 1).
Cancer models and tumour progression
At a molecular level the development of
intestinal cancers is well characterised, with the
most common genetic changes incorporated
into a paradigm of progression for colorectal
cancers in which loss of APC is a central early
event (as described by Bert Vogelstein’s lab
at Johns Hopkins University). Despite this it
has been shown that many other gene specific
mutations can also be associated with the
disease (Sjoblom et al., Science 2006; 314: 268).
APC has been deleted in animal models by
a variety of strategies that usually lead to the
development of benign adenomas. Introduction
of additional mutational events in candidate
genes has only been partly successful in creating
the full carcinomatous (cancer-like) disease. Our
ability to induce deletion of APC in the intestinal
epithelium lends itself to the investigation
of the nature of other gene mutations that
might interact with APC and contribute to the
formation of malignant disease. Therefore, as
an alternative unbiased approach to identifying
such genes we used our Cre models to mobilise
a Sleeping-Beauty activated transposable
element in mice predisposed to intestinal
tumorigenesis by virtue of APC deficiency
(Collier et al., Nature 2005; 436: 272). Cloning
and sequencing of the insertion sites in tumours
allows affected genes to be identified (as
common insertion sites or CISs) and associated
with tumour pathology.
Analysis of these insertion sites identifies
hundreds of gene that are mutated in multiple
tumours (March et al., Nat Genet 2011; 43: 1202).
In trying to determine the significance of this
observation we noted that: (1) tumours are very
oligoclonal and arise against a background of a
low rate of insertional mutation, presumably
due to the process of Darwinian selection; (2)
known oncogenic pathways are repeatedly
mutated with insertions found in most tumours
in one or more of the TGFβ superfamily, p53 or
K-ras pathways; (3) no insertions were uniquely
associated with subsets of tumours identified
by pathological or other features but that
certain genes were over-represented (e.g. in
the case of tumours with abnormal Paneth cell
differentiation); (4) some CISs co-occurred with
a higher frequency than would be expected by
chance.
Publications listed on page 82
Research Groups | 55
56 | Cambridge Research Institute Scientific Report 2011
Core Facilities
The CRI’s Core Facilities provide state-of-the-art services
and equipment to support the cutting-edge research of the
Institute, as well as working towards applying new technologies
to cancer research. Each facility has a team of scientific staff
who provide scientific support, advice, and training for all CRI
researchers and students in the use of their facility’s particular
speciality, as well as keeping fully up-to-date on developing
technologies.
Confocal microscopy image of a single colony formed from
purified tumour initiating cells (TIC) from an ovarian tumour.
TICs were isolated from a patient with stage III high-grade
serous ovarian cancer by disaggregation of an omental
metastasis and flow-sorting with stem cell markers. Purified
cells were plated in a colony forming assay and the TIC
frequency was approximately 1/250 cells. The development
of reproducible stem cell markers for ovarian cancer is now
being used to understand molecular heterogeneity in patients
and how differences in TICs can explain drug resistance.
Immunofluorescence staining shows cytokeratin in cancer cells
(Green = cytokeratin 7, Red = cytokeratin 18, blue = DAPI).
Image provided by Siru Virtanen (Stingl and Brenton
laboratories)
Core Facilities | 57
Bioinformatics
www.cambridgecancer.org.uk/bioinformatics-core
The Bioinformatics Core offers a data analysis and statistics
consulting service to CRI research scientists and develops software
and analysis pipelines to support high-throughput technologies
including next generation sequencing and microarrays.
Core Facility Manager
Matthew Eldridge
Staff
Richard Bowers
Tom Carroll*
Ben Davis
Sarah Dawson
Mark Dunning
Silvia Halim*
Stewart MacArthur †
Suraj Menon
Liz Merrall*†
Anne Pajon*
Roslin Russell
Rory Stark
Sarah Vowler
In the past year the facility has supported a large
number of research projects by consulting with
scientists, providing input into experimental
design, analysing the data generated and
assisting with the interpretation of results.
ChIP-seq experiments continue to be a major
theme and we have further developed our
analysis capability, recently releasing DiffBind, an
R package for identifying DNA/protein binding
sites that are differentially bound between two
or more sample groups based on sequencing
read densities. Researchers are increasingly
using multiple experimental techniques to study
the same set of tumour samples and we have
been applying statistical techniques as the basis
of an integrative analysis combining, for example,
transcription factor binding sites determined
using ChIP-seq and gene expression levels from
a microarray experiment.
The core has been developing a workflow
framework that provides a structured approach
for creating analysis pipelines, promoting
reuse of defined tasks and their efficient
execution on the institute’s high-performance
Differentially bound oestrogen
receptor sites can separate breast
cancer tumours from patients with
good or poor prognosis using data
from ChIP-seq (left) and expression
microarrays (right).
*
joined in 2011
†
left in 2011
58 | Cambridge Research Institute Scientific Report 2011
compute cluster. This underlies the automated
pipelines for contaminant screening and
alignment of sequencing data generated by
the high-throughput sequencing instruments
in the Genomics Core. We are also using the
framework to develop workflows for assessing
data quality and detecting somatic mutations
and structural variation in whole genome
sequencing datasets. We have been working
closely with Simon Tavaré’s group on an
oesophageal cancer project funded by Cancer
Research UK as part of the International Cancer
Genome Consortium.
Throughout the year we have been running
weekly experimental design sessions, jointly
with the Genomics Core, and statistics clinics
to provide bioinformatics and statistical input
into a wide range of experiments and research
questions. We ran training courses on DNA
motif searching and functional and network
analysis and a newly developed course on
introductory statistics has proved very popular.
Finally, a successful review of the core was
conducted in January by an external panel of
bioinformatics experts.
Publications listed on page 82
Biological Resources Unit
The BRU facility within the CRI has the ability to offer both
a state of the art animal facility and a variety of associated
services to Cancer Research UK and its collaborators.
Core Facility Manager
Allen Hazlehurst
Isolation and Containment
The isolation and containment suite was
primarily set up to allow animals with certain
named pathogens to be housed and imaged
within the CRI, using facilities such as our
Xenogen, MRI multi-proton microscope and
PET/SPECT scanners. Animals that have been
exposed to CL2 products will also be housed
within this area, allowing work with potentially
hazardous materials to be contained.
We also currently have four isolators. This will
ensure that shipments are kept isolated from
other shipments that may have the potential to
pass on pathogens as yet unseen in the colony.
It also ensures that they are segregated from
the main colonies in the CRI. From these we
will have the potential to re-derive mice into
the barrier should the goal be to establish a
breeding colony within the CRI.
A dedicated husbandry team ensures all
husbandry needs are met and are also able to
carry out specialist technical and licensed tasks
on any researcher’s behalf.
Figure 1
Part of the newly refurbished
surgical suite.
Figure 2
An isolation cabinet for housing
animals from outside the CRI.
*
joined in 2011
†
Import/export programme
The animal model service at the CRI has the
responsibility of arranging the importation
and exportation of whole animals, tissues and/
or embryos and sperm to/from any external
collaborators and/or commercial establishment
located either nationally or internationally. As
part of the service we carry out the following:
•Source particular strains
•Identify and apply for required government
licences
•Complete import and excise paperwork
•Find the most suitable shipping agents
•Arrange transportation
•Liaise with both the shipping agents and the
external collaborators.
Transgenic service
Our dedicated Transgenic team offer a variety
of bespoke transgenic services. These include
the cryopreservation of embryonic and sperm
cells, the derivation of new mouse ES cell lines,
siRNA transgenesis or embryo aggregation,
embryo or oocyte collection and transgenic
advice.
Regulatory compliance advisory service
The use of animals for scientific procedures is
controlled by the Animals (Scientific Procedures)
Act 1986. Three licences are required to be
in place before any regulated procedures can
take place. These are a personal licence (PIL),
a project licence (PPL) and a certificate of
designation (Cert Des). A regulated procedure
under the act is described as any experimental
procedure applied to a protected animal that
may cause pain, suffering, distress or lasting
harm. This also includes procedures such as
breeding. The regulatory compliance group
offers a wide range of licensing tools and up to
date advice designed to ensure that all local and
national requirements have been appropriately
addressed both at the beginning and throughout
the duration of research projects.
left in 2011
Core Facilities | 59
Biorepository and Cell Services
www.cambridgecancer.org.uk/biorepository-core
Our service allows simple access to storage, tracking and risk
management of tissue samples, cell lines and any other biological
samples, including human tissue samples, in accordance with
current legislation.
Core Facility Manager
Bob Geraghty
Senior Scientific Officer
Maria Vias
Scientific Officers
Petra Chovancova*
Jorgelina Trueba-Lopez
We provide up-to-date expertise, training and
troubleshooting in all aspects of cell and tissue
culture, to maintain a consistently high standard
throughout the Institute. The facility is used
extensively by most CRI research groups, and
this year there has been as significant increase in
the number of human tissue samples received,
and cell samples submitted for mycoplasma
testing.
Cell culture
We provide basic cell culture training for all
CRI scientists, a comprehensive mycoplasma
testing service, a batch testing service for serum
and other cell culture media components, and
quality controlled bulk culture of research cell
lines, including mouse embryonic fibroblasts
(MEFs). We also offer a routine human cell line
authentication service using multiplex PCR and
short tandem repeat (STR) profiling. Regular
mycoplasma testing and cell line authentication
is important to confirm integrity of data and is
becoming a requirement for publication in many
leading journals.
We currently support two Essen BioScience
IncuCyte™ instruments which are compact,
automated imaging platforms designed to
provide kinetic, non-invasive live cell imaging.
The instruments are located in a 5% CO2
incubator and acquire high definition phase
contrast and fluorescent images of live cells in
vitro in cell culture microplates, dishes and flasks.
Custom image processing software calculates
a variety of metrics, such as cell proliferation
and migration assays and growth curves,
optimisation of cell based assays and of cell
culture media components. These instruments
are very popular and in December 2011 we
ordered a third instrument to reduce the
waiting time for experiments.
*
joined in 2011
†
left in 2011
60 | Cambridge Research Institute Scientific Report 2011
The Human Tissue Act
Our staff advise on, monitor and control the
import, use, storage and disposal of human
tissue samples for research, to ensure full
compliance with the Human Tissue Act and
the Human Tissue Authority (HTA) Codes of
Practice, a statutory requirement for all research
involving human tissue samples. We advise on
how to request human tissue samples from the
Addenbrooke’s Hospital tissue bank and other
sources, and how to obtain local research ethics
committee approval for new research projects
involving the use of human tissues.
Future developments
We have identified a number of new services
and initiatives that will further facilitate
and enhance research at the CRI when
implemented. These include:
•Introducing a core cell culture karyotyping
service. This will complement our STR
profiling service and will, for example, allow
us to distinguish between mouse and human
cell lines and to visualise various chromosome
abnormalities which could lead to unexpected
STR profiles.
•Introducing and optimising a phage display
antibody library. A human single fold scFv
library will enable us to very rapidly derive
monoclonal antibodies to almost any target
molecule requested.
•We expect the number of human tissue
samples entering the CRI for research
purposes, in particular samples associated with
clinical trials, will continue to increase and we
will be involved in ensuring that these samples
fully comply with MHRA regulations.
Equipment Park
www.cambridgecancer.org.uk/equipment-park-core
The Equipment Park provides CRI scientists with access
to a range of state-of-the-art equipment and specialised
technologies.
Core Facility Manager
Jane Gray
Scientific Officers
Ian Hall*
Maeve McEnery †
Our lab offers technical/scientific advice,
troubleshooting support and appropriate
training for all the facility’s equipment. We also
routinely test the capabilities of our equipment,
optimise current techniques and horizon scan
to maximise the quality of data generated
and to provide the best possible advice to
CRI scientists. This year we have focused on
optimising Western blotting techniques as
these are routinely carried out by the Institute’s
researchers.
Protein gel electrophoresis
We provide access to a wide range of gel
electrophoresis equipment for analysis of
protein samples. We have the capability for
both 1- and 2-dimensional separation of proteins
including 2D fluorescence difference in‑gel
electrophoresis (2D-DiGE). Together with our
range of digital camera and scanner imaging
systems, we can digitise images which improves
accuracy of quantification, saves time and
reduces costs. This year we have investigated
the effectiveness of different housekeeping
proteins for quantitative Western blotting and
also introduced an intensive Western blotting
training course for CRI scientists.
Biosensor
The Biacore T100 measures molecular
interactions in real-time. It provides label-free
measurements of the affinity and kinetics of
interactions, as well as the thermodynamic
properties underlying association and
dissociation rates. This instrument has proved
pivotal in a number of research studies this year,
carried out by the Murphy, Balasubramanian and
Brindle laboratories, investigating the kinetics
of the following interactions: protein-protein,
small molecule binding to DNA, and proteinphospholipid.
*
joined in 2011
†
left in 2011
Plate readers and spectrophotometers
The Equipment Park provides access to three
high specification plate readers: the Tecan
Infinite M200 is used extensively by most
research groups at the CRI for absorbance,
fluorescence and/or luminescence assay work,
and we also house a BioTek Clarity, a dedicated
luminescence plate reader. This year we have
also introduced a third new plate reader, a BMG
PHERAstar FS, which allows users to perform
higher-end assays including time-resolved
fluorescence or fluorescence polarisation and
also increases sample throughput with its
automated plate stacker. A fourth UV‑visible
cuvette spectrophotometer, the Cecil Super
Aquarius 9500, is particularly suited to
quantification of low-concentration samples.
Imaging systems
Four imaging systems are available that
produce digital images from a wide range of
different samples. The Typhoon Trio produces
images of radioactive, visible fluorescent or
chemiluminescent samples while the Li-Cor
Odyssey images fluorescence specifically in the
infrared region. Both systems are used routinely
for Western blotting and cell-based assays. The
ImageScanner III is a high-resolution flatbed
scanner for imaging non-fluorescent samples.
We also have a high resolution camera system,
the Syngene Dyversity, capable of capturing
both fluorescent and chemiluminescent images.
Dedicated analysis software packages can
accurately quantify protein/DNA bands or spots
captured by any of these imaging systems.
Molecular biology applications
The Equipment Park houses an 8-channel
NanoDrop as well as a Qubit for quantification
of small volume nucleic acid (and protein)
samples and has the capability for both standard
and real-time PCR. We also have a pulsedfield gel electrophoresis system, CHEF III, for
separation of large DNA molecules and an
E-Gel iBase for fast separation of DNA and
RNA.
Core Facilities | 61
Flow Cytometry
www.cambridgecancer.org.uk/flow-cytometry-core
The Flow Cytometry core facility provides state-of-the-art flow
cytometric instrumentation, technical expertise, training, and
software analysis in a collaborative environment. Our mission
is to develop cytometric technologies that will best assist CRI
researchers in finding answers for the treatment, prevention,
and understanding of cancer.
Core Facility Manager
Richard Grenfell
Senior Scientific Officer
Reiner Schulte
Scientific Officer
Loïc Tauzin*
Visiting Worker
Lizz Grimwade
Services
Our lab offers a full range of educational
and cytometric services that includes
immunophenotyping, cell cycle analysis,
translocation and co-localisation of cell
activation markers, chromatin density, and
apoptotic and necrotic analysis. In addition
we are capable of performing cell sorting
for researchers so that they can isolate cell
populations needed for further studies.
Users are offered an array of educational
programs in the theory, anatomy, applications
and science of flow cytometry. Additional
workshops are offered on data analysis using
all of our software programs and on practical
applications of current protocols in cytometry.
We also collaborate with other scientists in the
Cambridge Cancer Centre on our specialised
equipment.
Equipment
FACS Aria SORP (BD Biosciences) – The
Aria is a high-speed sorter. It is equipped with
five lasers: a UV, 407nm, 445nm, 488nm, and
633nm. Our optical configuration allows us to
see three UV, six violet, three indigo, six blue
and three red parameters.
LSR II (BD Biosciences) – The LSR II is an
analytical bench top flow cytometer. It
is comprised of four lasers: a UV, a violet
(407nm), a blue (488 nm) and a red (633 nm).
Our optical configurations allow users to see
two UV, six violet, seven blue and three red
fluorescent parameters.
FACS Caliburs (BD Biosciences) – These flow
cytometers are routinely used for phenotyping
(to look at antigen, cytokine, or GFP
*
joined in 2011
†
left in 2011
62 | Cambridge Research Institute Scientific Report 2011
expression), cell cycle analysis, and apoptosis
studies. They are equipped with 488nm and
635nm lasers that allow users six parameter
analysis.
ImageStream (Amnis) – The powerful
combination of quantitative image analysis and
flow cytometry in a single platform creates
exceptional new experimental capabilities.
405nm, 488nm and 635nm lasers for four
colour/six parameter analysis as well as EDF
capability for FISH analysis are available.
Influx Cell Sorter (BD Biosciences) – This
high speed cell sorter is contained within a
biosafety cabinet to enable the isolation of cell
populations from human tissue. It has four
lasers at 405nm, 488nm, 561nm, 640nm and is
equipped with 12 fluorescence detectors.
RoboSep (Stem Cell Technologies) – This
magnetic bead separator unit has customisable
programs allowing positive or negative
selection of virtually any cell type from any
species. Up to four samples can be processed
simultaneously.
Vi-CELL (Beckman Coulter) – The Vi-CELL
automates the widely accepted trypan blue cell
exclusion method, with video imaging of the
flow-through cell, to obtain results in minutes.
The software conforms to key regulatory
requirements with its electronic signature
capability, audit trail, secure user sign on and
user level permissions for clinical or preclinical
studies.
Publications listed on page 83
Genomics
www.cambridgecancer.org.uk/genomics-core
The Genomics core facility allows researchers at the CRI
access to state-of-the-art DNA and RNA analysis instruments,
methods and applications.
Core Facility Manager
James Hadfield
Senior Scientific Officers
Sarah Aldridge†
Sarah Leigh-Brown*
Michelle Osborne
Scientific Officers
Claire Fielding
Hannah Haydon
Fatimah Madni*
The tools in Genomics help researchers to
understand the cancer genome and unravel
the genetic causes of cancer. Next-generation
sequencing technology is changing how we
look at cancer biology and cancer medicine and
the systems in the Genomics core help CRI
scientists answer questions in these areas.
In the last year it has become possible to
sequence the entire genome of a cancer patient
using next-generation sequencing (NGS). It is
likely to be several years before this technical
possibility becomes a clinical tool, however we
can now sequence a human genome in around
five days and perform unbiased genomewide experiments to see what the underlying
sequence differences are in cancer genomes.
We make extensive use of the Illumina NGS
technology (Figure 1) and the CRI has invested
significant time and resources in both the
Genomics and Bioinformatics core facilities
to become a centre of excellence in this
technology. Having the capability to access new
systems like this puts the CRI at the forefront of
genomic research.
Figure 1
The Illumina MiSeq 2000
next‑generation DNA sequencer.
*
joined in 2011
†
left in 2011
The genomics core has helped in the
development of ChIP-seq at the CRI (Schmidt
et al., Methods 2009; 48: 240) and this technique
has been used in many publications over the
last four years. The Carroll group recently
published work improving our understanding
of the estrogen receptor transcriptional
complex, which plays an important role in
breast cancer cells (Holmes et al., PNAS 2011;
Epub 2 May). The genomics core previously
worked with the Caldas group to assess miRNA
analysis methods. We were recently asked
to contribute the introductory chapter to a
new miRNA methods textbook (Aldridge and
Hadfield 2012; Methods Mol Biol 822: 19). We
also contributed to the Technology working
group of the Cancer Research UK Stratified
Medicines Initiative.
Microarrays allow us to analyse gene expression
(mRNA levels) and structural variation (DNA
copy number) on a genome-wide level.
Even with NGS we still use large numbers
of commercial arrays from Illumina, Agilent
and Affymetrix in a wide variety of research
projects. Gene expression analysis has become
a standard tool for biologists and microarrays
can be used to measure the amount of RNA
from a gene and help us to discover the genetic
drivers of cancer.
An important component of the Genomics core
facility is our staff. The technologies we use are
complicated and we undertake projects for the
Institute’s research groups as well as training
individuals to use Genomics core equipment.
We also offer support and access to other
genomics platforms including: Real-time PCR
for lower throughput gene expression and
copy number analysis; Pyrosequencing to look
at methylation of DNA; Agilent Bioanalyser
instruments to quality control RNA and DNA;
and Qiagen robotics for nucleic acid extraction.
We have recently installed a new system
from Fluidigm that allows us to run very high
throughput real-time PCR projects and also
to amplify regions of the genome for targeted
resequencing using NGS. This work is being
used to develop new diagnostic tests for cancer.
Publications listed on page 83
Core Facilities | 63
Histopathology and In Situ Hybridisation
www.cambridgecancer.org.uk/histopathology-core
The Histopathology/ISH core facility at the Cambridge
Research Institute offers a variety of histological techniques,
immunohistochemistry, in situ hybridisation, laser capture
microdissection as well as automatic slide digitisation and
analysis to CRI scientists.
Core Facility Manager
Will Howat
Senior Scientific Officers
Andrew Cassidy *
Julia Jones
Jodi Miller
Beverley Wilson
Scientific Officers
James Atkinson
Margarita Bennett
Louise Howard
Leigh-Anne McDuffus
Angela Seedhar †
Pooja Seedhar †
Cara Walters*
Visiting Workers
Fiona Blows
Kristy Driver †
Carrie-Ann Gilbey *
Amanda Khogali*†
Lyndsey Offord†
Histology
The facility processes, embeds and sections
human and animal tissues or cell lines into frozen
or paraffin formats and stains these with the
standard haematoxylin and eosin (H&E) or
special stains, as needed by the researcher to
complement their work. During the past year, we
modified our methods to allow for the acquisition
of sections from 3D spheroid co-cultures, as well
as adding these special stains: Masson Fontana
for demonstration of melanin/argentaffin; Perl’s
Prussian Blue for iron; Martius Scarlet Blue
for fibrin/collagen/muscle; AgNOR for nuclear
organiser regions; Millers Elastic for elastic fibres;
PAS diastase for the demonstration of glycogen.
Immunohistochemistry (IHC)
We have completed a collaborative project with
the Caldas laboratory and through this work have
validated 111 antibodies for use on the Bondmax
automated immunohistochemistry stainer. In
addition, a further 19 antibodies have been
validated through our routine IHC validation
request service. The combination of this, as well
as an increase in routine work has led to a further
increase in routine antibody staining to 22,000
slides this year. We aim to launch a protocol for
TUNEL to assist in the identification of apoptotic
cells in our studies in the next six months.
Figure 1 (left)
miR-205 in situ hybridisation
staining of breast lobules
demonstrating staining in the basal
cells of the lobule.
In situ hybridisation (ISH)
Our gold standard ISH protocol utilises
35
S‑labelled riboprobes and we offer
fluorescence ISH (FISH) for the Y chromosome
and human/mouse centromeric regions as part
of the routine service. We now also offer
micro-RNA ISH staining (Figure 1) as well as
having validated a method for detecting the 5′
leader sequence of rRNA. We aim to evaluate
and release methods for using the Panomics ISH
kits within the facility in the next year.
Digitisation and analysis
The number of slides being scanned through
the three scanners (Leica Ariol SL50, Aperio
XT, Zeiss Mirax) has increased by 17% to
35,000 slides over the course of the year. In
particular, the Ariol slide scanning system,
while scanning the same number of slides,
has changed its usage from 16% to 50% for
fluorescence images. We have continued
to train users in the use of image analysis for
automatically counting cellular staining and
have also purchased the TMALab, Microvessel
density and Aperio Genie tissue analysis
software to allow us to offer more extensive
automatic analysis of areas of necrosis and
tumour identification (Figure 2). We hope to
grow this further through collaborations with
the Breast Cancer Association Consortium and
International Cancer Genome Consortium.
A
Figure 2 (right)
Automated image segmentation.
(A) Tumour and associated
non-tumour tissue stained with
Haematoxylin and CD31. (B)
Automatic recognition of tumour
area using Aperio Genie software.
*
joined in 2011
†
left in 2011
64 | Cambridge Research Institute Scientific Report 2011
Tumour
B
Light Microscopy
www.cambridgecancer.org.uk/light-microscopy-core
The Light Microscopy Facility provides the CRI with state-ofthe-art light microscopy and develops new imaging modes.
Core Facility Manager
Stefanie Reichelt
Principal Scientific Officer
Heather Zecchini
Senior Scientific Officers
Lorraine Berry
John Harris†
Visiting Scientist
Brad Amos
The facility specialises in: advanced live-cell
imaging using wide-field and spinning disc
imaging systems; confocal scanning light
microscopy; non-linear imaging techniques such
as multi-photon, second harmonic, fluorescence
life-time imaging (FLIM); in vivo imaging at
high‑resolution; quantitative high throughput
image acquisition and analysis.
The CompuCyte iCys imaging cytometer is
the most popular quantitative high‑throughput
imaging and analysis system in the facility.
Current applications include measuring
ligand uptake, apoptosis, tumour vasculature
and drug distribution and DNA damage in
cancer cells, but also tissue microarrays. The
characterization of surface FAS with quantitative
morphological analysis using quantitative imaging
cytometry is being published in early 2012
(Ireland-Zechinni et al., Curr Protoc Cytometry
2012; Epub 1 Jan).
The LaVision TriMScope system, equipped
with an additional optical parametric oscillator,
provides fs-pulsed MP-excitation ranging from
690nm to 1600nm. The TriMScope is a very
sensitive and rapid multi-photon scanning
system, which is used mainly for imaging live
tumour samples.
We have upgraded the live cell imaging systems
with an EMCCD and the newest cCMOS
camera, which is simultaneously delivering
ultra‑low noise, fast frame rates, wide dynamic
range, high resolution and a large field of view
for fast live-cell imaging.
3D reconstruction of a confocal
image stack using Volocity ®
software of HeLa cells stained using
an anti-tubulin immunofluorescence
method. Image: Stefanie Reichelt,
Light Microscopy; sample: Joo-Hee
Sir, Gergely laboratory.
*
joined in 2011
†
We are also constantly monitoring new
developments in imaging techniques. We have
tested and applied high resolution imaging
(OMX) in collaboration with the Wellcome
Trust Gurdon Institute and CRI colleagues (Sir
et al., Nat Genet 2011, 43; 1147; Narita et al.,
Science 2011; 332: 966). We have successfully
used a supercontinuum white-light laser to carry
out interferometric biological imaging (Chiu et
al., J Microscopy 2012; In press).
An EMBO-funded annual course in Plymouth
has become a centre for training and discussion
in advanced optical microscope methods,
attracting leading lecturers, manufacturers and
students from the whole of Europe, including
students from the CRI. We continue to
contribute to the MONABIPHOT Erasmus
Course (coordinator: Prof. Zyss, ENS Cachan,
France) by hosting masters students.
Research and Development
Current projects include the following:
(1) We are using second harmonic imaging
based on a scattered signal, e.g. to demonstrate
the formation of vessels from endothelial cells
as well as the extracellular matrix in tumours,
and cell behaviour in collagen matrices. We
are working with Carola Schoenlieb (DAMTP,
Cambridge) to develop image analysis tools to
quantify tumour/stroma ratios in cancer.
(2) CARS imaging is being combined with
fluorescence imaging in cancer drug up-take
studies in collaboration with Sumeet Mahajan
(Department of Physics, Cambridge). This
project has been awarded a CCC pump-priming
grant.
Publications listed on page 83
left in 2011
Core Facilities | 65
Pharmacokinetics and Pharmacodynamics
We work with the remit to provide pharmacokinetic and
pharmacodynamic support for the Institute.
Core Facility Manager
Donna Smith
Senior Scientific Officer
Michael Williams
Scientific Officers
Dorentina Bexheti*
Zoe Drymoussi†
Deneen Holohan*
Pharmacokinetics (PK) is the study of what the
body does to drugs. It is the mathematical study
and description of the absorption, distribution,
metabolism and excretion processes used by
the body when a drug is administered. In order
to obtain good PK data, bioanalysis forms an
integral part of the science and to facilitate
this we have two liquid chromatography-mass
spectrometry systems (LC-MS/MS) within
the facility (Figure 1). These state-of-the-art
systems enable us to detect very low levels of
drugs in a variety of biological matrices such as
blood, plasma, tumour and cell cultures. We
have developed a range of bioanalytical assays
to support various research groups ranging from
small dicarboxylic acids to steroids. In addition
a number of bioanalytical assays have been
validated to support clinical trial studies.
We continue to evaluate the use of dried
blood spots (DBS) as a sampling technique
for bioanalysis. This leading edge bioanalytical
technology uses small volumes of blood for
sampling (typically around 30 µL). This can
have a positive impact on the refinement and
Figure 1
Liquid chromatography-mass
spectrometry system.
*
joined in 2011
†
left in 2011
66 | Cambridge Research Institute Scientific Report 2011
reduction of in vivo studies with the potential to
obtain high quality PK data from efficacy studies.
Initial results have been very promising and
there is strong interest for the application of this
technique in paediatric oncology clinical trials.
Pharmacodynamics (PD) is the study of what
the drug does to the body (i.e. its effect). By
relating PD effects to PK parameters, the PK/PD
relationship can be determined. To this end, a
variety of PD assays (e.g. biomarker assays) were
established to support several clinical trials. We
are looking to expand our portfolio for 2012. In
2011 Deneen Holohan replaced Zoe Drymoussi
as our PD contact in the core facility.
As we are working with clinical samples the
facility will be compliant to the MHRA guidelines
entitled ‘GCP in the Clinical Laboratory’.
In addition to the analysis of PK samples we
can also offer advice on the design of PK and
efficacy studies.
Publications listed on page 83
Pre-clinical Imaging
www.cambridgecancer.org.uk/preclinical-imaging-core
Pre-clinical imaging is a collaborative facility that manages a
wide range of imaging machines for the CRI.
Core Facility Manager
Kevin Brindle
Core Facility Manager
John Griffiths
Staff
Madhu Basetti
Mikko Kettunen
David Lewis
Scott Lyons
Dominick McIntyre
Mary McLean
Dmitry Soloviev
*
joined in 2011
†
left in 2011
Optical
IVIS 200 and IVIS lumina imaging systems (Caliper
Lifesciences) are available for whole-animal in vivo
photonic imaging, including sensitive and relatively
high-throughput in vivo bioluminescence imaging.
Typical scans take less than one minute and up to
five subjects can be imaged at a time.
MRI
We have two Varian MRI systems; a 9.4T with
higher sensitivity, and a 7T whose smaller
susceptibility effects make it more suitable for
techniques such as echo-planar imaging. Both
perform 1H MRI and multi-nuclear MRS, assisted
by integrated monitoring, gating, heating and
anaesthesia. We have produced DCE-MRI data
for vascular characterisation of autochthonous
pancreatic tumours (Tuveson laboratory). We
have implemented improved 1H MRS methods
that minimise chemical shift artefacts and we are
developing quantitative MT-MRI and motioninsensitive DW-MRI methods for abdominal
tumours, which are subject to respiratory
and cardiac motion. Our Hypersense system
is now routinely producing a wide range of
hyperpolarised substrates for high-sensitivity 13C
in vivo tumour metabolism studies.
Radiotracer methods
We have a NanoPET/SPECT/CT
(Mediso/Bioscan/Philips) system for
multimodality radionuclide imaging. This offers
the greatest sensitivity of any in vivo imaging
modality and provides non-invasive assessment
of pharmacological (target tissue exposure,
target engagement and functional activity) and
biological processes (blood flow, perfusion
and metabolism). These scanners can resolve
nanolitre volumes (~0.4mm for SPECT and
~1mm for PET) and so are ideal for small animal
imaging.
Static and dynamic imaging can be implemented,
with or without respiratory/cardiac gating. We
can label biologically active molecules with
radionuclides and perform kinetic imaging.
Simultaneous dual isotope studies are possible
using nanoSPECT and we are investigating
multimodal approaches for the integrated
molecular imaging of cancer.
A 68Ga generator was installed in 2011 – we are
setting up a laboratory for radiolabelling with
68
Ga and SPECT radionuclides in collaboration
with the radiopharmacy and PET/CT facility at
Addenbrooke’s Hospital, and are establishing
protein labelling methods with 68Ga.
Molecular imaging probes currently available
for PET include [18F]FDG, [18F]FLT, [18F]FMISO,
[18F]FET, [11C]acetate. Firstly we will focus on
novel molecular marker development and use of
PET and SPECT to measure early response of
tumours to therapy. The Brindle laboratory are
investigating the C2A domain of synaptotagmin,
labelled with 111In for SPECT and 68Ga for
PET, as a novel probe for detection of tumour
cell apoptosis post treatment and the use of
[11C]acetate for early detection of malignant
transformation.
Metabolomics
The facility is based on a Bruker 600MHz
NMR instrument. High resolution 1H, 13C
and 31P NMR studies are performed routinely
on solution samples. An HRMAS 1H and 31P
NMR probe allows biochemical analysis of
intact ex vivo clinical and preclinical biopsies.
Ongoing collaborations include studies on
cellular senescence (Narita laboratory) and 19F
NMR of anticancer drug metabolites (Tuveson
laboratory). With the Tavaré laboratory we are
developing metabolite correlation methods to
interpret the biochemical data.
Ultrasound
Our ultrasound imaging provision includes
two Vevo 2100 systems and one Vevo 770
system (Visualsonics). These image to as low
as 30 micron resolution, providing excellent
anatomical and soft tissue structural detail
instantaneously and in real-time, and also permit
rapid 3D imaging and dynamic vascular imaging
with power Doppler and non-linear contrast.
Core Facilities | 67
Proteomics
www.cambridgecancer.org.uk/proteomics-core
The Proteomics core facility focuses on the systematic study
of proteins, particularly their structures, interactions and
expression levels. The facility is equipped with state-of-theart instrumentation for CRI researchers requiring access to
proteomics technology and expertise.
Core Facility Manager
Clive D’Santos
Senior Scientific Officer
Xiaoping Yang
Scientific Officer
Chris Taylor
Figure 1
Dionex Ultimate 3000 RSLC
nanoHPLC system.
The Proteomics core facility provides help
in designing experimental strategies and
implements and validates previously developed
proteomic workflows to profile proteins from
diverse biological samples. We also aim to
modify or develop entirely new methods and
assays when warranted. In addition, we have
bioinformatics support for data management
and analysis as well as software development.
The facility has already been well equipped
with state-of-the-art analytical instrumentation
for proteomic studies, including the latest
orbitrap mass spectrometer, the LTQ Velos
Orbitrap (Thermo), which has been configured
to a Dionex Ultimate 3000 RSLC nanoHPLC
system (Figure 1). In addition an Agilent
6520 QToF with ChIP cube technology was
installed in 2010. The mass spectrometers
are supported by off-line chromatography
platforms: two Dionex Ultimate 3000
capHPLC systems for multidimensional
chromatography at the protein and peptide
level. These are supported by 1D and 2D
gel electrophoresis systems as well as a GE
Healthcare Typhoon Trio+ imager available
in the equipment park run by Jane Gray.
Data analysis is supported by an array of
bioinformatics and statistical analysis tools.
Specific methods and areas of interest include:
Protein profiling of complex biological samples,
e.g. serum, tissue, cell extracts
•Profiling by nanoLC/MS
•Multidimensional protein/peptide
fractionation by capLC and/or geLC.
*
joined in 2011
†
left in 2011
Targeted protein identification by
nanoLC/MS/MS
•Coomassie and silver stained gel bands of
purified proteins
•In solution digestion of purified proteins.
68 | Cambridge Research Institute Scientific Report 2011
Identification of protein and peptide
modifications
•Phosphorylation sites
•Protein modifications such as acetylation
and methylation,
•Coomassie stain only, purified proteins.
Relative quantitation by nanoLC/MS/MS
•SILAC - stable isotope labeling of amino
acids in cell culture
•ITRAQ - an isobaric peptide tagging system.
Publications listed on page 83
Oral cancer cells during the process of migration in a 2D
substrate. The actin cytoskeleton, shown in red, is a very
dynamic system of filaments that plays a role in cell motility
by pushing and pulling on different parts of the cell. The
protein paxillin, shown in green, indicates the location of focal
adhesions, the contact structures that fix the cells to the
substrate beneath. Image provided by Carles Escriu (Watt
laboratory).
Core Facilities | 69
The CRI graduate students meet with James D Watson
70 | Cambridge Research Institute Scientific Report 2011
Institute Information
Institute Information | 71
Research Publications
Shankar Balasubramanian (page 12)
Chemical biology of nucleic acids laboratory
Primary research papers
Dash J, Waller Bradbrook ZA, Pantos GD,
Balasubramanian S. Synthesis and binding studies
of novel diethynyl-pyridine amides with genomic
promoter DNA G-quadruplexes. Chemistry. 2011;
17: 4751-81
Hegde NS, Sanders DA, Rodriguez R,
Balasubramanian S. The transcription factor
FOXM1 is a cellular target of the natural product
thiostrepton. Nat Chem. 2011; 3: 725-31
Koirala D, Dhakal S, Ashbridge B, Sannohe Y,
Rodriguez R, Sugiyama H, Balasubramanian S, Mao
H. A single-molecule platform for investigation of
interactions between G-quadruplexes and smallmolecule ligands. Nat Chem. 2011; 3: 782-7
Lightfoot HL, Bugaut A, Armisen J, Lehrbach NJ,
Miska EA, Balasubramanian S. A LIN28-dependent
structural change of pre-let-7g directly inhibits Dicer
processing. Biochemistry. 2011; 50: 7514-21
McLuckie KI, Waller ZA, Sanders DA, Alves D,
Rodriguez R, Dash J, McKenzie GJ, Venkitaraman
AR, Balasubramanian S. G-quadruplex-binding
benzo[a]phenoxazines down-regulate c-KIT
expression in human gastric carcinoma cells. J Am
Chem Soc. 2011; 133: 2658-63
Raiber EA, Kranaster R, Lam E, Nikan M,
Balasubramanian S. A non-canonical DNA
structure is a binding motif for the transcription
factor SP1 in vitro. Nucleic Acids Res. 2011; Epub Oct
22
Sarkies P, Murat P, Phillips LG, Patel KJ,
Balasubramanian S, Sale JE. FANCJ coordinates
two pathways that maintain epigenetic stability at
G-quadruplex DNA. Nucleic Acids Res. 2011; Epub
Oct 22
Smith JS, Chen Q, Yatsunyk LA, Nicoludis JM,
Garcia MS, Kranaster R, Balasubramanian S,
Monchaud D, Teulade-Fichou MP, Abramowitz
L, Schultz DC, Johnson FB. Rudimentary
G-quadruplex-based telomere capping in
Saccharomyces cerevisiae. Nat Struct Mol Biol. 2011;
18: 478-85
Zhang AY, Bugaut A, Balasubramanian S. A
sequence-independent analysis of the loop length
dependence of intramolecular RNA G-quadruplex
stability and topology. Biochemistry. 2011; 50: 7251-8
Other publications
Balasubramanian S. Sequencing nucleic acids: from
chemistry to medicine. Chem Commun (Camb). 2011;
72 | Cambridge Research Institute Scientific Report 2011
47: 7281-6
Balasubramanian S. Decoding genomes at high
speed: implications for science and medicine. Angew
Chem Int Ed Engl. 2011; 50: 12406-10
Balasubramanian S, Hurley LH, Neidle S. Targeting
G-quadruplexes in gene promoters: a novel
anticancer strategy? Nat Rev Drug Discov. 2011; 10:
261-75
James Brenton (page 14)
Functional genomics of ovarian cancer laboratory
Primary research papers
Lawson MH, Cummings NM, Rassl DM, Russell
R, Brenton JD, Rintoul RC, Murphy G. Two novel
determinants of etoposide resistance in small cell
lung cancer. Cancer Res. 2011; 71: 4877-87
Ng CK, Cooke SL, Howe K, Newman S, Xian J,
Temple J, Batty EM, Pole JC, Langdon SP, Edwards
PA, Brenton JD. The role of tandem duplicator
phenotype in tumour evolution in high-grade
serous ovarian cancer. J Pathol. 2011; Epub Dec 19
Shearman JW, Myers RM, Brenton JD, Ley SV.
Total syntheses of subereamollines A and B. Organic
& Biomolecular Chemistry. 2011; 9: 62-5
Other publications
Cooke SL, Brenton JD. Evolution of platinum
resistance in high-grade serous ovarian cancer.
Lancet Oncol. 2011; 12: 1169-74
Gounaris I, Charnock-Jones DS, Brenton JD.
Ovarian clear cell carcinoma– bad endometriosis or
bad endometrium? J Pathol. 2011; 225: 157-60
Vaughan S, Coward JI, Bast RC, Jr., Berchuck A,
Berek JS, Brenton JD, et al. Rethinking ovarian
cancer: recommendations for improving outcomes.
Nat Rev Cancer. 2011; 11: 719-25
Kevin Brindle (page 16)
Molecular imaging of cancer laboratory
Primary research papers
Bohndiek SE, Kettunen MI, Hu DE, Kennedy BW,
Boren J, Gallagher FA, Brindle KM. Hyperpolarized
[1-13C]-ascorbic and dehydroascorbic acid: vitamin
C as a probe for imaging redox status in vivo. J Am
Chem Soc. 2011; 133: 11795-801
Crossan GP, van der Weyden L, Rosado IV,
Langevin F, Gaillard PH, McIntyre RE, Gallagher
F, Kettunen MI, Lewis DY, Brindle K, Arends
MJ, Adams DJ, Patel KJ. Disruption of mouse
Slx4, a regulator of structure-specific nucleases,
phenocopies Fanconi anemia. Nat Genet. 2011; 43:
147-52
Day SE, Kettunen MI, Cherukuri MK, Mitchell JB,
Lizak MJ, Morris HD, Matsumoto S, Koretsky AP,
Brindle KM. Detecting response of rat C6 glioma
tumors to radiotherapy using hyperpolarized [1-13C]
pyruvate and 13C magnetic resonance spectroscopic
imaging. Magn Reson Med. 2011; 65: 557-63
Gallagher FA, Kettunen MI, Day SE, Hu DE,
Karlsson M, Gisselsson A, Lerche MH, Brindle
KM. Detection of tumor glutamate metabolism in
vivo using 13C magnetic resonance spectroscopy and
hyperpolarized [1-13C]glutamate. Magn Reson Med.
2011; 66: 18-23
Gallagher FA, Tay KY, Vowler SL, Szutowicz H,
Cross JJ, McAuley DJ, Antoun NM. Comparing the
accuracy of initial head CT reporting by radiologists,
radiology trainees, neuroradiographers and
emergency doctors. Br J Radiol. 2011; 84: 1040-5
Massie CE, Lynch A, Ramos-Montoya A, Boren
J, Stark R, Fazli L, Warren A, Scott H, Madhu
B, Sharma N, Bon H, Zecchini V, Smith DM,
Denicola GM, Mathews N, Osborne M, Hadfield
J, Macarthur S, Adryan B, Lyons SK, Brindle KM,
Griffiths J, Gleave ME, Rennie PS, Neal DE, Mills
IG. The androgen receptor fuels prostate cancer
by regulating central metabolism and biosynthesis.
E MBO J. 2011; 30: 2719-33
Neves AA, Stockmann H, Harmston RR, Pryor
HJ, Alam IS, Ireland-Zecchini H, Lewis DY, Lyons
SK, Leeper FJ, Brindle KM. Imaging sialylated tumor
cell glycans in vivo. FASEB J. 2011; 25: 2528-37
Sleigh A, Raymond-Barker P, Thackray K, Porter
D, Hatunic M, Vottero A, Burren C, Mitchell C,
McIntyre M, Brage S, Carpenter TA, Murgatroyd
PR, Brindle KM, Kemp GJ, O’Rahilly S, Semple
RK, Savage DB. Mitochondrial dysfunction in
patients with primary congenital insulin resistance. J
Clin Invest. 2011; 121: 2457-61
Stockmann H, Neves AA, Day HA, Stairs S,
Brindle KM, Leeper FJ. (E,E)-1,5-Cyclooctadiene:
a small and fast click-chemistry multitalent. Chem
Commun (Camb). 2011; 47: 7203-5
Stockmann H, Neves AA, Stairs S, Brindle KM,
Leeper FJ. Exploring isonitrile-based click chemistry
for ligation with biomolecules. Org Biomol Chem.
2011; 9: 7303-5
Stockmann H, Neves AA, Stairs S, IrelandZecchini H, Brindle KM, Leeper FJ. Development
and evaluation of new cyclooctynes for cell surface
glycan imaging in cancer cells. Chemical Science. 2011;
2: 932-6
Witney TH, Kettunen MI, Brindle KM. Kinetic
modeling of hyperpolarized 13C label exchange
between pyruvate and lactate in tumor cells. J Biol
Chem. 2011; 286: 24572-80
Other publications
Brindle KM, Bohndiek SE, Gallagher FA, Kettunen
MI. Tumor imaging using hyperpolarized 13C
magnetic resonance spectroscopy. Magn Reson Med.
2011; 66: 505-19
Gallagher FA, Bohndiek SE, Kettunen MI, Lewis
DY, Soloviev D, Brindle KM. Hyperpolarized 13C
MRI and PET: In vivo tumor biochemistry. J Nucl
Med. 2011; 52: 1333-6
Gallagher FA, Kettunen MI, Brindle KM. Imaging
pH with hyperpolarized 13C. NMR Biomed. 2011; 24:
1006-15
Kurhanewicz J, Vigneron DB, Brindle K,
Chekmenev EY, Comment A, Cunningham
CH, Deberardinis RJ, Green GG, Leach MO,
Rajan SS, Rizi RR, Ross BD, Warren WS, Malloy
CR. Analysis of cancer metabolism by imaging
hyperpolarized nuclei: prospects for translation to
clinical research. Neoplasia. 2011; 13: 81-97
Carlos Caldas (page 18)
Breast cancer functional genomic laboratory
Primary research papers
Abraham JE, Maranian M, Driver KE, Platte R,
Kalmyrzaev B, Baynes C, Luccarini C, Earl HM,
Dunning AM, Pharoah PD, Caldas C. CYP2D6
gene variants and their association with breast
cancer susceptibility. Cancer Epidemiol Biomarkers
Prev. 2011; 20: 1255-8
Ali AM, Dawson SJ, Blows FM, Provenzano E, Ellis
IO, Baglietto L, Huntsman D, Caldas C, Pharoah
PD. Comparison of methods for handling missing
data on immunohistochemical markers in survival
analysis of breast cancer. Br J Cancer. 2011; 104:
693-9
Ali HR, Dawson SJ, Blows FM, Provenzano E,
Pharoah PD, Caldas C. Cancer stem cell markers in
breast cancer: pathological, clinical and prognostic
significance. Breast Cancer Res. 2011; 13: R118
Broeks A, Schmidt MK, Sherman ME, Couch FJ,
Hopper JL, [and 122 others], Caldas C, Lubinski J,
Jakubowska A, Huzarski T, Byrski T, Cybulski C,
Gorski B, Gronwald J, Brennan P, Sangrajrang S,
Gaborieau V, Shen CY, Hsiung CN, Yu JC, Chen
ST, Hsu GC, Hou MF, Huang CS, Anton-Culver
H, Ziogas A, Pharoah PD, Garcia-Closas M. Low
penetrance breast cancer susceptibility loci are
associated with specific breast tumor subtypes:
Findings from the Breast Cancer Association
Consortium. Hum Mol Genet. 2011; 20: 3289-303
Goh XY, Rees JR, Paterson AL, Chin SF, Marioni
JC, Save V, O’Donovan M, Eijk PP, Alderson D,
Ylstra B, Caldas C, Fitzgerald RC. Integrative
analysis of array-comparative genomic hybridisation
and matched gene expression profiling data
reveals novel genes with prognostic significance in
oesophageal adenocarcinoma. Gut. 2011; 60: 131726
Habashy HO, Powe DG, Glaab E, Ball G, Spiteri
Research Publications | 73
I, Krasnogor N, Garibaldi JM, Rakha EA, Green
AR, Caldas C, Ellis IO. RERG (Ras-like, oestrogenregulated, growth-inhibitor) expression in breast
cancer: a marker of ER-positive luminal-like subtype.
Breast Cancer Res Treat. 2011; 128: 315-26
Holland DG, Burleigh A, Git A, Goldgraben MA,
Perez-Mancera PA, Chin SF, Hurtado A, Bruna A,
Ali HR, Greenwood W, Dunning MJ, Samarajiwa
S, Menon S, Rueda OM, Lynch AG, McKinney S,
Ellis IO, Eaves CJ, Carroll JS, Curtis C, Aparicio
S, Caldas C. ZNF703 is a common Luminal B
breast cancer oncogene that differentially regulates
luminal and basal progenitors in human mammary
epithelium. EMBO Mol Med. 2011; 3: 167-80
Storr SJ, Mohammed RA, Woolston CM, Green
AR, Parr T, Spiteri I, Caldas C, Ball GR, Ellis
IO, Martin SG. Calpastatin is associated with
lymphovascular invasion in breast cancer. Breast.
2011; 50: 413-8
Wishart GC, Bajdik CD, Azzato EM, Dicks E,
Greenberg DC, Rashbass J, Caldas C, Pharoah
PD. A population-based validation of the prognostic
model PREDICT for early breast cancer. Eur J Surg
Oncol. 2011; 37: 411-7
Yuan Y, Curtis C, Caldas C, Markowetz F. A
sparse regulatory network of copy-number driven
gene expression reveals putative breast cancer
oncogenes. IEEE/ACM Trans Comput Biol Bioinform.
2011; Epub Jul 20
Yuan Y, Rueda OM, Curtis C, Markowetz F.
Penalized regression elucidates aberration hotspots
mediating subtype-specific transcriptional responses
in breast cancer. Bioinformatics. 2011; 27: 2679-85
Other publications
Caldas C. Translational genomics in breast cancer.
Eur J Cancer. 2011; 47: S381-2
Vollan HK, Caldas C. The breast cancer genome –
a key for better oncology. BMC Cancer. 2011; 11: 501
breast cancer oncogene that differentially regulates
luminal and basal progenitors in human mammary
epithelium. EMBO Mol Med. 2011; 3: 167-80
Holmes KA, Hurtado A, Brown GD, Launchbury
R, Ross-Innes CS, Hadfield J, Odom DT, Carroll
JS. Breast Cancer Special Feature: Transducin-like
enhancer protein 1 mediates estrogen receptor
binding and transcriptional activity in breast cancer
cells. Proc Natl Acad Sci USA. 2011; Epub May 2
Krijgsman O, Roepman P, Zwart W, Carroll JS,
Tian S, de Snoo FA, Bender RA, Bernards R,
Glas AM. A diagnostic gene profile for molecular
subtyping of breast cancer associated with
treatment response. Breast Cancer Res Treat. 2011;
Epub Aug 4
Meyer KB, Maia AT, O’Reilly M, Ghoussaini M,
Prathalingam R, Porter-Gill P, Ambs S, ProkuninaOlsson L, Carroll J, Ponder BA. A functional
variant at a prostate cancer predisposition locus
at 8q24 is associated with PVT1 expression. PLoS
Genet. 2011; 7: e1002165
Robinson JL, Macarthur S, Ross-Innes CS, Tilley
WD, Neal DE, Mills IG, Carroll JS. Androgen
receptor driven transcription in molecular apocrine
breast cancer is mediated by FoxA1. EMBO J. 2011;
30: 3019-27
Ross-Innes CS, Brown GD, Carroll JS. A coordinated interaction between CTCF and ER in
breast cancer cells. BMC Genomics. 2011; 12: 593
Zwart W, Theodorou V, Kok M, Canisius S,
Linn S, Carroll JS. Oestrogen receptor-cofactor-chromatin specificity in the transcriptional
regulation of breast cancer. EMBO J. 2011; 30: 476476
Other publications
Zaret KS, Carroll JS. Pioneer transcription factors:
establishing competence for gene expression. Genes
Dev. 2011; 25: 2227-41
Douglas Fearon (page 22)
Cancer and immunology laboratory
Primary research papers
Yeo CJ, Fearon DT. T-bet-mediated differentiation
of the activated CD8+ T cell. Eur J Immunol. 2011;
41: 60-6
Fanni Gergely (page 24)
Jason Carroll (page 20)
Nuclear receptor transcription laboratory
Primary research papers
Holland DG, Burleigh A, Git A, Goldgraben MA,
Perez-Mancera PA, Chin SF, Hurtado A, Bruna A,
Ali HR, Greenwood W, Dunning MJ, Samarajiwa
S, Menon S, Rueda OM, Lynch AG, McKinney S,
Ellis IO, Eaves CJ, Carroll JS, Curtis C, Aparicio
S, Caldas C. ZNF703 is a common Luminal B
74 | Cambridge Research Institute Scientific Report 2011
Centrosomes, microtubules and cancer laboratory
Primary research papers
Bakircioglu M, Carvalho OP, Khurshid M, Cox JJ,
Tuysuz B, Barak T, Yilmaz S, Caglayan O, Dincer
A, Nicholas AK, Quarrell O, Springell K, Karbani
G, Malik S, Gannon C, Sheridan E, Crosier M,
Lisgo SN, Lindsay S, Bilguvar K, Gergely F, Gunel
M, Woods CG. The essential role of centrosomal
NDE1 in human cerebral cortex neurogenesis. Am J
Hum Genet. 2011; 88: 523-35
Sir JH, Barr AR, Nicholas AK, Carvalho OP,
Khurshid M, Sossick A, Reichelt S, D’Santos C,
Woods CG, Gergely F. A primary microcephaly
protein complex forms a ring around parental
centrioles. Nat Genet. 2011; 43: 1147-53
Zyss D, Ebrahimi H, Gergely F. Casein kinase I
delta controls centrosome positioning during T cell
activation. J Cell Biol. 2011; 195: 781-97
John Griffiths (page 26)
Magnetic resonance imaging and spectroscopy (MRI
and MRS) laboratory
Primary research papers
Bapiro TE, Richards FM, Goldgraben MA, Olive
KP, Madhu B, Frese KK, Cook N, Jacobetz MA,
Smith DM, Tuveson DA, Griffiths JR, Jodrell DI.
A novel method for quantification of gemcitabine
and its metabolites 2′,2′-difluorodeoxyuridine and
gemcitabine triphosphate in tumour tissue by LCMS/MS: comparison with 19F NMR spectroscopy.
Cancer Chemother Pharmacol. 2011; 68: 1243-53
Barrett T, Gill AB, Kataoka MY, Priest AN,
Joubert I, McLean MA, Graves MJ, Stearn S,
Lomas DJ, Griffiths JR, Neal D, Gnanapragasam
VJ, Sala E. DCE and DW MRI in monitoring
response to androgen deprivation therapy in
patients with prostate cancer: A feasibility study.
Magn Reson Med. 2011; Epub Aug 29
Beloueche-Babari M, Arunan V, Troy H, Te Poele
RH, Wong Te Fong AC, Jackson LE, Payne GS,
Griffiths JR, Judson IR, Workman P, Leach MO,
Chung YL. Histone deacetylase inhibition increases
levels of choline kinase alpha and phosphocholine
facilitating non-invasive imaging in human cancers.
Cancer Res. 2011; Epub Dec 22
Candiota AP, Majos C, Julia-Sape M, Cabanas M,
Acebes JJ, Moreno-Torres A, Griffiths J, Arus C.
Non-invasive grading of astrocytic tumours from
the relative contents of myo-inositol and glycine
measured by in vivo MRS. JBR-BTR. 2011; 94: 319-29
Fuster-Garcia E, Navarro C, Vicente J, Tortajada
S, Garcia-Gomez JM, Saez C, Calvar J, Griffiths J,
et al. Compatibility between 3T 1H SV-MRS data
and automatic brain tumour diagnosis support
systems based on databases of 1.5T 1H SV-MRS
spectra. MAGMA. 2011; 24: 35-42
Golinska M, Troy H, Chung YL, McSheehy PM,
Mayr M, Yin X, Ly L, Williams KJ, Airley RE,
Harris AL, Latigo J, Perumal M, Aboagye EO,
Perrett D, Stubbs M, Griffiths JR. Adaptation
to HIF-1 deficiency by upregulation of the AMP/
ATP ratio and phosphofructokinase activation in
hepatomas. BMC Cancer. 2011; 11: 198
Julia-Sape M, Coronel I, Majos C, Candiota
AP, Serrallonga M, Cos M, Aguilera C, Acebes
JJ, Griffiths JR, Arus C. Prospective diagnostic
performance evaluation of single-voxel 1H MRS for
typing and grading of brain tumours. NMR Biomed.
2011; Epub Sep 23
Massie CE, Lynch A, Ramos-Montoya A, Boren
J, Stark R, Fazli L, Warren A, Scott H, Madhu
B, Sharma N, Bon H, Zecchini V, Smith DM,
Denicola GM, Mathews N, Osborne M, Hadfield
J, Macarthur S, Adryan B, Lyons SK, Brindle KM,
Griffiths J, Gleave ME, Rennie PS, Neal DE, Mills
IG. The androgen receptor fuels prostate cancer
by regulating central metabolism and biosynthesis.
E MBO J. 2011; 30: 2719-33
Mayr M, May D, Oren G, Madhu B, Gilon D, Yin
X, Xing Q, Drozdov I, Ainali C, Tsoka S, Xu Q,
Griffiths J, Horrevoets A, Keshet E. Metabolic
homeostasis is maintained in myocardial hibernation
by adaptive changes in the transcriptome and
proteome. J Mol Cell Cardiol. 2011; 506: 982-90
McLean MA, Barrett T, Gnanapragasam VJ, Priest
AN, Joubert I, Lomas DJ, Neal DE, Griffiths JR,
Sala E. Prostate cancer metabolite quantification
relative to water in 1H-MRSI in vivo at 3 Tesla. Magn
Reson Med. 2011; 65: 914-9
Tortajada S, Fuster-Garcia E, Vicente J, Wesseling
P, Howe FA, Julia-Sape M, Candiota AP, Monleon
D, Moreno-Torres A, Pujol J, Griffiths JR, et al.
Incremental Gaussian Discriminant Analysis based
on Graybill and Deal weighted combination of
estimators for brain tumour diagnosis. J Biomed
Inform. 2011; 44: 677-87
Other publications
Bell LK, Ainsworth NL, Lee SH, Griffiths JR.
MRI & MRS assessment of the role of the tumour
microenvironment in response to therapy. NMR
Biomed. 2011; 24: 612-35
Griffiths J. Editorial: Farewell and welcome. NMR
Biomed. 2011; 24: 113
Duncan Jodrell (page 28)
Pharmacology and drug development group
Primary research papers
Adams RA, Meade AM, Seymour MT, Wilson RH,
Madi A, Fisher D, Kenny SL, Kay E, Hodgkinson
E, Pope M, Rogers P, Wasan H, Falk S, Gollins
S, Hickish T, Bessell EM, Propper D, Kennedy
MJ, Kaplan R, Maughan TS. Intermittent versus
continuous oxaliplatin and fluoropyrimidine
combination chemotherapy for first-line treatment
of advanced colorectal cancer: results of the
randomised phase 3 MRC COIN trial. Lancet Oncol.
2011; 12: 642-53
Bapiro TE, Richards FM, Goldgraben MA, Olive
KP, Madhu B, Frese KK, Cook N, Jacobetz MA,
Smith DM, Tuveson DA, Griffiths JR, Jodrell DI.
A novel method for quantification of gemcitabine
and its metabolites 2′,2′-difluorodeoxyuridine and
gemcitabine triphosphate in tumour tissue by LCMS/MS: comparison with 19F NMR spectroscopy.
Cancer Chemother Pharmacol. 2011; 68: 1243-53
Courtin A, Communal L, Vilasco M, Cimino D,
Mourra N, de Bortoli M, Taverna D, Faussat AM,
Chaouat M, Forgez P, Gompel A. Glucocorticoid
receptor activity discriminates between
progesterone and medroxyprogesterone acetate
effects in breast cells. Breast Cancer Res Treat. 2011;
131: 49-63
Lin Y, Henderson P, Pettersson S, Satsangi J,
Hupp T, Stevens C. Tuberous sclerosis-2 (TSC2)
regulates the stability of death-associated protein
kinase-1 (DAPK) through a lysosome-dependent
degradation pathway. Febs J. 2011; 278: 354-70
Maughan TS, Adams RA, Smith CG, Meade AM,
Seymour MT, Wilson RH, Idziaszczyk S, Harris
R, Fisher D, Kenny SL, Kay E, Mitchell JK, Madi
A, Jasani B, James MD, Bridgewater J, Kennedy
Research Publications | 75
MJ, Claes B, Lambrechts D, Kaplan R, Cheadle
JP. Addition of cetuximab to oxaliplatin-based
first-line combination chemotherapy for treatment
of advanced colorectal cancer: results of the
randomised phase 3 MRC COIN trial. Lancet. 2011;
377: 2103-14
Rudman SM, Jameson MB, McKeage MJ, Savage
P, Jodrell DI, Harries M, Acton G, Erlandsson
F, Spicer JF. A phase 1 study of AS1409, a novel
antibody-cytokine fusion protein, in patients with
malignant melanoma or renal cell carcinoma. Clin
Cancer Res. 2011; 17: 1998-2005
Vilasco M, Communal L, Mourra N, Courtin A,
Forgez P, Gompel A. Glucocorticoid receptor and
breast cancer. Breast Cancer Res Treat. 2011; 130:
1-10
Other publications
Harrington JA, Jones RJ. Management of
metastatic castration-resistant prostate cancer after
first-line docetaxel. Eur J Cancer. 2011; 47: 2133-42
Florian Markowetz (page 30)
Computational biology laboratory
Primary research papers
Altay G, Asim M, Markowetz F, Neal DE.
Differential C3NET reveals disease networks of
direct physical interactions. BMC Bioinformatics.
2011; 12: 296
Szczurek E, Markowetz F, Gat-Viks I, Biecek P,
Tiuryn J, Vingron M. Deregulation upon DNA
damage revealed by joint analysis of context-specific
perturbation data. BMC Bioinformatics. 2011; 12: 249
Wang X, Terfve C, Rose JC, Markowetz F.
HTSanalyzeR: an R/Bioconductor package for
integrated network analysis of high-throughput
screens. Bioinformatics. 2011; 27: 879-80
Yuan Y, Curtis C, Caldas C, Markowetz F. A
sparse regulatory network of copy-number driven
gene expression reveals putative breast cancer
oncogenes. IEEE/ACM Trans Comput Biol Bioinform.
2011; Epub Jul 20
Yuan Y, Rueda OM, Curtis C, Markowetz F.
Penalized regression elucidates aberration hotspots
mediating subtype-specific transcriptional responses
in breast cancer. Bioinformatics. 2011; 27: 2679-85
Yuan Y, Savage RS, Markowetz F. Patient-specific
data fusion defines prognostic cancer subtypes.
PLoS Comput Biol. 2011; 7: e1002227
Gillian Murphy (page 32)
Proteases and the tumour microenvironment
laboratory
Primary research papers
Lawson MH, Cummings NM, Rassl DM, Russell
R, Brenton JD, Rintoul RC, Murphy G. Two novel
determinants of etoposide resistance in small cell
lung cancer. Cancer Res. 2011; 71: 4877-87
McGinn OJ, English WR, Roberts S, Ager A,
Newham P, Murphy G. Modulation of integrin α4β1
by ADAM28 promotes lymphocyte adhesion and
transendothelial migration. Cell Biol Int. 2011; 35:
1043-53
76 | Cambridge Research Institute Scientific Report 2011
Nunes GL, Simoes A, Dyszy FH, Shida CS, Juliano
MA, Juliano L, Gesteira TF, Nader HB, Murphy
G, Chaffotte AF, Goldberg ME, Tersariol IL,
Almeida PC. Mechanism of heparin acceleration
of tissue inhibitor of metalloproteases-1 (TIMP-1)
degradation by the human neutrophil elastase. PLoS
ONE. 2011; 6: e21525
Tape CJ, Willems SH, Dombernowsky SL, Stanley
PL, Fogarasi M, Ouwehand W, McCafferty J,
Murphy G. Cross-domain inhibition of TACE
ectodomain. Proc Natl Acad Sci USA. 2011; 108:
5578-83
Winder DM, Chattopadhyay A, Muralidhar B,
Bauer J, English WR, Zhang X, Karagavriilidou
K, Roberts I, Pett MR, Murphy G, Coleman N.
Overexpression of the oncostatin M receptor in
cervical squamous cell carcinoma cells is associated
with a pro-angiogenic phenotype and increased cell
motility and invasiveness. J Pathol. 2011; 225: 448-62
Yan D, Chen D, Cool SM, van Wijnen AJ, Mikecz
K, Murphy G, Im HJ. Fibroblast growth factor
receptor 1 is principally responsible for fibroblast
growth factor 2-induced catabolic activities in
human articular chondrocytes. Arthritis Res Ther.
2011; 13: R130
Other publications
Murphy G. Tissue inhibitors of metalloproteinases.
Genome Biol. 2011; 12: 233
Murphy G, Nagase H. Localizing matrix
metalloproteinase activities in the pericellular
environment. Febs J. 2011; 278: 2-15
Adele Murrell (page 34)
Epigenetics and imprinting laboratory
Primary research papers
Huntriss J, Woodfine K, Huddleston JE, Murrell A,
Rutherford AJ, Elder K, Khan AA, Hemmings K,
Picton H. Quantitative analysis of DNA methylation
of imprinted genes in single human blastocysts by
pyrosequencing. Fertil Steril. 2011; 95: 2564-7
Nativio R, Sparago A, Ito Y, Weksberg R,
Riccio A, Murrell A. Disruption of genomic
neighbourhood at the imprinted IGF2-H19 locus in
Beckwith-Wiedemann syndrome and Silver-Russell
syndrome. Hum Mol Genet. 2011; 20: 1363-74
Sandovici I, Smith NH, Nitert MD, AckersJohnson M, Uribe-Lewis S, Ito Y, Jones RH,
Marquez VE, Cairns W, Tadayyon M, O’Neill LP,
Murrell A, Ling C, Constancia M, Ozanne SE.
Maternal diet and aging alter the epigenetic control
of a promoter-enhancer interaction at the Hnf4a
gene in rat pancreatic islets. Proc Natl Acad Sci USA.
2011; 108: 5449-54
Sun B, Ito M, Mendjan S, Ito Y, Brons IG, Murrell
A, Vallier L, Ferguson-Smith AC, Pedersen RA.
Status of genomic imprinting in epigenetically
distinct pluripotent stem cells. Stem Cells. 2011; 30:
161-8
Woodfine K, Huddleston JE, Murrell A.
Quantitative analysis of DNA methylation at all
human imprinted regions reveals preservation
of epigenetic stability in adult somatic tissue.
Epigenetics Chromatin. 2011; 4: 1
Other publications
Murrell A. Setting up and maintaining differential
insulators and boundaries for genomic imprinting.
Biochem Cell Biol. 2011; 89: 469-78
Uribe-Lewis S, Woodfine K, Stojic L, Murrell A.
Molecular mechanisms of genomic imprinting and
clinical implications for cancer. Expert Rev Mol Med.
2011; 13: e2
Masashi Narita (page 36)
Mechanisms of cellular senescence laboratory
Primary research papers
Al-Mayhani MT, Grenfell R, Narita M, Piccirillo
S, Kenney-Herbert E, Fawcett JW, Collins
VP, Ichimura K, Watts C. NG2 expression in
glioblastoma identifies an actively proliferating
population with an aggressive molecular signature.
Neuro Oncol. 2011; 13: 830-45
Bergboer JG, Tjabringa GS, Kamsteeg M, van
Vlijmen-Willems IM, Rodijk-Olthuis D, Jansen
PA, Thuret JY, Narita M, Ishida-Yamamoto A,
Zeeuwen PL, Schalkwijk J. Psoriasis risk genes of
the late cornified envelope-3 group are distinctly
expressed compared with genes of other LCE
groups. Am J Pathol. 2011; 178: 1470-7
Narita M, Young AR, Arakawa S, Samarajiwa
SA, Nakashima T, Yoshida S, Hong S, Berry
LS, Reichelt S, Ferreira M, Tavaré S, Inoki K,
Shimizu S, Narita M. Spatial coupling of mTOR and
autophagy augments secretory phenotypes. Science.
2011; 332: 966-70
Wu J, Liu Z, Shao C, Gong Y, Hernando E, Lee
P, Narita M, Muller W, Liu J, Wei JJ. HMGA2
overexpression-induced ovarian surface epithelial
transformation is mediated through regulation of
EMT genes. Cancer Res. 2011; 71: 349-59
Other publications
Hoare M, Young AR, Narita M. Autophagy in
cancer: Having your cake and eating it. Semin Cancer
Biol. 2011; 21: 397-404
Sikora E, Arendt T, Bennett M, Narita M. Impact
of cellular senescence signature on ageing research.
Ageing Research Reviews. 2011; 10: 146-52
Young AR, Narita M, Narita M. Spatio-temporal
association between mTOR and autophagy during
cellular senescence. Autophagy. 2011; 7: 1387-8
David Neal (page 38)
Prostate research laboratory
Primary research papers
Altay G, Asim M, Markowetz F, Neal DE.
Differential C3NET reveals disease networks of
direct physical interactions. BMC Bioinformatics.
2011; 12: 296
Barbiere JM, Saeb-Parsy K, Greenberg DC,
Wright KA, Brown CH, Neal DE, Lyratzopoulos
G. Trends in the use of radiotherapy and radical
surgery for patients with bladder urothelial cell
carcinoma in East Anglia, 1995-2006. BJU Int. 2011;
108: 1106-14
Barrett T, Gill AB, Kataoka MY, Priest AN,
Joubert I, McLean MA, Graves MJ, Stearn S,
Lomas DJ, Griffiths JR, Neal D, Gnanapragasam
VJ, Sala E. DCE and DW MRI in monitoring
response to androgen deprivation therapy in
patients with prostate cancer: A feasibility study.
Magn Reson Med. 2011; Epub Aug 29
Batra J, Lose F, O’Mara T, Marquart L, Stephens
C, Alexander K, Srinivasan S, Eeles RA, Easton
DF, Olama AA, Kote-Jarai Z, Guy M, Muir K,
Lophatananon A, Rahman AA, Neal DE, et al.
Association between prostinogen (KLK15) genetic
variants and prostate cancer risk and aggressiveness
in Australia and a meta-analysis of GWAS data.
PLoS ONE. 2011; 6: e26527
Burton AJG, Martin RM, Holly JM, Hamdy FC,
Neal DE, Donovan JL, Tilling KM. Association
of anthropometric and lifestyle factors with
prostate specific antigen (Psa) trajectories in men
with localised prostate cancer undergoing active
monitoring. Eur Urol Suppl. 2011; 10: 237
Cho HS, Kelly JD, Hayami S, Toyokawa G,
Takawa M, Yoshimatsu M, Tsunoda T, Field HI,
Neal DE, Ponder BA, Nakamura Y, Hamamoto
R. Enhanced expression of EHMT2 is involved in
the proliferation of cancer cells through negative
regulation of SIAH1. Neoplasia. 2011; 13: 676-84
Cho HS, Toyokawa G, Daigo Y, Hayami S, Masuda
K, Ikawa N, Yamane Y, Maejima K, Tsunoda T,
Field HI, Kelly JD, Neal DE, Ponder BA, Maehara
Y, Nakamura Y, Hamamoto R. The JmjC domaincontaining histone demethylase KDM3A is a
positive regulator of the G1/S transition in cancer
cells via transcriptional regulation of the HOXA1
gene. Int J Cancer. 2011; Epub Oct 23
Collin SM, Metcalfe C, Palmer TM, Refsum H,
Lewis SJ, Smith GD, Cox A, Davis M, Marsden
G, Johnston C, Lane JA, Donovan JL, Neal DE,
Hamdy FC, Smith AD, Martin RM. The causal
roles of vitamin B12 and transcobalamin in prostate
cancer: can Mendelian randomization analysis
provide definitive answers? Int J Mol Epidemiol
Genet. 2011; 2: 316-27
Dev H, Sharma NL, Dawson SN, Neal DE, Shah
N. Detailed analysis of operating time learning
curves in robotic prostatectomy by a novice
surgeon. BJU Int. 2011; Epub Oct 28
Dimitropoulou P, Martin RM, Turner EL, Lane JA,
Gilbert R, Davis M, Donovan JL, Hamdy FC, Neal
DE. Association of obesity with prostate cancer:
Research Publications | 77
a case-control study within the population-based
PSA testing phase of the ProtecT study. Br J Cancer.
2011; 104: 875-81
Down L, Metcalfe C, Martin RM, Neal DE, Hamdy
FC, Donovan JL, Lane JA. Seasonal variation
in prostate-specific antigen levels: a large crosssectional study of men in the UK. BJU Int. 2011; 108:
1409-14
Gilbert R, Metcalfe C, Fraser WD, Donovan
J, Hamdy F, Neal DE, Lane JA, Martin RM.
Associations of circulating 25-hydroxyvitamin D
with prostate cancer diagnosis, stage and grade. Int J
Cancer. 2011; Epub Oct 27
Kote-Jarai Z, Amin Al Olama A, Leongamornlert
D, Tymrakiewicz M, Saunders E, Guy M, Giles
GG, Severi G, Southey M, Hopper JL, Sit KC,
Harris JM, Batra J, Spurdle AB, Clements JA,
Hamdy F, Neal D, et al. Identification of a novel
prostate cancer susceptibility variant in the KLK3
gene transcript. Hum Genet. 2011; 129: 687-94
Kote-Jarai Z, Olama AA, Giles GG, Severi G,
Schleutker J, Weischer M, Campa D, Riboli E,
Key T, Gronberg H, Hunter DJ, Kraft P, Thun MJ,
Ingles S, Chanock S, Albanes D, Hayes RB, Neal
DE, et al. Seven prostate cancer susceptibility loci
identified by a multi-stage genome-wide association
study. Nat Genet. 2011; 43: 785-91
Lamb AD, Qadan M, Roberts S, Timlin H, Vowler
SL, Campbell FM, Grigor K, Bartlett JM, McNeill
SA. CD4+ and CD8+ T-lymphocyte scores cannot
reliably predict progression in patients with benign
prostatic hyperplasia. BJU Int. 2011; 108: E43-50
Lamb AD, Vowler SL, Johnston R, Dunn N,
Wiseman OJ. Meta-analysis showing the beneficial
effect of alpha-blockers on ureteric stent
discomfort. BJU Int. 2011; 108: 1894-902
Lane JA, Wade J, Down L, Bonnington S, Holding
PN, Lennon T, Jones AJ, Elizabeth Salter C, Neal
DE, Hamdy FC, Donovan JL. A Peer Review
Intervention for Monitoring and Evaluating sites
(PRIME) that improved randomized controlled trial
conduct and performance. J Clin Epidemiol. 2011; 64:
628-36
Lose F, Batra J, O’Mara T, Fahey P, Marquart L,
Eeles RA, Easton DF, Al Olama AA, Kote-Jarai
Z, Guy M, Muir K, Lophatananon A, Rahman AA,
Neal DE, et al. Common variation in Kallikrein
genes KLK5, KLK6, KLK12, and KLK13 and risk of
prostate cancer and tumor aggressiveness. Urol
Oncol. 2011; Epub Jul 7
Madden T, Doble A, Aliyu SH, Neal DE. Infective
complications after transrectal ultrasound-guided
prostate biopsy following a new protocol for
antibiotic prophylaxis aimed at reducing hospitalacquired infections. BJU Int. 2011; 108: 1597-602
Massie CE, Lynch A, Ramos-Montoya A, Boren
J, Stark R, Fazli L, Warren A, Scott H, Madhu
B, Sharma N, Bon H, Zecchini V, Smith DM,
Denicola GM, Mathews N, Osborne M, Hadfield
J, Macarthur S, Adryan B, Lyons SK, Brindle KM,
Griffiths J, Gleave ME, Rennie PS, Neal DE, Mills
IG. The androgen receptor fuels prostate cancer
by regulating central metabolism and biosynthesis.
78 | Cambridge Research Institute Scientific Report 2011
MBO J. 2011; 30: 2719-33
E
McLean MA, Barrett T, Gnanapragasam VJ, Priest
AN, Joubert I, Lomas DJ, Neal DE, Griffiths JR,
Sala E. Prostate cancer metabolite quantification
relative to water in 1H-MRSI in vivo at 3 Tesla. Magn
Reson Med. 2011; 65: 914-9
Mills N, Donovan JL, Wade J, Hamdy FC, Neal
DE, Lane JA. Exploring treatment preferences
facilitated recruitment to randomized controlled
trials. J Clin Epidemiol. 2011; 64: 1127-36
Morgan R, Boxall A, Bhatt A, Bailey M, Hindley
R, Langley S, Whitaker HC, Neal DE, Ismail
M, Whitaker H, Annels N, Michael A, Pandha
H. Engrailed-2 (EN2): A tumor specific urinary
biomarker for the early diagnosis of prostate cancer.
Clin Cancer Res. 2011; 17: 1090-8
Pashayan N, Duffy SW, Chowdhury S, Dent T,
Burton H, Neal DE, Easton DF, Eeles R, Pharoah
P. Polygenic susceptibility to prostate and breast
cancer: implications for personalised screening. Br J
Cancer. 2011; 104: 1656-63
Robinson JL, Macarthur S, Ross-Innes CS, Tilley
WD, Neal DE, Mills IG, Carroll JS. Androgen
receptor driven transcription in molecular apocrine
breast cancer is mediated by FoxA1. EMBO J. 2011;
30: 3019-27
Rowlands MA, Holly JM, Gunnell D, Donovan
JL, Lane JA, Hamdy F, Neal DE, Oliver S, Davey
Smith G, Martin RM. Circulating insulin-like growth
factors (IGFs) and IGF binding proteins (IGFBPs)
in PSA-detected prostate cancer: the large case
control study ProtecT. Cancer Res. 2011; Epub Nov
21
Rowlands MA, Holly JM, Hamdy F, Phillips J,
Goodwin L, Marsden G, Gunnell D, Donovan J,
Neal DE, Martin RM. Serum insulin-like growth
factors and mortality in localised and advanced
clinically detected prostate cancer. Cancer Causes
Control. 2011; Epub Dec 20
Rowlands MAE, Holly JMP, Gunnell D, Donovan J,
Lane JA, Hamdy F, Neal DE, Oliver SE, Smith GD,
Martin RM. Insulin-like growth factors (Igfs) and Igf
binding proteins in Psa-detected prostate cancer: A
large population-based case control study (Protect).
Eur Urol Suppl. 2011; 10: 208
Schumacher FR, Berndt SI, Siddiq A, Jacobs KB,
Wang Z, Lindstrom S, Stevens VL, Chen C,
Mondul AM, Travis RC, Stram DO, Eeles RA,
Easton DF, Giles G, Hopper JL, Neal DE, et al.
Genome-wide association study identifies new
prostate cancer susceptibility loci. Hum Mol Genet.
2011; 20: 3867-75
Stacey SN, Sulem P, Jonasdottir A, Masson
G, Gudmundsson J, [and 83 others], Neal
DE, Catalona WJ, Wrensch M, Wiencke J,
Jenkins RB, Nagore E, Vogel U, Kiemeney LA,
Kumar R, Mayordomo JI, Olafsson JH, Kong A,
Thorsteinsdottir U, Rafnar T, Stefansson K. A
germline variant in the TP53 polyadenylation signal
confers cancer susceptibility. Nat Genet. 2011; 43:
1098-103
Toyokawa G, Cho HS, Iwai Y, Yoshimatsu M,
Takawa M, Hayami S, Maejima K, Shimizu N,
Tanaka H, Tsunoda T, Field H, Kelly JD, Neal DE,
Ponder BA, Maehara Y, Nakamura Y, Hamamoto
R. The histone demethylase JMJD2B plays an
essential role in human carcinogenesis through
positive regulation of cyclin-dependent kinase 6.
Cancer Prev Res (Phila). 2011; 4: 2051-61
Toyokawa G, Cho HS, Masuda K, Yamane Y,
Yoshimatsu M, Hayami S, Takawa M, Iwai Y,
Daigo Y, Tsuchiya E, Tsunoda T, Field HI, Kelly
JD, Neal DE, Maehara Y, Ponder BA, Nakamura
Y, Hamamoto R. Histone lysine methyltransferase
Wolf-Hirschhorn syndrome candidate 1 is involved
in human carcinogenesis through regulation of the
Wnt pathway. Neoplasia. 2011; 13: 887-98
Toyokawa G, Masuda K, Daigo Y, Cho HS,
Yoshimatsu M, Takawa M, Hayami S, Maejima K,
Chino M, Field HI, Neal DE, Tsuchiya E, Ponder
BA, Maehara Y, Nakamura Y, Hamamoto R.
Minichromosome Maintenance Protein 7 is a
potential therapeutic target in human cancer and
a novel prognostic marker of non-small cell lung
cancer. Mol Cancer. 2011; 10: 65
Williams N, Hughes LJ, Turner EL, Donovan JL,
Hamdy FC, Neal DE, Martin RM, Metcalfe C.
Prostate-specific antigen testing rates remain low in
UK general practice: a cross-sectional study in six
English cities. BJU Int. 2011; 108: 1402-8
Wong LM, Johnston R, Sharma N, Shah NC,
Warren AY, Neal DE. General application of
the National Institute for Health and Clinical
Excellence (NICE) guidance for active surveillance
for men with prostate cancer is not appropriate in
unscreened populations. BJU Int. 2011; Epub Nov 11
Other publications
Gnanapragasam VJ, Mason MD, Shaw GL, Neal
DE. The role of surgery in high-risk localised
prostate cancer. BJU Int. 2011; Epub Sep 27
Neal DE, Shah NC, Gnanpragasam VJ, Pati V.
Randomised surgical trials need good surgical
outcomes in the control arm. BMJ. 2011; 343: d7520
Duncan Odom (page 40)
Regulatory systems biology laboratory
Primary research papers
Holmes KA, Hurtado A, Brown GD, Launchbury
R, Ross-Innes CS, Hadfield J, Odom DT, Carroll
JS. Breast Cancer Special Feature: Transducin-like
enhancer protein 1 mediates estrogen receptor
binding and transcriptional activity in breast cancer
cells. Proc Natl Acad Sci USA. 2011; Epub May 2
Ip JY, Schmidt D, Pan Q, Ramani AK, Fraser AG,
Odom DT, Blencowe BJ. Global impact of RNA
polymerase II elongation inhibition on alternative
splicing regulation. Genome Research. 2011; 21: 390401
Kutter C, Brown GD, Goncalves A, Wilson MD,
Watt S, Brazma A, White RJ, Odom DT. Pol III
binding in six mammals shows conservation among
amino acid isotypes despite divergence among
tRNA genes. Nat Genet. 2011; 43: 948-55
Laudadio I, Manfroid I, Achouri Y, Schmidt D,
Wilson MD, Cordi S, Thorrez L, Knoops L,
Jacquemin P, Schuit F, Pierreux CE, Odom DT,
Peers B, Lemaigre FP. A feedback loop between
the liver-enriched transcription factor network
and Mir-122 controls hepatocyte differentiation.
Gastroenterology. 2011; Epub Sep 12
Nascimento EM, Cox CL, Macarthur S, Hussain
S, Trotter M, Blanco S, Suraj M, Nichols J, Kubler
B, Benitah SA, Hendrich B, Odom DT, Frye M.
The opposing transcriptional functions of Sin3a and
c-Myc are required to maintain tissue homeostasis.
Nat Cell Biol. 2011; 13: 1395-405
Other publications
Odom DT. Identification of transcription factorDNA interactions in vivo. Subcell Biochem. 2011; 52:
175-91
Bruce Ponder (page 42)
Genetic susceptibility to cancer laboratory
Primary research papers
Cho HS, Kelly JD, Hayami S, Toyokawa G,
Takawa M, Yoshimatsu M, Tsunoda T, Field HI,
Neal DE, Ponder BA, Nakamura Y, Hamamoto
R. Enhanced expression of EHMT2 is involved in
the proliferation of cancer cells through negative
regulation of SIAH1. Neoplasia. 2011; 13: 676-84
Cho HS, Toyokawa G, Daigo Y, Hayami S, Masuda
K, Ikawa N, Yamane Y, Maejima K, Tsunoda T,
Field HI, Kelly JD, Neal DE, Ponder BA, Maehara
Y, Nakamura Y, Hamamoto R. The JmjC domaincontaining histone demethylase KDM3A is a
positive regulator of the G1/S transition in cancer
cells via transcriptional regulation of the HOXA1
gene. Int J Cancer. 2011; Epub Oct 23
Meyer KB, Maia AT, O’Reilly M, Ghoussaini M,
Prathalingam R, Porter-Gill P, Ambs S, ProkuninaOlsson L, Carroll J, Ponder BA. A functional
variant at a prostate cancer predisposition locus
at 8q24 is associated with PVT1 expression. PLoS
Genet. 2011; 7: e1002165
Takawa M, Masuda K, Kunizaki M, Daigo Y,
Takagi K, Iwai Y, Cho HS, Toyokawa G, Yamane
Y, Maejima K, Field HI, Kobayashi T, Akasu T,
Sugiyama M, Tsuchiya E, Atomi Y, Ponder BA,
Nakamura Y, Hamamoto R. Validation of the
histone methyltransferase EZH2 as a therapeutic
target for various types of human cancer and as a
prognostic marker. Cancer Sci. 2011; 102: 1298-305
Toyokawa G, Cho HS, Iwai Y, Yoshimatsu M,
Takawa M, Hayami S, Maejima K, Shimizu N,
Tanaka H, Tsunoda T, Field H, Kelly JD, Neal DE,
Ponder BA, Maehara Y, Nakamura Y, Hamamoto
R. The histone demethylase JMJD2B plays an
essential role in human carcinogenesis through
positive regulation of cyclin-dependent kinase 6.
Cancer Prev Res (Phila). 2011; 4: 2051-61
Toyokawa G, Cho HS, Masuda K, Yamane Y,
Yoshimatsu M, Hayami S, Takawa M, Iwai Y,
Daigo Y, Tsuchiya E, Tsunoda T, Field HI, Kelly
JD, Neal DE, Maehara Y, Ponder BA, Nakamura
Y, Hamamoto R. Histone lysine methyltransferase
Wolf-Hirschhorn syndrome candidate 1 is involved
in human carcinogenesis through regulation of the
Wnt pathway. Neoplasia. 2011; 13: 887-98
Toyokawa G, Masuda K, Daigo Y, Cho HS,
Research Publications | 79
Yoshimatsu M, Takawa M, Hayami S, Maejima K,
Chino M, Field HI, Neal DE, Tsuchiya E, Ponder
BA, Maehara Y, Nakamura Y, Hamamoto R.
Minichromosome Maintenance Protein 7 is a
potential therapeutic target in human cancer and
a novel prognostic marker of non-small cell lung
cancer. Mol Cancer. 2011; 10: 65
John Stingl (page 46)
Mammary stem cell biology laboratory
Other publications
Del Mar Vivanco M, Stingl J, Clarke RB, BentiresAlj M. The devil is in the methods: lineage tracing,
functional screens and sequencing, hormones,
tumour-stroma interactions, and expansion of
human breast tumours as xenografts. Breast Cancer
Res. 2011; 13: 316
Stingl J. Estrogen and progesterone in normal
mammary gland development and in cancer. Horm
Cancer. 2011; 2: 85-90
Simon Tavaré (page 48)
Computational biology and statistics laboratory
Primary research papers
Cairns J, Spyrou C, Stark R, Smith ML, Lynch AG,
Tavaré S. BayesPeak - An R package for analysing
ChIP-seq data. Bioinformatics. 2011; 27: 713-4
Halper-Stromberg E, Frelin L, Ruczinski I, Scharpf
R, Jie CF, Carvalho B, Hao HP, Hetrick K, Jedlicka
A, Dziedzic A, Doheny K, Scott AF, Baylin S,
Pevsner J, Spencer F, Irizarry RA. Performance
assessment of copy number microarray platforms
using a spike-in experiment. Bioinformatics. 2011; 27:
1052-60
Harman AN, Lai J, Turville S, Samarajiwa S, Gray
L, Marsden V, Mercier SK, Jones K, Nasr N,
Rustagi A, Cumming H, Donaghy H, Mak J, Gale
M, Jr., Churchill M, Hertzog P, Cunningham AL.
HIV infection of dendritic cells subverts the IFN
induction pathway via IRF-1 and inhibits type 1 IFN
production. Blood. 2011; 118: 298-308
Hertzog P, Forster S, Samarajiwa S. Systems
biology of interferon responses. J Interferon Cytokine
Res. 2011; 31: 5-11
Holland DG, Burleigh A, Git A, Goldgraben MA,
Perez-Mancera PA, Chin SF, Hurtado A, Bruna A,
Ali HR, Greenwood W, Dunning MJ, Samarajiwa
S, Menon S, Rueda OM, Lynch AG, McKinney S,
Ellis IO, Eaves CJ, Carroll JS, Curtis C, Aparicio
S, Caldas C. ZNF703 is a common Luminal B
breast cancer oncogene that differentially regulates
luminal and basal progenitors in human mammary
80 | Cambridge Research Institute Scientific Report 2011
epithelium. EMBO Mol Med. 2011; 3: 167-80
Manolopoulou I, Legarreta L, Emerson BC,
Brooks S, Tavaré S. A Bayesian approach to
phylogeographic clustering. Interface Focus. 2011; 1:
909-21
Marko NF, Quackenbush J, Weil RJ. Why is
there a lack of consensus on molecular subgroups
of glioblastoma? Understanding the nature of
biological and statistical variability in glioblastoma
expression data. PLoS One. 2011; 6: e20826
Marko NF, Xu Z, Gao T, Kattan MW, Weil RJ.
Predicting survival in women with breast cancer
and brain metastasis: A nomogram outperforms
current survival prediction models. Cancer. 2011;
Epub Dec 16
Massie CE, Lynch A, Ramos-Montoya A, Boren
J, Stark R, Fazli L, Warren A, Scott H, Madhu
B, Sharma N, Bon H, Zecchini V, Smith DM,
Denicola GM, Mathews N, Osborne M, Hadfield
J, Macarthur S, Adryan B, Lyons SK, Brindle KM,
Griffiths J, Gleave ME, Rennie PS, Neal DE, Mills
IG. The androgen receptor fuels prostate cancer
by regulating central metabolism and biosynthesis.
E MBO J. 2011; 30: 2719-33
Narita M, Young AR, Arakawa S, Samarajiwa
SA, Nakashima T, Yoshida S, Hong S, Berry
LS, Reichelt S, Ferreira M, Tavaré S, Inoki K,
Shimizu S, Narita M. Spatial coupling of mTOR and
autophagy augments secretory phenotypes. Science.
2011; 332: 966-70
Ritchie ME, Liu R, Carvalho BS, Irizarry RA,
Multiple ANZ. Comparing genotyping algorithms
for Illumina’s Infinium whole-genome SNP
BeadChips. BMC Bioinformatics. 2011; 12: 68
Siegmund KD, Marjoram P, Tavaré S, Shibata
D. High DNA methylation pattern intratumoral
diversity implies weak selection in many human
colorectal cancers. PLoS ONE. 2011; 6: e21657
Speed D, Tavaré S. Sparse partitioning: Nonlinear
regression with binary or tertiary predictors, with
application to association studies. Ann Appl Stat.
2011; 5: 873-93
Sottoriva A, Vermeulen L, Tavaré S. Modeling
evolutionary dynamics of epigenetic mutations in
hierarchically organized tumors. PLoS Comput Biol.
2011; 7: e1001132
Wilkinson RD, Steiper ME, Soligo C, Martin RD,
Yang ZH, Tavaré S. Dating primate divergences
through an integrated analysis of palaeontological
and molecular data. Systematic Biology. 2011; 60:
16-31
Yegnasubramanian S, Wu ZJ, Haffner MC, Esopi
D, Aryee MJ, Badrinath R, He TL, Morgan JD,
Carvalho B, Zheng QZ, De Marzo AM, Irizarry
RA, Nelson WG. Chromosome-wide mapping of
DNA methylation patterns in normal and malignant
prostate cells reveals pervasive methylation
of gene-associated and conserved intergenic
sequences. BMC Genomics. 2011; 12: 313
Yuan Y, Curtis C, Caldas C, Markowetz F. A
sparse regulatory network of copy-number driven
gene expression reveals putative breast cancer
oncogenes. IEEE/ACM Trans Comput Biol Bioinform.
2 011; Epub Jul 20
Yuan Y, Rueda OM, Curtis C, Markowetz F.
Penalized regression elucidates aberration hotspots
mediating subtype-specific transcriptional responses
in breast cancer. Bioinformatics. 2011; 27: 2679-85
Other publications
Ritchie ME, Dunning MJ, Smith ML, Shi W,
Lynch AG. BeadArray expression analysis using
bioconductor. PLoS Comput Biol. 2011; 7: e1002276
David Tuveson (page 50)
Tumour modelling and experimental medicine
laboratory
Primary research papers
Bapiro TE, Richards FM, Goldgraben MA, Olive
KP, Madhu B, Frese KK, Cook N, Jacobetz MA,
Smith DM, Tuveson DA, Griffiths JR, Jodrell DI.
A novel method for quantification of gemcitabine
and its metabolites 2′,2′-difluorodeoxyuridine and
gemcitabine triphosphate in tumour tissue by LCMS/MS: comparison with 19F NMR spectroscopy.
Cancer Chemother Pharmacol. 2011; 68: 1243-53
Caldwell ME, Denicola GM, Martins CP, Jacobetz
MA, Maitra A, Hruban RH, Tuveson DA. Cellular
features of senescence during the evolution of
human and murine ductal pancreatic cancer.
Oncogene. 2011; Epub Aug 22
DeNicola GM, Karreth FA, Humpton TJ,
Gopinathan A, Wei C, Frese K, Mangal D, Yu
KH, Yeo CJ, Calhoun ES, Scrimieri F, Winter JM,
Hruban RH, Iacobuzio-Donahue C, Kern SE,
Blair IA, Tuveson DA. Oncogene-induced Nrf2
transcription promotes ROS detoxification and
tumorigenesis. Nature. 2011; 475: 106-9
Froeling FE, Feig C, Chelala C, Dobson R,
Mein CE, Tuveson DA, Clevers H, Hart IR,
Kocher HM. Retinoic acid-induced pancreatic
stellate cell quiescence reduces paracrine Wntβ-catenin signaling to slow tumor progression.
Gastroenterology. 2011; 141: 1486-97 e14
Gopinathan A, Denicola GM, Frese KK, Cook
N, Karreth FA, Mayerle J, Lerch MM, Reinheckel
T, Tuveson DA. Cathepsin B promotes the
progression of pancreatic ductal adenocarcinoma in
mice. Gut. 2011; Epub Dec 9
Holland DG, Burleigh A, Git A, Goldgraben MA,
Perez-Mancera PA, Chin SF, Hurtado A, Bruna A,
Ali HR, Greenwood W, Dunning MJ, Samarajiwa
S, Menon S, Rueda OM, Lynch AG, McKinney S,
Ellis IO, Eaves CJ, Carroll JS, Curtis C, Aparicio
S, Caldas C. ZNF703 is a common Luminal B
breast cancer oncogene that differentially regulates
luminal and basal progenitors in human mammary
epithelium. EMBO Mol Med. 2011; 3: 167-80
Karreth FA, Frese K, DeNicola GM, Baccarini M,
Tuveson D. C-Raf is required for the initiation of
lung cancer by K-RasG12D. Cancer Discovery. 2011; 1:
128-36
Karreth FA, Tay Y, Perna D, Ala U, Tan SM,
Rust AG, Denicola G, Webster KA, Weiss D,
Perez-Mancera PA, Krauthammer M, Halaban R,
Provero P, Adams DJ, Tuveson DA, Pandolfi PP.
In vivo identification of tumor-suppressive PTEN
ceRNAs in an oncogenic BRAF-induced mouse
model of melanoma. Cell. 2011; 147: 382-95
Maniati E, Bossard M, Cook N, Candido JB,
Emami-Shahri N, Nedospasov SA, Balkwill FR,
Tuveson DA, Hagemann T. Crosstalk between
the canonical NF-κB and Notch signaling pathways
inhibits Pparγ expression and promotes pancreatic
cancer progression in mice. J Clin Invest. 2011; 121:
4685-99
Massie CE, Lynch A, Ramos-Montoya A, Boren
J, Stark R, Fazli L, Warren A, Scott H, Madhu
B, Sharma N, Bon H, Zecchini V, Smith DM,
Denicola GM, Mathews N, Osborne M, Hadfield
J, Macarthur S, Adryan B, Lyons SK, Brindle KM,
Griffiths J, Gleave ME, Rennie PS, Neal DE, Mills
IG. The androgen receptor fuels prostate cancer
by regulating central metabolism and biosynthesis.
E MBO J. 2011; 30: 2719-33
Pearson HB, Perez-Mancera PA, Dow LE, Ryan
A, Tennstedt P, Bogani D, Elsum I, Greenfield
A, Tuveson DA, Simon R, Humbert PO. SCRIB
expression is deregulated in human prostate cancer,
and its deficiency in mice promotes prostate
neoplasia. J Clin Invest. 2011; 121: 4257-67
Varela I, Tarpey P, Raine K, Huang D, Ong CK,
[and 25 others], Tuveson DA, Perez-Mancera PA,
Mustonen V, Fischer A, Adams DJ, Rust A, Chanon W, Subimerb C, Dykema K, Furge K, Campbell
PJ, Teh BT, Stratton MR, Futreal PA. Exome
sequencing identifies frequent mutation of the SWI/
SNF complex gene PBRM1 in renal carcinoma.
Nature. 2011; 469: 539-42
Other publications
Tuveson D, Hanahan D. Translational Medicine:
Cancer lessons from mice to humans. Nature. 2011;
471: 316-7
Fiona Watt (page 52)
Epithelial cell biology laboratory
Primary research papers
Arwert EN, Mentink RA, Driskell RR, Hoste
E, Goldie SJ, Quist S, Watt FM. Upregulation of
CD26 expression in epithelial cells and stromal
cells during wound-induced skin tumour formation.
Oncogene. 2011; Epub Jul 18
Blanco S, Kurowski A, Nichols J, Watt FM,
Benitah SA, Frye M. The RNA-methyltransferase
Misu (NSun2) poises epidermal stem cells to
differentiate. PLoS Genet. 2011; 7: e1002403
Collins CA, Kretzschmar K, Watt FM.
Reprogramming adult dermis to a neonatal
state through epidermal activation of β-catenin.
Development. 2011; 138: 5189-99
Connelly JT, Mishra A, Gautrot JE, Watt FM.
Shape-induced terminal differentiation of human
epidermal stem cells requires p38 and is regulated
by histone acetylation. PLoS ONE. 2011; 6: e27259
Driskell RR, Juneja VR, Connelly JT, Kretzschmar
K, Tan DW, Watt FM. Clonal growth of dermal
papilla cells in hydrogels reveals intrinsic differences
between Sox2-positive and -negative cells in vitro
and in vivo. J Invest Dermatol. 2011; Epub Dec 22
Fujiwara H, Ferreira M, Donati G, Marciano
Research Publications | 81
DK, Linton JM, Sato Y, Hartner A, Sekiguchi K,
Reichardt LF, Watt FM. The basement membrane
of hair follicle stem cells is a muscle cell niche. Cell.
2011; 144: 577-89
Giangreco A, Hoste E, Takai Y, Rosewell I, Watt
FM. Epidermal Cadm1 expression promotes
autoimmune alopecia via enhanced T cell adhesion
and cytotoxicity. J Immunol. 2011; Epub Dec 30
Giangreco A, Lu L, Vickers C, Teixeira VH, Groot
KR, Ilieva EV, George J, Nicholson AG, Sage
EK, Watt FM, Janes SM. β-catenin determines
upper airway progenitor cell fate and pre-invasive
squamous lung cancer progression by modulating
epithelial-to-mesenchymal transition. J Pathol. 2011;
Epub Nov 14
Hunziker L, Aznar Benitah S, Braun KM, Jensen
K, McNulty K, Butler C, Potton E, Nye E, Boyd
R, Laurent G, Glogauer M, Wright NA, Watt FM,
Janes SM. Rac1 deletion causes thymic atrophy. PLoS
ONE. 2011; 6: e19292
Narita M, Young AR, Arakawa S, Samarajiwa
SA, Nakashima T, Yoshida S, Hong S, Berry
LS, Reichelt S, Ferreira M, Tavaré S, Inoki K,
Shimizu S, Narita M. Spatial coupling of mTOR and
autophagy augments secretory phenotypes. Science.
2011; 332: 966-70
Other publications
Burdick JA, Watt FM. High-throughput stem-cell
niches. Nat Methods. 2011; 8: 915-6
Driskell RR, Clavel C, Rendl M, Watt FM. Hair
follicle dermal papilla cells at a glance. J Cell Sci. 2011;
124: 1179-82
Watt FM. Stem cells: on the front line. J Cell Sci.
2011; 124: 3527-8
Watt FM, Fujiwara H. Cell-extracellular matrix
interactions in normal and diseased skin. Cold Spring
Harb Perspect Biol. 2011; 3: pii: a005124
Douglas Winton (page 54)
Cancer and intestinal stem cells laboratory
Primary research papers
Ahmad I, Morton JP, Singh LB, Radulescu SM,
Ridgway RA, Patel S, Woodgett J, Winton DJ,
Taketo MM, Wu XR, Leung HY, Sansom OJ.
β-catenin activation synergizes with PTEN loss to
cause bladder cancer formation. Oncogene. 2011; 30:
178-89
March HN, Rust AG, Wright NA, Ten Hoeve J, de
Ridder J, Eldridge M, van der Weyden L, Berns A,
Gadiot J, Uren A, Kemp R, Arends MJ, Wessels
LF, Winton DJ, Adams DJ. Insertional mutagenesis
identifies multiple networks of cooperating genes
driving intestinal tumorigenesis. Nat Genet. 2011; 43:
1202-9
Stamataki D, Holder M, Hodgetts C, Jeffery R,
Nye E, Spencer-Dene B, Winton DJ, Lewis J.
Delta1 expression, cell cycle exit, and commitment
to a specific secretory fate coincide within a few
hours in the mouse intestinal stem cell system. PLoS
ONE. 2011; 6: e24484
Other publications
March HN, Winton DJ. mTOR regulation by
JNK: Rescuing the starving intestinal cancer cell?
82 | Cambridge Research Institute Scientific Report 2011
Gastroenterology. 2011; 140: 1387-91
Bioinformatics (page 58)
Matthew Eldridge
Primary research papers
Cairns J, Spyrou C, Stark R, Smith ML, Lynch AG,
Tavaré S. BayesPeak - An R package for analysing
ChIP-seq data. Bioinformatics. 2011; 27: 713-4
Dev H, Sharma NL, Dawson SN, Neal DE, Shah
N. Detailed analysis of operating time learning
curves in robotic prostatectomy by a novice
surgeon. BJU Int. 2011; Epub Oct 28
Gallagher FA, Tay KY, Vowler SL, Szutowicz H,
Cross JJ, McAuley DJ, Antoun NM. Comparing the
accuracy of initial head CT reporting by radiologists,
radiology trainees, neuroradiographers and
emergency doctors. Br J Radiol. 2011; 84: 1040-5
Gelson W, Hoare M, Dawwas MF, Vowler
S, Gibbs P, Alexander G. The pattern of late
mortality in liver transplant recipients in the United
Kingdom. Transplantation. 2011; 91: 1240-4
Holland DG, Burleigh A, Git A, Goldgraben MA,
Perez-Mancera PA, Chin SF, Hurtado A, Bruna A,
Ali HR, Greenwood W, Dunning MJ, Samarajiwa
S, Menon S, Rueda OM, Lynch AG, McKinney S,
Ellis IO, Eaves CJ, Carroll JS, Curtis C, Aparicio
S, Caldas C. ZNF703 is a common Luminal B
breast cancer oncogene that differentially regulates
luminal and basal progenitors in human mammary
epithelium. EMBO Mol Med. 2011; 3: 167-80
Lamb AD, Qadan M, Roberts S, Timlin H, Vowler
SL, Campbell FM, Grigor K, Bartlett JM, McNeill
SA. CD4+ and CD8+ T-lymphocyte scores cannot
reliably predict progression in patients with benign
prostatic hyperplasia. BJU Int. 2011; 108: E43-50
Lamb AD, Vowler SL, Johnston R, Dunn N,
Wiseman OJ. Meta-analysis showing the beneficial
effect of alpha-blockers on ureteric stent
discomfort. BJU Int. 2011; 108: 1894-902
Lawson MH, Cummings NM, Rassl DM, Russell
R, Brenton JD, Rintoul RC, Murphy G. Two novel
determinants of etoposide resistance in small cell
lung cancer. Cancer Res. 2011; 71: 4877-87
March HN, Rust AG, Wright NA, Ten Hoeve J, de
Ridder J, Eldridge M, van der Weyden L, Berns A,
Gadiot J, Uren A, Kemp R, Arends MJ, Wessels
LF, Winton DJ, Adams DJ. Insertional mutagenesis
identifies multiple networks of cooperating genes
driving intestinal tumorigenesis. Nat Genet. 2011; 43:
1202-9
Massie CE, Lynch A, Ramos-Montoya A, Boren
J, Stark R, Fazli L, Warren A, Scott H, Madhu
B, Sharma N, Bon H, Zecchini V, Smith DM,
Denicola GM, Mathews N, Osborne M, Hadfield
J, Macarthur S, Adryan B, Lyons SK, Brindle KM,
Griffiths J, Gleave ME, Rennie PS, Neal DE, Mills
IG. The androgen receptor fuels prostate cancer
by regulating central metabolism and biosynthesis.
E MBO J. 2011; 30: 2719-33
Nascimento EM, Cox CL, Macarthur S, Hussain
S, Trotter M, Blanco S, Suraj M, Nichols J, Kubler
B, Benitah SA, Hendrich B, Odom DT, Frye M.
The opposing transcriptional functions of Sin3a and
c-Myc are required to maintain tissue homeostasis.
Nat Cell Biol. 2011; 13: 1395-405
Robinson JL, Macarthur S, Ross-Innes CS, Tilley
WD, Neal DE, Mills IG, Carroll JS. Androgen
receptor driven transcription in molecular apocrine
breast cancer is mediated by FoxA1. EMBO J. 2011;
30: 3019-27
Other publications
Drummond GB, Vowler SL. Data interpretation:
using probability. J Physiol. 2011; 589: 2433-5 (also
published in Adv Physiol Educ., Br J Pharmacol., Clin
Exp Pharmacol Physiol., Exp Physiol., Microcirculation)
Drummond GB, Vowler SL. Show the data, don’t
conceal them. J Physiol. 2011; 589: 1861-3 (also
published in Adv Physiol Educ., Br J Pharmacol., Clin
Exp Pharmacol Physiol., Exp Physiol., Microcirculation)
Ritchie ME, Dunning MJ, Smith ML, Shi W,
Lynch AG. BeadArray expression analysis using
bioconductor. PLoS Comput Biol. 2011; 7: e1002276
cell glycans in vivo. FASEB J. 2011; 25: 2528-37
Sir JH, Barr AR, Nicholas AK, Carvalho OP,
Khurshid M, Sossick A, Reichelt S, D’Santos C,
Woods CG, Gergely F. A primary microcephaly
protein complex forms a ring around parental
centrioles. Nat Genet. 2011; 43: 1147-53
Stockmann H, Neves AA, Stairs S, IrelandZecchini H, Brindle KM, Leeper FJ. Development
and evaluation of new cyclooctynes for cell surface
glycan imaging in cancer cells. Chemical Science. 2011;
2: 932-6
Flow cytometry (page 62)
Richard Grenfell
Primary research papers
Al-Mayhani MT, Grenfell R, Narita M, Piccirillo
S, Kenney-Herbert E, Fawcett JW, Collins
VP, Ichimura K, Watts C. NG2 expression in
glioblastoma identifies an actively proliferating
population with an aggressive molecular signature.
Neuro Oncol. 2011; 13: 830-45
Genomics (page 63)
James Hadfield
Primary research papers
Holmes KA, Hurtado A, Brown GD, Launchbury
R, Ross-Innes CS, Hadfield J, Odom DT, Carroll
JS. Breast Cancer Special Feature: Transducin-like
enhancer protein 1 mediates estrogen receptor
binding and transcriptional activity in breast cancer
cells. Proc Natl Acad Sci USA. 2011; Epub May 2
Massie CE, Lynch A, Ramos-Montoya A, Boren
J, Stark R, Fazli L, Warren A, Scott H, Madhu
B, Sharma N, Bon H, Zecchini V, Smith DM,
Denicola GM, Mathews N, Osborne M, Hadfield
J, Macarthur S, Adryan B, Lyons SK, Brindle KM,
Griffiths J, Gleave ME, Rennie PS, Neal DE, Mills
IG. The androgen receptor fuels prostate cancer
by regulating central metabolism and biosynthesis.
E MBO J. 2011; 30: 2719-33
Light microscopy (page 65)
Stefanie Reichelt
Primary research papers
Narita M, Young AR, Arakawa S, Samarajiwa
SA, Nakashima T, Yoshida S, Hong S, Berry
LS, Reichelt S, Ferreira M, Tavaré S, Inoki K,
Shimizu S, Narita M. Spatial coupling of mTOR and
autophagy augments secretory phenotypes. Science.
2011; 332: 966-70
Neves AA, Stockmann H, Harmston RR, Pryor
HJ, Alam IS, Ireland-Zecchini H, Lewis DY, Lyons
SK, Leeper FJ, Brindle KM. Imaging sialylated tumor
Pharmacokinetics and
pharmacodynamics (page 66)
Donna-Michelle Smith
Primary research papers
Massie CE, Lynch A, Ramos-Montoya A, Boren
J, Stark R, Fazli L, Warren A, Scott H, Madhu
B, Sharma N, Bon H, Zecchini V, Smith DM,
Denicola GM, Mathews N, Osborne M, Hadfield
J, Macarthur S, Adryan B, Lyons SK, Brindle KM,
Griffiths J, Gleave ME, Rennie PS, Neal DE, Mills
IG. The androgen receptor fuels prostate cancer
by regulating central metabolism and biosynthesis.
E MBO J. 2011; 30: 2719-33
Proteomics (page 68)
Clive D’Santos
Primary research papers
Finch AJ, Hilcenko C, Basse N, Drynan LF,
Goyenechea B, Menne TF, Gonzalez Fernandez
A, Simpson P, D’Santos CS, Arends MJ, Donadieu
J, Bellanne-Chantelot C, Costanzo M, Boone C,
McKenzie AN, Freund SM, Warren AJ. Uncoupling
of GTP hydrolysis from eIF6 release on the
ribosome causes Shwachman-Diamond syndrome.
Genes Dev. 2011; 25: 917-29
Lewis AE, Sommer L, Arntzen MO, Strahm
Y, Morrice NA, Divecha N, D’Santos CS.
Identification of nuclear phosphatidylinositol
4,5-bisphosphate-interacting proteins by neomycin
extraction. Mol Cell Proteomics. 2011; 10: M110
003376
Sir JH, Barr AR, Nicholas AK, Carvalho OP,
Khurshid M, Sossick A, Reichelt S, D’Santos C,
Woods CG, Gergely F. A primary microcephaly
protein complex forms a ring around parental
centrioles. Nat Genet. 2011; 43: 1147-53
Research Publications | 83
External Funding
The Cambridge Research Institute gratefully acknowledges those
organisations that have provided support during the period of this
report to the individuals and laboratories listed.
Academy of Medical Sciences
Christine Parkinson (Brenton laboratory)
Addenbrooke’s Charitable Trust
Christine Parkinson (Brenton laboratory)
Tom Booth (Brindle laboratory)
Caldas laboratory
American Association of Neurological Surgeons
Nick Marko (Tavaré laboratory)
Biotechnology and Biological Sciences Research
Council (BBSRC)
Hélène Bon (Neal laboratory)
Tavaré laboratory
Holly Canuto (Brindle laboratory)
Breast Cancer Campaign
Kelly Holmes (Carroll laboratory)
Breast Cancer Research Foundation
Ana-Teresa Maia (Ponder laboratory)
Brian Cross Memorial Trust
Nicola Ainsworth (Griffiths laboratory)
Caja Madrid Foundation
Pedro Perez Mancera (Tuveson laboratory)
Cambridge Commonwealth Trust and Overseas
Research Studentship
Charlotte Ng (Brenton laboratory)
Cancer Research UK (competitive bursary)
Kamarul Zaki (Carroll laboratory)
Cancer Research UK (competitive fellowship)
Athena Matakidou (Tuveson laboratory)
Natalie Cook (Tuveson laboratory)
Cancer Research UK (competitive grant)
Narita laboratory
Watt laboratory
Caring for Carcinoid Foundation
Tuveson laboratory
China Scholarship Council
Ruiling Xu (Jodrell laboratory)
84 | Cambridge Research Institute Scientific Report 2011
Commonwealth Scholarship and Fellowships Plan
Sarah-Jane Dawson (Caldas laboratory)
Michelle Ward (Odom laboratory)
Deutscher Akademischer Austausch Dienst
(DAAD)
Albrecht Neesse (Tuveson laboratory)
Alexander Kuznetsov (Watt Laboratory)
EC (Health)
Brindle laboratory
Caldas laboratory
Tuveson laboratory
Watt laboratory
EC Initial Training Network
Irene Marco and Eva Serrao (Brindle laboratory)
EC Innovative Medicines Initiative
Griffiths/Brindle laboratories
EC Marie Curie Initial Training Network
Joana Borlido (Neal laboratory)
Esther Hoste (Watt laboratory)
Bianca Schmitt (Odom laboratory)
Christine Weber (Watt laboratory)
EC Marie Curie International Re-integration
Grant
Klara Stefflova (Odom laboratory)
EC Marie Curie Intra-European Fellowship
Joan Boren (Brindle laboratory)
Alejandra Bruna (Caldas laboratory)
Christine Feig (Tuveson laboratory)
Tiago Rodrigues (Brindle laboratory)
Engineering and Physical Sciences Research
Council (EPSRC)
Brenton laboratory
EU Seventh Framework Programme (FP7)
Helen Ross-Adams (Neal laboratory)
EUREKA EU
Light Microscopy (with the Universities of Utrecht
and Heidelberg, the MRC Laboratory of Molecular
Biology and Nikon Europe)
European Molecular Biology Organisation (EMBO)
Carroll laboratory
Odom laboratory
Michelle Ward (Odom laboratory)
European Research Council (ERC)
Carroll laboratory
Odom laboratory
European Science Foundation (ESF)
Markowetz laboratory
Experimental Cancer Medicine Centre
Bin Liu (Caldas laboratory)
Federation of European Biochemical Sciences
Sara Cipolat (Watt laboratory)
Fibrogen Inc.
Tuveson laboratory
Gates Foundation
Timothy Humpton (Tuveson laboratory)
GlaxoSmithKline
Jodrell laboratory
Neal/Brindle Laboratories
Hales Clinical Fellowship
Carles Escriu (Watt laboratory)
Lucy Gossage (Jodrell laboratory)
Human Frontier Science Program (HFSP)
Narita laboratory
Institut National du Cancer/DoH
Anthea Messent (Murphy laboratory)
Irish Research Council for Science, Engineering
and Technology
Aisling Redmond (Carroll laboratory)
Microsoft Research
Brenton laboratory
National Institute for Health Research
Christine Parkinson (Brenton laboratory)
Alastair Lamb (Neal laboratory)
Maxine Tran (Neal laboratory)
Naomi Sharma (Neal laboratory)
Aileen Marshall (Odom laboratory)
Ponder laboratory
National Institutes of Health, USA (NIH)
Griffiths laboratory
Tuveson laboratory
Ryan Fiehler (Watt Laboratory)
Nuffield Foundation
Gergely laboratory
Pancreatic Cancer UK
Shivan Sivakumar (Tuveson laboratory)
Portuguese Science Foundation
Pedro Correa de Sampaio (Murphy laboratory)
Jose Sandoval (Caldas laboratory)
Prostate Cancer Charity
Neal laboratory
Prostate UK
Ajoeb Baridi (Stingl and Neal laboratories)
Neal laboratory
Royal Society University Research Fellow
Fanni Gergely
Schultheiss-Reiser Foundation
Sven Quist (Watt laboratory)
Science and Technology Facilities Council
Brenton laboratory
Italian Association for Cancer Research
Daniele Perna (Tuveson laboratory)
The Leukemia and Lymphoma Society, USA
Piotr Dzien (Brindle laboratory)
Japan Foundation for the Promotion of Science
Ken Natsuga (Watt Laboratory)
Uehara Memorial Foundation
Hiro Fujiwara (Watt laboratory)
Mahito Sadaie (Narita laboratory)
Japan Society for the Promotion of Science
Narita laboratory
KWF Dutch Royal Fellowship
Wilbert Zwart (Carroll laboratory)
Yousef Jameel Scholarship
Sui Seng Tee (Brindle laboratory)
Kyowa Hakko Kirin Co., Ltd.
Narita laboratory
Medical Research Council (MRC)
Jonathan Cairns (Tavaré laboratory)
Ioannis Gounaris (Brenton laboratory)
Tom Booth (Brindle laboratory)
Stephen Goldie (Watt laboratory)
Murphy Laboratory
External Funding | 85
Seminars and Conferences
The CRI hosts a number of seminar series, covering basic to
translational aspects of cancer research, and quantitative biology.
CRI Seminars in Cancer
Conferences
Stephan Beck, UCL Cancer Institute
Reverse phenotyping: towards an integrated
(epi)genomic approach to complex phenotypes and
common disease.
Attended by over 250 delegates, the meeting
featured a series of diverse talks from
invited speakers and a keynote lecture by
Robert Weinberg (Whitehead Institute). See
page 88 for more information.
We welcomed the following speakers to present
in our international seminar series, CRI Seminars in
Cancer:
Richard Treisman, Cancer Research UK London
Research Institute, Lincoln’s Inn Fields
MAL: linking the actin cytoskeleton to
transcriptional regulation
Hing Leung, Beatson Institute for Cancer Research
Analysis of sprouty2 in human prostate
carcinogenesis
Richard Peto, University of Oxford
Changing cancer mortality
Edison Liu, Genome Institute of Singapore
Integrated solutions in cancer genomics
David Threadgill, North Carolina State University
Modeling non-familial colon cancer susceptibility in
mice
Richard Young, Whitehead Institute for Biomedical
Research and MIT
Transcriptional regulation of cell state
Sean J Morrison, University of Texas Southwestern
Medical Center
The intrinsic and extrinsic regulation of stem cells
self-renewal
Details of all CRI seminar series – CRI Seminars in
Cancer, Cambridge Oncology Seminars, Lectures
in Cancer Biology and CRI Seminars in Quantitative
Biology – can be found on www.cambridgecancer.
org.uk/seminars-conferences/
86 | Cambridge Research Institute Scientific Report 2011
Cambridge Cancer Centre Annual Symposium
23 June
Institute Retreat
6–7 October
This year’s retreat was once again held at the
CRI. We continued with the popular talks for
non‑scientists series, which was well attended. This
year we reduced the number of research talks and
added two new sessions, which focussed on recent
results papers and clinical case presentations. This
provided an excellent opportunity to review work
in progress, assess successfully published results, and
gain insight into the clinical applications of the CRI’s
research. The team building activity of producing a
collage of a Cambridge cityscape resulted in a new
work of art for the Institute.
CRI Symposium, Unanswered Questions in
Transcription
4–5 November
This year we welcomed a distinguished line-up of
international speakers to speak about and discuss
questions in transcription, in a programme put
together by the scientific organising committee of
Jason Carroll, Florian Markowetz, Adele Murrell and
Duncan Odom.
Session 1: Transcriptional regulation in mammalian
cells
Ronald Evans, Chair, Salk Institute for Biological
Studies. Your genome on steroids: re-thinking
glucocorticoid receptor “trans-repression”
Tony Kouzarides, Wellcome Trust/Cancer
Research UK Gurdon Institute. Chromatin modifying
enzymes: their function and role in cancer
Stephan Beck, University College London. EWAS:
the new kid on the block for epigenome-wide
association studies
Nicola Reynolds, Wellcome Trust Centre for Stem
Cell Research. NuRD interacts with PRC2 to control
gene expression in embryonic stem cells (Selected
talk from submitted abstracts)
Paolo Sassone-Corsi, University of California, Irvine.
Synergy of metabolome and transcriptome by the
circadian clock
Shelley Berger, University of Pennsylvania.
Signaling kinases activate transcription via histone
phosphorylation
Session 2: Regulatory networks
Daphne Koller, Chair, Stanford University.
Probabilistic methods for reconstructing gene
regulatory networks from high-throughput data
Joe Gray, Oregon Health and Science University.
Systems biology approaches to predictive markers
in breast cancer
Jan Korbel, EMBL Heidelberg. Mapping structural
variants by population-scale genome sequencing
Klaas Mulder, Cancer Research UK Cambridge
Research Institute. A network of epigenetic
strategies controls adult stem cell fate (Selected talk
from submitted abstracts)
Shirley Liu, Harvard University. Distinct modes of
transcription factor binding and chromatin dynamics
Dana Pe’er, Columbia University. Integrative cancer
genomics: drivers, pathways and drugs
Session 4: RNA regulators of gene expression
David Baulcombe, Chair, University of Cambridge.
Small RNA and epigenetic regulation
Carlos Caldas, Cancer Research UK Cambridge
Research Institute. The complex life of small RNA
(or what I have learned about it in breast cancer!)
Vihandha Wickramasinghe, MRC Cancer Cell Unit.
Selective mRNA export from the mammalian cell
nucleus mediated by GANP (Selected talk from
submitted abstracts)
Jernej Ule, MRC Laboratory of Molecular Biology.
Competition between hnRNP C and U2AF65
controls the exonization of cryptic exons
John Lis, Cornell University. Focused and genomewide analysis of promoter-proximal pausing
2012 events
22 June: Cambridge Cancer Centre Annual
Symposium
11–12 October: CRI Retreat
2–3 November: CRI Annual International
Symposium – Unanswered Questions in Cancer
Sequencing
Session 3: Cancer epigenetics
Stephen Baylin, Chair, Johns Hopkins University.
Some molecular mechanisms underlying
epigenetically mediated transcription profiles in
cancer
Alan Clarke, Cardiff School of Biosciences.
Epigenetic regulators of intestinal homeostasis and
tumorigenesis
Keith Robertson, Georgia Health Sciences University.
Interplay between DNA methylation and histone
modifications in cancer cells
Santiago Uribe-Lewis, Cancer Research UK
Cambridge Research Institute. Cytosine
hydroxymethylation in colorectal cancer (Selected
talk from submitted abstracts)
Peter Jones, University of Southern
California. Chromatin choreography after
5‑Aza‑2′‑deoxycytidine treatment
Kristian Helin, University of Copenhagen. Functional
roles of TET proteins and hydroxymethylation in
stem cells and cancer
Seminars and Conferences | 87
Cambridge Cancer Centre
Director
Bruce Ponder
Coordinator
Katrien Van Look
Director of Scientific
Development
Kenneth Seamon
Development
Director – Clinical
Simon Oberst
The Cambridge Cancer Centre (CCC) is a partnership between
Cancer Research UK, the University of Cambridge, Cambridge
University Hospitals NHS Foundation Trust and the Medical Research
Council. The vision of the CCC is to build strong links across
disciplines from the laboratory to the clinic.
In October this year, the CCC appointed two
Development Directors, Kenneth Seamon and
Simon Oberst. Kenneth’s role as Director
of Scientific Development is to progress the
development of scientific interactions across
Cambridge, as part of the new role of the Cancer
Centre as a designated University “Strategic
Initiative”. Among other things, this will provide us
with a clear description of cancer-related research
activities and resources across Cambridge, which
we hope will be an effective “shop window”
for potential pharma partners. Simon is the
Development Director – Clinical. He is working
on a detailed analysis of our current cancer services
and the interface between those and clinical
research. He is also providing financial data on
which to base a case for new initiatives in the
clinical area. The Cancer Centre began as a virtual
framework for interaction; these appointments are
helping to make it more concrete.
In October, the first four PhD students appointed
under the Cancer Centre began their studies.
Nikola Novcic started in Mariann Bienz’s lab
at the MRC Laboratory of Molecular Biology,
and Henrike Resemann in the Department of
Pathology with Christine Watson. Christine Hänni
will work with Philip Zegerman at the Gurdon
Institute, and Leah Bury with David Glover in the
Department of Genetics. Three Clinical Fellows
also began their PhD studies in the Cancer Centre
in 2011. Amit Roshan will work with Phil Jones
at the MRC Cancer Cell Unit, Jonathan Sive at
the Cambridge Institute for Medical Research
with Bertie Göttgens, and Morteza Jalali with
Ludovic Vallier in the Stem Cell Programme within
the Department of Surgery. We have recently
appointed a further four non-clinical PhD students
and three Clinical Fellows to start in October
2012.
88 | Cambridge Research Institute Scientific Report 2011
The pump-priming awards of the CCC were
launched again in September, following a twoyear gap. These awards are of £50K for one
year, and are to fund novel and interdisciplinary
projects between at least two different University
Departments or Institutes. Twenty-four high
quality applications were received, a record
number. Funding was sufficient to support four of
these after a keen competition.
The 5th Annual CCC Symposium took place in
June with around 300 delegates from a wide
range of University departments, institutes,
biotech companies and journal editorial offices
in Cambridge. The Keynote Lecture was given
by Robert Weinberg from MIT, who gave a
stimulating talk on the epithelial-mesenchymal
transition, cancer stem cells and malignant
progression. The talks from Cambridge-based
researchers spanned a range of disciplines: Kevin
Brindle (Department of Biochemistry/CRI) molecular imaging; Douglas Easton (Centre for
Cancer Genetic Epidemiology/Departments of
Public Health and Primary Care, and Oncology)
- genetics of cancer susceptibility; Douglas
Fearon (Departments of Medicine/CRI) - novel
approaches to cancer immuno-therapy; Stephen
Jackson (Gurdon Institute/Department of
Chemistry) - DNA repair and therapeutic targets
for cancer; Roger Williams (MRC Laboratory of
Molecular Biology) - molecular mechanisms of
activation of phosphoinositide 3-kinases; Chris
Abell (Department of Chemistry) - disrupting
protein-protein interactions; Rebecca Fitzgerald
(MRC Cancer Cell Unit/Department of Medicine)
- Barrett’s oesophagus and early diagnosis of
oesophageal cancer; and Duncan Jodrell (CRI)
- evaluating new therapeutics in the clinic. A
successful poster session took place over lunch,
and the day ended with the well-known barbecue!
Outreach and Fundraising
Coordinator
Katrien Van Look
CRI staff are actively involved in public engagement and fundraising
events.
This year we again took part in the Cambridge
Science Festival and in the Fen Edge Family
Festival. The Cambridge Science Festival is the
UK’s largest free science festival and attracts
around 35,000 visitors over a two week period.
The Fen Edge Family Festival in Cottenham is a
small local village festival, but was equally fun!
We exhibited a mock-up lab and got children
and adults involved with DNA extraction, lots of
pipetting and running gels – it was a great hit.
who organise or take part in a range of activities
and events: this year we met Phil Purdy and Jeff
Crooke who are climbing Mount Everest to raise
money for pancreatic and oesphageal cancer,
and in January 2012 we met Matt Wallace who is
walking from John O’ Groats to Land’s End raising
money for Cancer Research UK. Matt started his
adventure in September and is still walking!
CRI staff take on their own fundraising challenges and
many of the women participate in the Race for Life
events locally. In addition to the staff running in the
Cambridge Race for Life on the 3rd July, around 50
researchers and scientific support staff volunteered
on the day. Dr Hayley Whitaker (Principal Scientific
Officer, Neal laboratory) gave a short, moving and
motivational speech, and thanked all the participants
for supporting Cancer Research UK.
We again hosted the East Regional Final of the
Institute of Ideas Debating Matters competition
at the CRI and a few members of staff were
involved in judging the debates. The competition
is held nationally for sixth-form students to debate
current issues in science, politics, the arts and
other subjects.
CRI staff regularly give talks at local primary and
secondary schools. We also give many talks and
tours of our laboratories for groups of Cancer
Research UK fundraisers and supporters, around 30
per year. It is very inspiring to meet our fundraisers
CCC, Outreach and Fundraising | 89
Academic Administration
The graduate student body in the Cambridge Research Institute is
composed of PhD students, MB/PhD students, MPhil students and
clinical research training fellows. Details of the graduate training
programme are co‑determined by Cancer Research UK, the Institute,
the University Department with which a student is affiliated, and
the University of Cambridge. The entire staff of the Institute are
committed to making it a great place to work and study and all
provide support at all levels to our students.
The Cambridge Experience
There are currently 65 graduate students at the
CRI which corresponds to approximately one third
of our entire research population. Our graduate
students are fully integrated into their research
groups where they are expected to make valuable
contributions to the success of their groups. Eleven
students commenced study in October 2011 with a
further three starting in January 2012, of these, two
are MB/PhD students, and two are MPhil students.
Our student body is highly international – out of 14
new starters, four are from the UK, three from the
EU and the remaining seven from further afield.
although some are members of the Departments
of Genetics (Watt laboratory), Biochemistry
(Brindle laboratory), Medicine (Fearon laboratory)
or Applied Maths and Theoretical Physics
(Tavaré laboratory). All students also belong to a
Cambridge college so they gain the full collegiate
experience while studying in the Institute. Many
colleges provide graduate student accommodation
and an active social network as well as sporting
facilities. Students also have access to a personal
tutor in college who is available to provide pastoral
care if required.
First year graduate students
Support and Mentoring
Each student has a supervisor who is a group leader
and is also assigned a second supervisor who acts
as a mentor and provides support. In addition, Ann
Kaminski (the Head of Scientific Administration)
acts as the first point of contact for any student
with a query or difficulty that is not directly related
to their scientific work. All student matters in the
Institute are overseen by the Studentships and
Fellowships Committee, chaired by Fiona Watt.
This committee has the well-being of our students
at heart, while ensuring that they are fulfilling the
requirements of the University of Cambridge for
obtaining their degree.
Aspiring students apply to specific group leaders via
Cancer Research UK’s on-line application system.
Group leaders select students for interview in
December and January and successful applicants are
then given support to apply for graduate entry to
the University of Cambridge. Most of our students
are members of the Department of Oncology,
The Graduate Programme
Soon after their arrival, all of our new graduate
students join the University graduate intake to
attend the compulsory introductory safety and
induction courses organised by the University,
followed by similar couses specific to the Institute.
All students and group leaders are invited
to attend a reception in the Institute, where
the students are welcomed by the Institute’s
director Bruce Ponder and the members of the
90 | Cambridge Research Institute Scientific Report 2011
administration team. This is their first opportunity
to meet some of the staff who will help them over
the years to come.
All first year graduate students are required to
attend a series of around 30 lectures in cancer
biology, which are organised by the Department
of Oncology. As our students come from diverse
backgrounds, such as medicine, basic biology,
mathematics and statistics, this course goes
some way to ensuring that they all obtain a good
grounding in cancer biology. The lectures are given
by specialists in their fields and they provide the
students with a comprehensive overview of cancer
biology, ranging from basic cell biology through to
cancer diagnosis and treatment. This excellent and
unique resource is available to all members of the
University and is widely attended. The students
are also briefed by the core facilities managers to
learn of the services available to them. They also
attend courses specific to the demands of their
projects.
After two months in the Institute all first year
students give 15 minute talks to all members of the
Institute to explain the nature of the projects. In
accordance with University regulations, all graduate
students studying Biological Sciences in Cambridge
are not at first registered for PhD studies and must
qualify for registration by successfully completing a
first year report followed by a viva. Two examiners
assess a student’s report and then write a report
on their progress over the past year.
Our second and third year PhD students give
research talks as part of the Institute Lunchtime
Seminar series, attended by all Institute staff. In
addition, students complete a written report
towards the end of their second year which
summarises their work to date and also forms
the basis for discussions regarding further work.
Our graduate students all follow the three year
graduate programme supported by the University
of Cambridge; a further year is available if necessary
to complete their thesis, which must be submitted
within four years.
Like their colleagues in London, our students are
encouraged to attend numerous courses planned
to hone their transferable skills. These courses
range from advice on how to make scientific
posters to the Cancer Research UK-organised
Graduate Students Public Engagement with Science
and Technology (GRADPEST) course.
The Graduate Society
The graduate students have organised themselves
into a very active society which organises monthly
journal clubs and a wide variety of social events
including movie nights, punting and the occasional
wine-tasting. The students also arrange meetings
with visiting speakers and have a Christmas dinner
with an invited speaker — this year’s speaker was
the CEO of Abcam, Dr Jonathan Milner. The
society has also introduced a highly effective
mentoring scheme in which all first year students
have two mentors located in different parts of
the building. This provides new students with
recognisable friendly faces in other labs and also
helps them to settle in much quicker.
The graduate students meet with James D Watson during his
recent visit to the institute.
Other Student Activities
Many of the students have attended conferences
and workshops both in the UK and overseas,
including the graduate student conference, held this
year in Glasgow. This yearly meeting is organised
by students from the host institution and attendees
are invited from a range of European research
institutes.
Awards, Prizes and Achievements
Several of our students won prizes or were invited
to speak at meetings this year:
Simon Buczacki (Winton lab) won the best
oral presentation prize at the Association of
Coloproctology of Great Britain and Ireland
Eastern Chapter Annual Meeting
Natalie Cook (Tuveson lab) won the McElwain
prize – a talk at the annual Association of Cancer
Physicians meeting and £1000 prize.
Sarah Kozar (Winton lab) was selected to give a
10 minute presentation at this year’s International
Society for Stem Cell Research conference in
Toronto.
Dominic Schmidt (Odom/Carroll labs) has been
invited to speak at “From beads on a string to the
pearls of regulation: the structure and dynamics of
chromatin.” A joint Biochemical Society / Wellcome
Trust conference. 3–4 August 2011
Mike Smith (Tavaré lab) was awarded runner-up
in the “Best technical poster” category at the 2011
UseR conference, Warwick, UK
Chris Tape (Murphy lab) was invited to speak at
the Gordon Research Conference on Regulated
Proteolysis of Cell Surface Proteins.
Michelle Ward (Odom lab) has been awarded an
EMBO short-term fellowship.
Academic Administration | 91
Institute Administration
The administration team facilitates the smooth running of
the Institute by providing infrastructure and support to the
Director.
Director of Operations
John Wells
PA to the Director of
Operations
Belinda Ledgerton
Cambridge Cancer Centre and
Outreach Coordinator
Katrien Van Look
The team provides administrative support to
group leaders and supports research activities
through management of the laboratories and
core facilities. The team also coordinates
graduate student administration and laboratory
finance, the Cambridge Cancer Centre
(page 88) and outreach activities (page 89).
In addition to laboratory management each of
the group leaders has administrative support
provided by one of the dedicated research
administrators.
Scientific Administration
Graduate student and summer student
administration is overseen by Ann Kaminski
(page 90). The team organises tenure reviews
and mid-term reviews for the research groups,
and reviews for the core facilities.
Head of Scientific Administration
Ann Kaminski
Scientific Communications
Officer
Laura Blackburn
Scientific Communications
Administrator
Julie Bailey
Audio Visual and Graphic Design
Officer
Charles Thomson
PA to the Director
Jean Miller
Director’s Office Assistant
Brenda Wright
Human Resources Manager
Sophie Duncan
Finance Manager
Ruth Bennett
*
joined in 2011
†
left in 2011
The scientific administration team is responsible
for the running of symposia, seminars, chalk talks
and committees that take place in the Institute,
including providing full audio visual cover. The
team also organise the CRI symposium and
the group leader and Institute retreats. We
produce the Institute’s publications including the
annual report, the CRI newsletter, leaflets and
posters for fundraising and outreach activities,
write for and edit the intranet, and provide
content for the internet.
guidance in the areas of recruitment, personal
and team development, pay and grading,
employment law and staff wellbeing. The
Institute has a mixed economy of staff from
Cancer Research UK and the University of
Cambridge so collaboration is essential to
provide a seamless employment experience,
allowing the Institute to focus on its research.
This year has seen the CRI bring together a
local HR network of HR professionals in the
area of biomedical research. Cambridge is
internationally recognised for its transformative
research, greatly enhanced by scientific
collaboration. The HR Biomedical Network aims
to mirror this collaborative approach to ensure
the provision of the HR infrastructure necessary
for our researchers. 2011 has also seen the
launch of the CRI’s postdoctoral research fellow
development programme offering a portfolio of
development opportunities to support scientists
on their journey to scientific success.
As we move into 2012 the CRI’s HR will
continue to interact with the wider Cambridge
HR community in creating an environment
conducive to high quality research.
The team is also responsible for internal and
external communications, coordination with the
Cancer Research UK press office, and is involved
in the organisation of fundraising visits. We have
also been heavily involved in the co-ordination
of volunteering efforts at Cancer Research UK
fundraising events this year. CRI staff took part
in the Cambridge Race for Life, for more details
see page 89.
Finance
The finance team:
•Help budget holders efficiently manage their
budgets.
•Provide financial analysis to CRI management
to inform decision-making.
•Assist with the budget and business planning
process for the CRI.
•Assist with the acquisition and management
of grants.
•Provide a link between the Cancer
Research UK finance department and the CRI.
•Help with ad hoc queries and concerns.
Human Resources
Human Resources (HR) work in partnership
with the Institute to provide support and
Laboratory Management
The Laboratory Management team form part
of the Institute administration and continue to
92 | Cambridge Research Institute Scientific Report 2011
Senior Grants Administrator
Emma Ryley
Finance Assistant
Paulina Annison
Research Administrators
Paula Baines
Julie Barlow
Kate Davenport
Frankie Dubery *
Marion Karniely
Catharine Savin
Denise Schofield
Tania Smith
Tina Thorn
provide a vital role in underpinning the Institute’s
ever-evolving research activities. Doug Fearon
and his group joined in the summer from the
MRC Laboratory of Molecular Biology and
have settled in well on the second floor. It has
also been a busy year within the team with
Ross Coates being appointed as the Scientific
Porter, assisting the groups with deliveries and
replenishment of specialised gases and other key
consumables on the lab floors. More recently
Chris Lehane has joined as the CRI Laboratory
Manager, overseeing the Laboratory Management
team, Glasswash and Media, and liaising with
Property Services and Health and Safety to
ensure the smooth running of the laboratories.
The team continues to work closely with
other departments both within the CRI
and other Cancer Research UK buildings, as
well as neighbouring research Institutes and
Addenbrooke’s Hospital.
Assistant Director of Operations
Katy Smith
Laboratory Manager
Christine Lehane*
Floor Managers
Chris Harley
Catherine Pauley
Siân Webster
Laboratory Support Assistants
Carienne Bailey
Caroline Edwards
Scientific Porter
Ross Coates
Administrators
Katherine Balch*
Laura Bluer †
Liz Heselwood
Glasswash and Media Supervisor
Mark Jay
Deputy Glasswash and Media
Supervisor
Jackie Coulson
Lab Support Technicians
Susan Boddy
Ursula Clarke†
Marcia Da Silva*
Andrew Greaves*
Kinga James†
Health and Safety Manager
Susannah Rush
Health and Safety Officer
Mark Earthrowl
Purchasing and Supplies Manager
Janice Sutton
Building Services Facilities
Manager
Martin Frohock
*
joined in 2011
†
left in 2011
Major projects planned for this year include
the refurbishment of some of the tissue culture
facilities on the first floor. The team will be
liaising closely with the groups involved to
ensure as little disruption as possible. Working
with Property Services, we are in the process
of creating a new database to manage the
Institute’s asset register and associated service
contracts and documentation. The database
will also aid with the establishment of a new
management system as the Institute works
towards becoming GCP (Good Clinical Practice)
compliant.
Glasswash and Media
The Glasswash and Media Core Facility
provides a high quality, centralised glass
washing and sterile supplies service. We
play an essential role in supporting the
Institute’s research, by providing a range of
basic solutions and liquid/solid media, which
are replenished in the laboratories on a daily
basis. The team can also supply more complex
solutions and media to order. In addition, the
team provides a variety of general support
activities on the laboratory floors, such as
ordering compressed gas cylinders, waste
recycling and sterilisation of class 1 genetically
modified (GM) and containment level 2 (CL2)
waste, contributing positively to the site’s
environmental goals and initiatives.
In addition the team are responsible
for the overall management of the core
consumables supply chain for the Institute,
including laboratory tissue culture plastics and
consignment stocks primarily for tissue culture
reagents. Recently a vending machine has been
installed dispensing tissue culture reagents for
easy access.
We have had some personnel changes in the
last few months, with Andrew Greaves and
Marcia Da Silva joining the team, replacing
Ursula Clark and Kinga James who had been
part of the Institute for almost five years. The
Glasswash and Media Facility has recently
undertaken a survey within the institute, as
part of the Core Facility Review Process,
receiving excellent feedback and we continue
to look for new ways to improve and make
further additions to the service.
Health and Safety
We are committed to the continuous
improvement of the safety performance of the
Institute. Our philosophy is to encourage staff
to take ownership for their own safety, and
thereby create a positive safety culture. As a
Health and Safety team we help staff develop
pragmatic safety solutions that safeguard their
welfare, and also ensure regulatory compliance.
During 2011 we conducted an audit of the
safety management system and conducted
safety inspections with a view to improving
safety performance. We reviewed existing
safety policies, developed a number of new
policies, and delivered safety training. We also
conducted a series of safety inspections to
ensure that good safety standards were being
maintained in the laboratories.
In order to improve the understanding of safety
issues we have held three IOSH ‘Managing
Safety’ training sessions. More than 25 staff
have successfully completed the course. We
also held a ‘Health and Safety Awareness’ day to
further raise the awareness of health and safety.
In 2012 we will aim to continue to improve
safety performance at the CRI, and encourage
managers to promote good safety performance
within their departments through toolbox talks
and safety presentations. We will deliver safety
training to meet the needs of the Institute and
continue our inspection program. We also
intend to set up an internal audit program so
that we can monitor safety compliance more
effectively.
Procurement
The CRI procurement team is supported
by the Purchasing Operations Team, which
is based in London. Procurement helps
departments accomplish their objectives by
undertaking a range of activities that achieve
value for money. We do this through good
procurement practice and increased efficiency
whilst mitigating operational, commercial and
compliance risk to Cancer Research UK.
•We are working with key suppliers to set up
central discounted price agreements, reducing
Institute Administration | 93
Assistant Facilities Manager
Allan Graham†
Facilities Team Leader
Colin Weir
Property Services Administrator
Hannah Newell*
Assistant Facilities Coordinator
David Willsher
Facilities Operative
Anthony Millard
Security Supervisor
David Maguire
Porters
Helder Jose de Jesus Oliveira
Ligia Maria Rodrigues Ramos
Manuel da Silva†
Lead Building Technician
Tony Rose
Maintenance
Jim Baglee
Luke King
Jack Miles
Wayne Shelbourne
Darron Young
Catering Manager
Peter Houghton*
Assistant Catering Manager
Lynda Duck †
Head Chef
Adam Cattel
Restaurant
Tim Bygrave*
Lex Demydov †
Gergely Kiss
Mike Potter †
Parlagatan Sitorus
Paul Rowley
Mark Watson
Head of IT
Peter Maccallum
IT Staff
Beauty Bapiro
Richard Bemrose
Nigel Berryman
Karen Fernandes
Jason Flory †
Luis Huang
Naomi Neil*
Marc O’Brien
Dominic Oyeniran
the cost of consumable items.
•We are working with laboratory
management teams in London and Cambridge
to set up central service contracts.
•We are working across directorates on
a number of proposals to implement new
ways of buying goods and services into the
organisation, enabling further efficiencies
through increased automation of buying
processes.
•We are playing our part in helping Cancer
Research UK meet its environmental targets.
We are integrating sustainability into our
procurement process and are working with
our suppliers to minimise waste and packaging
and to support recycling schemes.
Property Services
Property Services adopt a Total Facilities
Management strategy, working towards
integrating the provision of all services to the
Institute. We are ambitious and progressive
and we work alongside the end-users of these
services to deliver and maintain a world-class
facility, whilst reducing our environmental and
economic impact wherever possible.
The Property Services team provide three core
services: maintenance, facilities and security.
Our key objective is to ensure the smooth, safe
and efficient running of Institute operations, in
order to support the CRI’s research efforts.
We ensure that the building is clean, well
maintained and secure for staff and visitors.
Property Services provide and administers
the following services: electrical maintenance,
cleaning, car parking, environmental and climate
control systems, post room, recycling, catering,
mechanical engineering, security, decorating
and repairs, furniture, energy management and
carbon reduction, waste removal and disposal,
reprographics and photocopying, catering and
hospitality, stationery.
The CRI hosts a large number of seminars
and events for staff and external visitors. The
Facilities Team manages the infrastructure and
the catering requirements for these meetings.
The team liaises with other Institutes on the
Cambridge Biomedical Campus to ensure that
the CRI is kept up to date with developments
on the site, and contributes to the site’s
environmental goals and initiatives.
IT and Scientific Computing
Researchers at the Institute depend on
information technology for nearly every aspect
of their work, from the collection and analysis of
experimental data to the development of new
*
joined in 2011
†
left in 2011
94 | Cambridge Research Institute Scientific Report 2011
theoretical techniques. The IT and Scientific
Computing Department provides technology
and expertise to support these needs.
The department has nine staff with a variety
of technical and programming skills to provide
a helpdesk, systems administration, database
support, application development, and design
and build services for new information systems.
They are drawn from a variety of scientific
computing and IT backgrounds with experience
in industry and academic research organisations.
The Institute’s scientists (and their laboratory
equipment) use a variety of Apple, Windows
and Linux desktops and laptops to support their
work, with Voice Over IP phone systems and
1 Gb network connectivity to labs and offices.
All users have access to backed-up network file
systems to store and share their data.
Around 20% of the researchers at the Institute
have roles which are primarily mathematical or
computational. Many of the technologies in use
(sequencing, proteomics, imaging, etc.) depend
on large scale computational data analysis,
with several terabytes of data being generated
each week. To support this the department
provides a blade-based computer cluster,
parallel file system storage for high throughput
analysis, mirrored disk storage for working data,
archive storage for long-term retention of raw
experimental data and final analysis results, and
servers for databases or web applications.
The expansion of data and computational
analysis continued through 2011. A rolling
programme of desktop and laptop replacement
was started to ensure that all researchers
have access to up to date personal computing
equipment and software. The department
expanded the cluster from 512 to 1300
computer cores; the parallel file system was
expanded from 48TB to 96TB; the archive was
expanded from 240TB to 480TB. New firewalls
and wireless networking equipment have been
purchased and work is underway to upgrade
the Institute’s network infrastructure.
Theses
The following CRI students submitted theses in 2011:
Tamir Chandra
PhD
Natalie Cook, Tuveson laboratory
The Notch pathway is a therapeutic target in
pancreatic ductal adenocarcinoma
Caryn Ross-Innes, Carroll and Odom
laboratories
Genomic approaches to understanding
oestrogen receptor alpha biology
Tamir Chandra, Narita laboratory
Chromatin dynamics in cellular senescence
Dominic Schmidt, Odom and Carroll
laboratories
Dynamics and evolution of vertebrate
transcriptional regulator binding
Sarah Jane Dawson, Caldas laboratory
Molecular biomarkers in breast cancer
Stephen Goldie, Watt laboratory
Studies of FRMD4A in a new model of head and
neck cancer
Stephen Goldie
Monika Golinska, Griffiths laboratory
The molecular and metabolic adaptations of
HIF-1b deficient tumour cells
Mahesh Iddawela, Caldas laboratory
Genome-wide copy number and gene
expression profiling using archived tissue for
molecular marker studies in breast cancer
Sarah Leigh-Brown
Sergii Ivakhno, Tavaré laboratory
Statistical framework for the analysis of copy
number aberrations in high-throughput cancer
genomic data
Alastair Lamb, Neal laboratory
A study of Hes6 as a transcriptional regulator in
castrate resistant prostate cancer
Naomi Sharma, Neal laboratory
The transcriptional role of the androgen
receptor in prostate cancer
Henning Stöckmann, Brindle laboratory and
Leeper laboratory, Department of Chemistry
The development of new agents for molecular
imaging in cancer
Chris Tape, Murphy laboratory
Discovery and development of therapeutic
TACE antibodies
Feng Wang, Brenton laboratory
The role of TGFBI in development and cancer
Julie Woolford, Tavaré laboratory and Miska
laboratory, Wellcome Trust/Cancer Research
UK Gurdon Institute
Statistical analysis of small RNA high-throughput
sequencing data
Sarah Leigh-Brown, Odom laboratory
The evolution of gene regulation in vertebrates
Caryn Ross-Innes
Charlotte Ng, Brenton laboratory
Tumour evolution in ovarian cancer using highthroughput genomics technologies
Roheet Rao, Neal laboratory
Understanding the functional role of HIP1 in
prostate cancer
Chris Tape
Theses | 95
Contact Details
Cancer Research UK Cambridge Research Institute
Li Ka Shing Centre
Robinson Way
Cambridge CB2 0RE
Telephone: +44 1223 404209
www.cambridgecancer.org.uk
An electronic copy of this report is available on our website.
Long Road
To City Centre
and Railway Station
(A1134)
ith’s Way
oa
d
on
R
Fe
nd
Adrian W
ay
Hutchison/MRC
Research Centre
Staff Car Park
h
Keit
Day
Cancer Research UK
Cambridge Research Institute
(Li Ka Shing Centre)
Main Entrance
Roa
Addenbrooke’s
Hospital
Main Visitor
Car Park
MRC Laboratory of
Molecular Biology
Bus stops
d
ical
Clin ol
o
Sch
N
ga
tin
ig h
u
en
Av
le
e
Ro
ad
Ward S4
l ls
Hi
Site of new
MRC Laboratory of
Molecular Biology
(opening 2012)
Wellcome Trust/MRC
Building (CIMR)
d
Goods
Entrance
be Way
a
Ro
Puddico
m
ls
Hil
Robinson W
ay
Queen Ed
Long Road
Sixth Form
College
1
(A
7)
30
a
y
Ave
nue
nW
inso
Rob
Francis Crick
ACCI/Phase I facility
C
Red
e
Gre
ds
m
30
7)
Cyclepath to
Great Shelford
By road: from M11 junction 11, follow the signs to Addenbrooke’s Biomedical Campus, or follow the signs to
Trumpington Park and Ride and take the Guided Bus.
By rail: take the train to Cambridge, then take the Stagecoach Citi 1 or Citi 7 bus to Addenbrooke’s.
By air: the nearest airport to the CRI is London Stansted Airport.
Cancer Research UK
Registered charity number in England and Wales: 1089464; in Scotland: SC041666
Registered as a company limited by guarantee in England and Wales number: 4325234
Registered address: Angel Building, 407 St. John Street, London EC1V 4AD
Telephone: +44 20 7242 0200
www.cancerresearchuk.org
96 | Cambridge Research Institute Scientific Report 2011
A1
d(
W ay
a
Ro
s
bin
on
aus
ewa
y
Strangeways
Research
Laboratory
aha
University of Cambridge
Forvie Site
Ro
To M11
Junction 11
Wo
r t’s
C
r
Bab
ity
n
ater
ie M al
Ros ospit
H
nlan
’s
oke
bro entre
C
den
Ad ment
at
Tre
Cyclepath to
Trumpington
e
Lan
ross
Oncology Centre
(Outpatients)
Cover images
Top
Primary human prostate cells grown in culture in
order to identify progenitor cells. Cells were
dissociated from benign patient tissue and were
seeded on growth medium, giving rise to the large
colony shown. The cells were stained for human
epithial markers cytokeratin 5 (green) and
cytokeratin 18 (red). Nuclear staining with DAPI
(blue). Image provided by Ajoeb Baridi (Stingl and
Neal laboratories).
Bottom
Podocyte cells wrap around the capillaries of the
glomerulus within the kidney and are a core element
of the filtration barrier that is the first stage in
removing waste products from the blood to form
urine. Podocytes play a critical role in the constant
turnover of the glomerular basement membrane as
well as endothelium maintenance by secreting
extracellular matrix components, vascular endothelial
growth factor (VEGF) and many other factors.
These cells stain positive for the recently described
TOR autophagy spatial coupling compartment
(TASCC). This intracellular ‘factory’ compartment
may conceivably allow them to maintain their high
levels of secretion. Image provided by Andy Young
(Narita laboratory).
Cancer Research UK Cambridge Research Institute
Scientific Report 2011
Editor: Laura Blackburn
Page setting: Charles D N Thomson
Cancer Research UK Cambridge Research Institute
Li Ka Shing Centre
Robinson Way
Cambridge CB2 0RE
ISSN 1756-8994
Copyright © 2011 Cancer Research UK
Cambridge Research Institute Scientific Report 2011
Cambridge
Research
Institute
Scientific Report 2011
Cancer Research UK Cambridge Research Institute
Li Ka Shing Centre
Robinson Way
Cambridge CB2 0RE
Telephone +44 (0) 1223 404209
www.cambridgecancer.org.uk