Download terry fox cancer research - Terry Fox Research Institute

Document related concepts
no text concepts found
Transcript
15 n
20 sio
r
ve
terry fox cancer research
portfolio 2015
Photo: Gail Harvey & The Terry Fox Foundation
TFRI_portfolio_2015.indd 1
15-10-28 2:24 PM
general overview
Terry Fox (1958-1981)
Terry Fox has become an inspiration to us all. Terry Fox was 18 when he was diagnosed with cancer
and lost one leg to it by amputation. In 1980 he started to run across Canada in his Marathon of
Hope to support cancer research. He wrote in his letter seeking support, “I will be ready to achieve
something that for me was once only a distant dream reserved for the world of miracles—to run across
Canada to raise money for the fight against cancer. We need your help. The people in cancer clinics all
over the world need people who believe in miracles.”
He has received many honours and awards and today is recognized by many as Canada’s greatest
hero. His legacy lives on through The Terry Fox Foundation, the millions of people who participate
in Terry Fox Runs around the world, the millions of generous donors worldwide who give to cancer
research, and the Terry Fox Research Institute.
The Terry Fox Foundation (TFF)
www.terryfox.org
The Terry Fox Foundation (TFF) maintains the vision and principles of Terry Fox while raising money
for cancer research through the annual Terry Fox Run, National School Run Day and other fundraising
initiatives. To date, over $700 million has been raised worldwide for cancer research in Terry Fox’s
name. The first Terry Fox Run was held in 1981, with The Terry Fox Foundation being created in 1988.
Its national headquarters are located in Burnaby, BC and it has offices in 9 provinces.
The Foundation invests in cure-oriented, biomedical research through its flagship program, The Terry
Fox New Frontiers Program Project Grants. It also supports capacity-building research through its New
Investigator awards. The Foundation research portfolio is managed by The Terry Fox Research Institute
and affiliated partners.
The Terry Fox Research Institute (TFRI)
www.tfri.ca
Launched in October 2007, The Terry Fox Research Institute (TFRI) is the brainchild of The Terry Fox
Foundation and today acts as its research arm, overseeing its complete portfolio of cancer research
projects. TFRI seeks to improve significantly the outcomes of cancer research for the patient through a
highly collaborative, team-oriented, milestone-based approach to research that will enable discoveries
to translate quickly into practical solutions for cancer patients worldwide. TFRI collaborates with over
73 cancer hospitals and research organizations across Canada. Headquartered in Vancouver, BC, the
Institute has six nodes across Canada which interact with regional partners and support the mission
and vision of the Institute.
Annual scientific participants visit the statue of Terry Fox during an early morning run in May 2015
TFRI_portfolio_2015.indd 2
15-10-28 2:24 PM
On the occasion of the 35th anniversary of the Marathon of Hope, TFRI’s 6th Annual Scientific
Meeting was held in St. John’s, Newfoundland, where Terry began his run in 1980.
The projects and publications listed in this summary of Terry Fox research represent some of
the best cancer research being conducted in Canada. Provided by our project leaders, these
scientific summaries describe research funded by The Terry Fox Foundation and its partners.
Funding partners are acknowledged for specific projects. We are deeply grateful to the patients
who participate in this research, and to our researchers, clinicians, scientists and their staff for
their dedication, expertise and commitment to making a difference for all cancer patients.
The Terry Fox Research Institute supports five areas of cancer research:
Terry Fox New Frontiers Program Project Grants These programs support Canadian
research teams exploring new frontiers in cancer research through the funding of three or more
outstanding independent research projects around a common theme. An open competition is
offered annually through TFRI to select the best program projects for funding.
Terry Fox New Investigator Awards These awards provide a three-year operating grant to
independent cancer researchers within the first five years of their first faculty appointment. New
Investigators are sponsored and mentored by an existing translational project or New Frontiers
Program Project team.
Terry Fox Translational Pan-Canadian Cancer Research These programs support Canadian
multidisciplinary teams to develop collaborative pan-Canadian research projects to align with
its translational research mandate. These projects mostly target a specific cancer with a focus
on moving discoveries and knowledge into practical solutions for patients within a relatively
short time frame. This is accomplished via an iterative process of developing milestone-driven
“business plans” to focus on the outcomes impact of the research. An open competition is
offered annually through TFRI to select the best project for funding. TFRI provides one award
per year.
Terry Fox Cancer Research Training Program TFRI currently supports five integrated training
programs across the country.
Terry Fox International Run Program Grants Globally, The Terry Fox Foundation fundraises
through its International Run Program. Funds raised support research projects in countries where
the run is held. www.terryfox.org/InternationalRun/
Oral poster presentation trainees are congratulated by Terry’s brother Darrell Fox (sixth from left) and Judith-Fox Alder (right), Terry Fox’s sister,
at our meeting in May 2015.
TFRI_portfolio_2015.indd 3
15-10-28 2:24 PM
table of contents
Terry Fox New Investigator Operating Grants
Auer, Rebecca
A personalized oncolytic vaccine: Using oncolytic viruses to exploit neo-antigens
derived from the tumour mutanome
1
Boutros, Paul
The systems biology of tumour hypoxia
3
Bridle, Byram
Evaluation of oncolytic immunotherapy in canine cancer trials: A stepping stone
towards successful translation into human patients
4
eciphering the role of chromatin demethylases in high-risk pediatric acute myeloid
D
leukemia (AML)
6
Hirst, Martin:
Epigenetic basis of myeloid malignancies
7
McCaffrey, Luke
Epithelial polarity in tumour invasion and metastasis
8
Morin, Ryan
Exploring clonal evolution in Non-Hodgkin lymphomas using serial tumour sampling
and liquid biopsies
9
Cellot, Sonia
O’Brien, Catherine
nderstanding cancer stem cell heterogeneity and dynamics: Implications for
U
therapy in human colorectal cancer
11
Rodier, Francis
Understanding the impact of cancer cell fate decisions during ovarian
cancer treatment
13
Shah, Sohrab
Are genomic instability and clonal diversity prognostic indicators of high-grade
serous ovarian cancer?
15
Stagg, John
The role of CD73-adenosinergic pathway in prostate cancer
16
Wilhelm, Brian
Transcriptional and epigenetic consequences of MLL-AF9 translocations
17
Zadeh, Gelareh
Exploring novel mechanisms of tumour vascularization in malignant brain tumours
19
Terry Fox New Frontiers Program Project Grants
Bell, John
The Terry Fox New Frontiers Program Project Grant: Canadian Oncolytic Virus
Consortium (COVCo)
20
Czarnota, Gregory
The Terry Fox New Frontiers Program Project Grant in ultrasound and MRI for
cancer therapy
22
he Terry Fox New Frontiers Program Project Grant: The development of stemnessT
based prognostic biomarkers and therapeutic targets
24
The Terry Fox New Frontiers Program in killing the hydra: Genetic dissection of
actionable targets required for maintenance of metastatic disease
25
Gascoyne, Randy
The Terry Fox New Frontiers Program Project Grant in molecular correlates of
treatment failure in lymphoid cancers
26
Giguère, Vincent
he Terry Fox New Frontiers Program Project Grant in oncometabolism and the
T
molecular pathways that fuel cancer
28
Dick, John
Egan, Sean
Gleave, Martin
The Terry Fox New Frontiers Program Project Grant in prostate cancer progression
30
Humphries, Keith
The Terry Fox New Frontiers Program Project Grant in core pathogenic pathways in
human leukemia
32
Huntsman, David
The Terry Fox New Frontiers Program Project Grant in the genomics of forme fruste
tumours: New vistas on cancer biology and management
33
The Terry Fox New Frontiers Program Project Grant in unraveling metabolic
adaptations associated with disease progression and therapeutic response in
metastatic breast cancer
35
Jones, Russell;
Park Morag
TFRI_portfolio_2015.indd 4
15-10-28 2:24 PM
Terry Fox New Frontiers Program Project Grants (continued)
Jones, Steven
The Terry Fox New Frontiers Program Project in discovery and therapeutic
development of antibody-based targets in oncology
36
Malkin, David
Li-Fraumeni Syndrome: Applying genetic determinants of cancer risk to cancer
surveillance and prevention
37
Nagy, Andras
The Terry Fox New Frontiers Program Project Grant in genetic analysis of signaling
pathways for vascular development and tumour angiogenesis
38
Paige, Christopher
The Terry Fox New Frontiers Program Project Grant in molecular and cellular
differentiation: New targets and treatments
39
Wilson, Brian
The Terry Fox New Frontiers Program Project in nanoparticle-enhanced
photoacoustic imaging for cancer localization and therapeutic guidance
41
Wouters, Brad;
Bristow, Robert
A research pipeline for hypoxia-directed precision cancer medicine
43
Terry Fox Research Institute Translational Cancer Research Projects
Babcook, John;
Rottapel, Robert
STP collaboration with the Centre for Drug Research and Development
Batist, Gerald;
Gallinger, Steven
Canadian Colorectal Cancer Consortium (C4)
Bell, John;
Chen, Pei-Jer
Development of new treatment and biomarker for hepatocellular carcinoma; From
woodchuck to human
47
Benard, François;
Kai Yuan Tzen
Development of 2-[18F]fluoro-2-deoxy-D-galactose as a new molecular imaging
probe for hepatocellular carcinoma diagnosis
48
Cairncross, Gregory
Modeling and therapeutic targeting of the clinical and genetic diversity of
glioblastoma
49
45
46
Humphries, Keith;
Tien, Hwei-Fang;
Kuo,Yuan-Yen;
Chou, Wen-Chien
Investigation of the pathogenesis of ASXL1 mutation in acute myeloid leukemia
Lam, Stephen;
Tsao, Ming
Pan-Canadian Early Lung Cancer Detection Study
Lam Stephen;
Lam, Wan;
Yan, Pan-Chyr;
Yu, Chong-Je
Translational research in lung cancer: From molecular markers/targets to
therapeutic applications
50
51
53
Mes-Masson, Anne-Marie; A pan-Canadian platform for the development of biomarker-driven subtype-specific
Rottapel, Robert
management of ovarian cancer
54
Mes-Masson, Anne-Marie; Selective Therapies Program collaboration: Therapeutic targets validation in ovarian
cancer
Provencher, Diane;
Huntsman David
55
Rosin, Miriam;
Poh, Catherine
Efficacy of optically guided surgery in the management of early-stage oral cancer:
The Canadian optically guided approach for oral lesions surgical (COOLS) trial
56
Saad, Fred
The Canadian Prostate Cancer Biomarker Network(CPCBN)
58
TFRI_portfolio_2015.indd 5
15-10-28 2:24 PM
A personalized oncolytic vaccine: Using oncolytic viruses to exploit neo-antigens derived
from the tumour mutanome
Terry Fox New Investigator Operating Grant (2012-2015)
Investigator: Rebecca Auer, Ottawa Hospital Research Institute
Mentoring Program: Canadian Oncolytic Virus Consortium (COVCo)
Scientific Summary: The immune system plays a central role in the cancer outcomes of most solid tumours,
in particular in the eradication of micrometastatic disease following surgical resection of the primary tumour.
Cancer immunotherapies must effectively target ‘self’-derived tumours while avoiding autoimmune side
effects, a potentially fatal penalty for effective immunotherapy. A recent focus in the field of cancer
immunotherapy is “immunomics”-combining immunology with genomics to identify neo-epitopes, based on
tumour-specific somatic mutations (the ‘mutanome’), with the goal of designing personalized cancer vaccines
that are less likely to produce autoimmune pathology.
The rapid advances in next-generation sequencing means that personalized cancer vaccines will be realized in
the next five years. A promising cancer vaccine platform is based on oncolytic viruses (OV). OVs selectively
replicate in tumour cells resulting in immunogenic cell death. Oncolytic vaccines (OV expressing tumour
antigens) provide an even more powerful boost to pre-existing antitumour immunity by combining viral
oncolysis with the presentation of tumour antigens. The Terry Fox Canadian Oncolytic Virus Consortium is a
leader in the development of oncolytic vaccines and combining this strategy with a personalized approach to
antigen selection, based on the tumour ‘mutanome’ represents a highly novel application of both
technologies. When used in combination with surgery, this cancer vaccine strategy has the potential to
impact over 65,000 Canadians who undergo surgical resection of their solid tumour every year.
Hypothesis: We hypothesize that the antitumoural immunity induced by viral oncolysis can be utilized to
identify immunogenic neo-antigens derived from the tumour ‘mutanome’. We further hypothesize that
immunizations with oncolytic vaccines expressing these mutated sequences will result in a therapeutic
antitumour immune response without development of autoimmunity and that this strategy can be
successfully combined with surgical resection of the primary tumour to eradicate micrometastatic disease.
Specific Aims:
(1) Determine the effect of tumour oncolysis on the in-vivo generation of T cell mediated immune responses
to mutant peptide neo-antigens derived from the B16 ‘mutanome’.
(2) Assess the therapeutic and autoimmune effects of a prime-boost vaccination strategy using a
recombinant adenovirus vaccine vector and a complementary replicating rhabdoviral oncolytic vaccine
expressing selected mutant peptide neo-antigens derived from the B16 ‘mutanome’.
(3) Evaluate a perioperative prime-boost immunization strategy to treat metastatic disease in combination
with surgery in a B16 tumour model
Methods: In this project we will make use of the murine B16 melanoma model, one of the most widely used
models for scientific validation of T cell-based immunotherapies, and for which the ‘mutanome’ has been
published. This project will establish the pre-clinical ‘proof of concept’ for a personalized oncolytic
vaccination strategy that can ultimately be employed in patients following deep sequencing of their own
tumour.
1
List of Key Publications:
1.
Ananth AA, Tai LH, Alkayyal A, Zhang J, Tanese de Souza C, Stephenson K, Pol J, Parato, K, Stojdl DF, Atkins HL, Bell JC,
Lichty BD, Auer RC*. Surgical stress attenuates pre-existing anti-tumour immunity resulting in postoperative metastases and
local recurrence in a murine model. PLOS One (in revision Sept 2015).
2.
Tai LH, Tanese de Souza C, Sahi S, Zhang J, Alkayyal A, Ananth A, Auer RC*. A Mouse Tumor Model of Surgical Stress to
Explore the Mechanisms of Postoperative Immunosuppression and Evaluate Novel Perioperative Immunotherapies. J
Visualized Experiments 2014;85. IF: 1.9
Zhang J, Tai LH, Ilkow C, Alkayyal AA, Abhirami AA, Tanese de Souza C, Lefevbre C, Falls TJ, Bell JC, Stojdl D, Auer RC*.
Preventing metastases by perioperative targeting of natural killer cell dysfunction with Maraba MG1 virotherapy. Mol Ther
2014;22(7):1320-32. IF:6.825
3.
4.
Tai LH, Auer R*. Attacking postoperative metastases using perioperative oncolytic viruses and viral vaccines. Front Oncol
2014;12(4):217.
5.
Tai LH, Zhang J, Auer RC*. Preventing surgery-induced NK cell dysfunction and cancer metastases with influenza
vaccination. OncoImmunology 2013;2(11):e26618.
6.
Tai LH, Zhang J, Tanese de Sousa C, Alkayyal A, Ananth AA, Sahi S, Mahmoud AB, Bell JC, Makrigiannis A, Auer RC*.
Perioperative influenza vaccination reduces postoperative metastatic disease by reversing surgery-induced dysfunction in
natural killer cells. Clin Cancer Res 2013;19(18):5104-15. IF:7.742
7.
Seth R, Tai LH, Falls T, Tanese de Sousa C, Bell J, Carrier M, Atkins H, Boushey R, Auer RC. Surgical stress promotes the
development of cancer metastases by a coagulation-dependent mechanism involving Natural-Killer cells in a murine model.
Ann Surg 2013; 258(1):158-68. IF:6.329
8.
Tai LH, Tanese de Sousa C, Rintoul J, Ly L, Zhang J, Falls TJ, Belanger S, Bell JC, Makrigiannis A, Auer RC. Preventing
postoperative metastases by enhancing natural killer cell function with novel oncolytic virus therapy. Cancer Res
2012;73(1):97-107. IF:8.650
9.
Lemay CG, Rintoul JL, Kus A, Paterson JM, Garcia V, Falls TJ, Ferreira L, Brindle BW, Conrad DP, Tang VA, Diallo JS,
Arulanandam R, LeBoef F, Stojdl DF, Lichty BD, Atkins HL, Parato KA, Bell JC, Auer RC. Harnessing oncolytic virusmediated anti-tumour immunity in an infected cell vaccine. Mol Ther 2012;20(9):1791-9. IF:7.041
10. Auer RC, Bell JC. Oncolytic viruses: smart therapeutics for smart cancers. Future Oncol. 2012 Jan;8(1):1-4. IF:2.455
11. Rintoul JL, Lemay CG, Tai LH, Stanford MM, Falls TJ, de Souza C, Bridle BW, Ohashi PS, Wan Y, Lichty BD, Mercer AA,
Auer RC, Atkins HL, Bell JC*. ORFV:A Novel Oncolytic and Immnune Stimulating Parapoxvirus Therapeutic. Mol Ther
2012;20(5):1148-57. IF:7.041
2
The systems biology of tumour hypoxia
Terry Fox New Investigator Operating Grant (2014-2016)
Investigator: Dr. Paul C. Boutros, Ontario Institute for Cancer Research
Mentoring Program: A research pipeline for hypoxia-directed precision cancer medicine
Scientific Summary: Human cancers reside in a local micro-environment unlike that of any normal tissue. Through a variety of
processes, tumour cells interact with and modulate their immediate micro-environment. These micro-environments contain
areas of very low oxygen concentration, called hypoxic regions, which can contain cells that have adapted to severe metabolic
stressors and are resistant to cytotoxic chemotherapy and radiotherapy. These cells can also acquire genetic instability and
lead to an increased capacity for metastatic spread. A greater understanding of tumour hypoxia within a clinical context is
critical to improving outcomes across multiple cancer types. To achieve this, I am creating computational techniques that relate
the molecular characteristics of hypoxic cells and their surrounding micro-environment to clinical heterogeneity in patient
response to therapy and overall prognosis.
Aims: The first key aim of this project is to develop ways to accurately predict specific micro-environmental phenomena from
molecular data. We are taking two complementary approaches to this problem: one based on using Bayesian probabilistic
models to distinguish distinct cellular populations, and the other employing genomic data to predict specific microenvironmental characteristics. The second key aim of this project is to create joint micro-environmental-molecular models to
predict patient outcome and response to therapy. We are extending our previous graph-theory- based approaches for
biomarker prediction, integrating known functional pathways with micro-environmental and other molecular characteristics.
Validation of both aims is occurring with new datasets being generated by the TFRI Tumour Hypoxia Program Project.
Updates: We have created a way of merging tumour microenvironmental and genomic data into a model that very accurately
predicts patient survival. This model is now undergoing validation in new cohorts in a CLIA setting, with the aim of it being
used in routine clinical practice. We also developed a series of computational tools that are now in wide-use by scientists both
within and outside of Canada to improve the way that they analyze cancer genomic data. Finally, we created the first spatial
map of genomic heterogeneity within prostate cancer, giving specific guidance on how biomarkers for personalized medicine
can account for this inherent variability in tumours.
List of Key Publications:
3
Evaluation of oncolytic immunotherapy in canine cancer trials: A stepping stone towards
successful translation into human patients
Terry Fox New Investigator Operating Grant (2015-2018)
Investigator: Byram W. Bridle, University of Guelph
Mentoring Program: Canadian Oncolytic Virus Consortium (COVCo)
Collaborators: J. Paul Woods, Anthony Mutsaers and Geoffrey Wood, University of Guelph; Jean-Simon Diallo, Hesham
Abdelbary and Joel Werier, Ottawa Hospital Research Institute
Scientific Summary: Osteosarcoma (OSA) is the most common form of primary bone cancer. When patients present
with metastases, the prognosis is dismal despite aggressive surgical resection and chemotherapy. Dogs have a 10-fold
higher incidence of OSA and its pathogenesis closely mimics the human disease, with a similarly poor prognosis. The
hypothetical basis of this proposal is that adjunct oncolytic immunotherapy can augment the standard of care in OSA
patients. The applicant has published a strategy to synergize cancer immunotherapy that directs the power of the
immune system against tumours, with oncolytic virotherapy that utilizes viruses that replicate in and kill only cancerous
cells. This synergy could be achieved by using an oncolytic rhabdovirus as a tumour-associated, antigen-expressing
vaccine to boost adenovirus-primed, tumour-specific immune responses. The primary objective of this proposal is to
test this novel biotherapy in the context of a clinical canine OSA trial. The long-term goal is to refine the approach for
translation into human patients. Primary, secondary and tertiary endpoints of the trial are induction of detectable
OSA-specific T-cell responses and increase median progression-free and overall survival between dogs receiving the
standard of care with adjunct oncolytic immunotherapy versus those treated with the standard of care alone.
Predicted benefits include prolonged remission time, extended survival and enhanced quality of life. Attempts will be
made to correlate tumour-antigen expression, intra-tumoural gene and cytokine signatures with clinical outcomes in
an effort to identify novel biomarkers and new therapeutic targets. Two small-scale projects will assess the potential
benefits of incorporating histone deacetylase inhibition or treatment with liposome-encapsulated clodronate into the
oncolytic vaccine therapy. The goal of these pre-clinical projects is to develop more advanced forms of the therapy
that can be tested in future canine clinical trials. Access to the state-of-the-art Animal Cancer Centre at the University
of Guelph, which has a huge catchment area, combined with the logistical infrastructure, intellectual property (e.g. the
potential to license novel veterinary therapeutics) and expertise developed through this project will be a valuable and
unique Canadian resource that can be leveraged by the sponsoring partner (COVCo) and TFRI to facilitate translation of
the most promising and highly refined biotherapeutics into human cancer patients.
Aims:
1. Primary objective: Evaluate the efficacy of an MG1-vectored oncolytic booster vaccine in a canine osteosarcoma
clinical trial.
2. Pipeline project #1: Test a more advanced iteration of the oncolytic vaccine therapy that incorporates histone
deacetylase inhibition. This study will determine a safe dose of the MG1 oncolytic booster vaccine that can be used in
combination with the HDI entinostat in a dose-escalation study in Ad-vaccinated purpose-bred research dogs. The
objective is to develop the rationale to propose testing of a next-generation therapy in a new clinical canine
osteosarcoma trial at the time of renewal of this grant.
3. Pipeline project #2: Use liposome-encapsulated clodronate to deplete splenic marginal zone macrophages to
simultaneously enhance oncolytic vaccine-induced secondary T-cell responses, improve intratumoural delivery of the
virus, remove immunosuppressive myeloid-derived suppressor cells and deplete osteoclasts that cause bone loss and
spontaneous fractures in osteosarcoma patients. This represents an extremely novel pre-clinical project and aims to
generate data to inform the design of a future study in purpose-bred research dogs.
Updates: Although funding only commenced three months ago, a PhD-level graduate student and postdoctoral fellow
have been recruited to conduct research. In addition, two summer students were hired to assist with the research.
4
The graduate student has begun constructing the viral vectors needed for the pre-clinical, mouse-based project that
has been proposed. The postdoctoral researcher is constructing the viruses needed for the clinical canine
osteosarcoma trial. One of the summer students optimized a flow cytometry-based method to quantify activated Tcells. This will be used to monitor immune responses in the upcoming dog trial. The other summer student confirmed,
by Western blotting, that our proposed target antigen (survivin) is highly expressed by osteosarcomas in our
companion animal tumour bank but not matched normal tissues. We have also optimized a method to transiently
deplete splenic marginal zone macrophages as well as the method needed to assess this. Once viral vectors are
rescued, the next steps will include: working with collaborators to produce veterinary clinical trial-grade batches and
then assessing their safety in a Canadian Food Inspection Agency/Canadian Centre for Veterinary Biologics approved
study.
List of Key Publications:
1.
Pol JG, Zhang L, Bridle BW, Stephenson KB, Rességuier J, Hanson S, Chen L, Kazdhan N, Bramson JL, Stojdl DF, Wan Y, Lichty
BD. Maraba virus as a potent oncolytic vaccine vector. Molecular Therapy. 2014 Feb;22(2):420-9.
2.
Bridle BW, Clouthier D, Zhang L, Pol J, Chen L, Lichty BD, Bramson JL, Wan Y. Oncolytic vesicular stomatitis virus quantitatively
and qualitatively improves primary CD8+ T-cell responses to anticancer vaccines. Oncoimmunology. 2013 Aug 1;2(8):e26013.
3.
Kim JJ, Bridle BW, Ghia JE, Wang H, Syed SN, Manocha MM, Rengasamy P, Shajib MS, Wan Y, Hedlund PB, Khan WI. Targeted
inhibition of serotonin type 7 (5-HT7) receptor function modulates immune responses and reduces the severity of intestinal
inflammation. Journal of Immunology. 2013 May 1;190(9):4795-804.
4.
Zhang L, Bridle BW, Chen L, Pol J, Spaner D, Boudreau JE, Rosen A, Bassett JD, Lichty BD, Bramson JL, Wan Y. Delivery of
viral-vectored vaccines by B cells represents a novel strategy to accelerate CD8(+) T-cell recall responses. Blood. 2013 Mar
28;121(13):2432-9.
5.
Bridle BW, Chen L, Lemay CG, Diallo JS, Pol J, Nguyen A, Capretta A, He R, Bramson JL, Bell JC, Lichty BD, Wan Y. HDAC
inhibition suppresses primary immune responses, enhances secondary immune responses, and abrogates autoimmunity during
tumor immunotherapy. Molecular Therapy. 2013 Apr;21(4):887-94.
6.
Lemay CG, Rintoul JL, Kus A, Paterson JM, Garcia V, Falls TJ, Ferreira L, Bridle BW, Conrad DP, Tang VA, Diallo JS,
Arulanandam R, Le Boeuf F, Garson K, Vanderhyden BC, Stojdl DF, Lichty BD, Atkins HL, Parato KA, Bell JC, Auer RC.
Harnessing Oncolytic Virus-mediated Antitumor Immunity in an Infected Cell Vaccine. Molecular Therapy. 2012 Sep;20(9):17919.
7.
Rintoul JL, Lemay CG, Tai L-H, Falls TJ, de Souza CT, Bridle BW, Stanford MM, Ohashi PS, Wan Y, Lichty BD, Mercer AA, Auer
RC, Atkins HL, Bell JC. ORFV: A Novel Oncolytic and Immune Stimulating Parapoxvirus Therapeutic. Molecular Therapy. 2012
Jan 24.
8.
Eftimie R, Dushoff J, Bridle BW, Bramson JL, Earn DJD. Multi-stability and multi-instability phenomena in a mathematical
model of tumor-immune-virus interactions. Bulletin of Mathematical Biology. 2011 Dec; 73(12):2932-61.
9.
Bridle BW. Neuroendocrine cancer vaccines in clinical trials. Expert Reviews in Vaccines. 2011 Jun;10(6):811-23. Invited review.
10. Bridle BW, Stephenson KB, Boudreau JE, Koshy S, Kazdhan N, Pullenayegum E, Brunellière J, Bramson JL, Lichty BD, Wan Y.
Potentiating cancer immunotherapy using an oncolytic virus. Molecular Therapy. 2010 Aug;18(8):1430-9.
11. Bridle BW, Hanson S, Lichty BD. Combining oncolytic virotherapy and tumour vaccination. Cytokine Growth Factor Reviews.
2010; 21(2-3):143-8. Invited review.
12. Bridle BW, Li J, Jiang S, Chang R, Lichty BD, Bramson JL, Wan Y. Immunotherapy can reject intracranial tumor cells without
damaging the brain despite sharing the target antigen. Journal of Immunology. 2010 Apr 15;184(8):4269-75.
13. Bridle BW, Boudreau JE, Lichty BD, Brunellière J, Stephenson K, Koshy S, Bramson JL, Wan Y. Vesicular stomatitis virus as a
novel cancer vaccine vector to prime antitumor immunity amenable to rapid boosting with adenovirus. Molecular Therapy. 2009
Oct;17(10):1814-21.
14. Boudreau JE, Bridle BW, Stephenson KB, Jenkins KM, Brunellière J, Bramson JL, Lichty BD, Wan Y. Recombinant vesicular
stomatitis virus transduction of dendritic cells enhances their ability to prime innate and adaptive antitumor immunity.
Molecular Therapy. 2009 Aug;17(8):1465-72.
15. Grinshtein N, Bridle B, Wan Y, Bramson JL. Neoadjuvant vaccination provides superior protection against tumor relapse
following surgery compared with adjuvant vaccination. Cancer Research. 2009 May 1;69(9):3979-85.
5
Deciphering the role of chromatin demethylases in high-risk pediatric acute myeloid
leukemia
Terry Fox New Investigator Operating Grant (2015-2017)
Investigator: Sonia Cellot, CHU Ste-Justine
Mentoring Program: Core pathogenic pathways in human leukemia
Funding Partner: The Cole Foundation, La Fondation CHU Sainte-Justine, La Fondation du Centre de Cancérologie
Charles-Bruneau
Scientific Summary: Chromatin methylation patterns impact on cell fate decisions, such as stem cell self-renewal and
differentiation, in both hematopoietic stem cells (HSCs) and leukemia stem cells (LSCs). Regulators of chromatin
methylation dynamics are involved in normal blood development, and their perbutation can lead to cancer. This is best
exemplified by the histone methyl transferase MLL (mixed lineage leukemia), which is critical in regulating normal HSC
fate decisions, while mutations in the MLL gene are found in more than 70% of infant leukemia. Like MLL, several
epigenetic regulators are deregulated in aggressive forms of leukemia. Epigenetic regulators are under active
pharmacological scrutiny as promising drug targets in leukemia, and chemical probes now become available at a rapid
pace. Pediatric acute myeloid leukemia (AML) is a heterogeneous disease with sub-optimal survival rates. Using an
RNAi- and chemical-based screening strategy, we want to identify demethylases that are critical to sustain leukemia,
and assess their their role in normal HSC fate regulation. The research program greatly benefits from synergistic
collaborations with stem cell biology leaders and members of the thematically related TFRI Program Project Grant in
Core Pathogenic Pathways in Human Leukemia, led by Dr. K. Humphries (BC Cancer Agency).
Aims: To develop a functional screening platform using RNAi- and chemical-based approaches to assess the role of
epigenetic regulators, and in particular chromatin demethylases, in the context of pediatric acute myeloid leukemia.
We anticipate that AML subtypes will display specific demthylase dependencies, given the heterogeneity of the
disease, and in particular the distinct entity that represents pediatric leukemia. A counterscreen will be performed
using cord blood isolated HSC, to gauge for potential toxicity of demethylase inhibition on the normal hematopoietic
system, a critical step in drug target identification. Identified hits will be validated in vivo using xeno-transplantation
based assays. Epigenetic landscapes and expression profiles of HSC and LSC will be determined.
Updates: We have performed functional genetics screens to identify HSC regulators, using a pipeline of standardized
HSC isolation, transduction and transplantation strategies. Among the Jumonji family of histone demethylases, we
identified both positive (Jhdm1f/Phf8) and negative (Kdm5b/Jarid1b) regulators of mouse HSC activity, through an
RNAi-based approach. These studies suggest that Jarid1b contributes to transcriptional repression of stemness
associated genes and promotes cell differentiation. We now want to identify demethylases that modulate human
HSC/LSC fate, using newly optimized culture conditions that sustain human cells ex vivo and novel high-risk acute
myeloid leukemia (AML) models. For epigenetic and expression profile studies of AML specimens, we will exploit the
pediatric branch of the Quebec Leukemia Cell Bank (www.bclq.gouv.qc.ca). Overall, we aim to identify pediatric
leukemia specific drug targets, and investigate the role demethylases in normal human HSC.
List of Key Publications:
Cellot S, Hope KJ, Chagraoui J, Sauvageau M, Deneault E, Macrae T, Mayotte N, Wilhelm BT, Landry JR, Ting SB, Krosl J, Humphries
K, Thompson A, Sauvageau G. RNAi screen identifies Jarid1b as a major regulator of mouse HSC activity. Blood. 2013. Aug
29;122(9):1545-55.
Hope KJ, Cellot S, Ting SB, MacRae T, Mayotte N, Iscove NN, Sauvageau G. An RNAi screen identifies Msi2 and Prox1 as having
opposing roles in the regulation of hematopoietic stem cell maintenance. Cell Stem Cell. 2010 Jul 2; 7(1):101-13.
Denault É, Cellot S, Faubert A, Laverdure J-P, Fréchette M, Chagraoui J, Mayotte N, Sauvageau M, Ting S.B, and Sauvageau G. A
Functional Screen to Identify Novel Effectors of Hematopoietic Stem Cell Activity. Cell. 2009 Apr 17; 137(2):369-79.
6
Epigenetic basis of myeloid malignancies
Terry Fox New Investigator Operating Grant (2015-2017)
Investigators: Martin Hirst, UBC; R. Keith Humphries, Aly Karsan, BC Cancer Agency;
Mentoring Program: Core pathogenic pathways in human leukemia
Funding Partner: BC Cancer Foundation
Scientific Summary: Acute myeloid leukemia (AML) remains one of the most lethal of adult malignancies, with longterm survival rates of <20% in patients under 65 and little improvement in treatment options for several decades. The
goal of our research is to dissect the role of the epigenome in the pathogenesis of primary AML and quantitate the
molecular and phenotypic changes associated with exposure to ascorbic acid (Vitamin C) in the context of AML
harbouring inactivating TET or neomorphic IDH mutations. Comprehensive and quantitative epigenetic profiling of
clinically annotated and genotyped primary leukemic cells combined with the study of an essential nutrient recently
shown to have the capacity to activate TET will enhance our understanding of the mechanisms of epigenetics in cancer
and the potential to reverse genetically driven epi-mutations.
We have recently demonstrated that vitamin C alters DNA methylation homeostasis and, in turn, the expression of
specific genes by enhancing TET activity. In a TET dependent manner, vitamin C reduces CpG methylation at CpG
islands (CGIs) via a hydroxymethyl-cytosine intermediate promoting a more primitive DNA methylation state in normal
hematopoietic stem cells. My hypothesis is that vitamin C will partially or completely reverse epigenetic effects
specifically of heterozygous TET and IDH mutations in AML via activation of the remaining wild type TET protein and
hmC driven demethylation of aberrantly methylated CGIs. If successful, our research will provide evidence for a new
class of therapeutics that act by upregulating endogenous epigenetic pathways and the opportunity of rapid
translation for Canadian patients diagnosed with this deadly cancer type.
List of Key Publications:
1. Adam Deveau, A. Forrester, Andrew Coombs, Gretchen Wagner, Clemens Grabher, Ian Chute, Daniel Léger, Matthew
Mingay, Gabriela Alexe, Vinothkumar Rajan, Robert Liwski, Martin Hirst, Kimberly Stegmaier, Stephen Lewis, Thomas
Look, and Jason Berman. Epigenetic therapy restores normal hematopoiesis in a zebrafish model of NUP98-HOXA9-induced
myeloid disease. Leukemia, Leukemia. 2015 May 28. doi: 10.1038/leu.2015.126. PMID: 26017032.
2. Mingay M, Hirst M. The Epigenomic Toolkit. Drug Discovery Today: Disease Models. DOI: 10.1016/j.ddmod.2014.05.004.
Epub 2014 August 22.
3. Nguyen LV, Makarem M, Carles A, Moksa M, Kannan N, Pandoh P, Eirew P, Osako T, Kardel M, Cheung AM, Kennedy W,
Tse K, Zeng T, Zhao Y, Humphries RK, Aparicio S, Eaves CJ, Hirst M. Clonal Analysis via Barcoding Reveals Diverse Growth
and Differentiation of Transplanted Mouse and Human Mammary Stem Cells. Cell Stem Cell.2014 Feb 6;14(2):253-63. doi:
10.1016/j.stem.2013.12.011. Epub 2014 Jan 16.PMID: 24440600
4. Xiang P, Wei W, Lo C, Rosten P, Hou J, Hoodless PA, Bilenky M, Bonifer C, Cockerill PN, Kirkpatrick A, Gottgens B, Hirst M,
Humphries KR. Delineating MEIS1 cis-regulatory elements active in hematopoietic cells.Leukemia.2013 Oct 7.doi:
10.1038/leu.2013.287. [Epub ahead of print].PMID:24097337
5. Blaschke K, Ebata KT, Karimi MM, Zepeda-Martínez JA, Goyal P, Mahapatra S, Tam A, Laird DJ, Hirst M, Rao A, Lorincz
MC, Ramalho-Santos M. Vitamin C induces Tet-dependent DNA demethylation and a blastocyst-like state in ES cells.
Nature. 2013 Aug 8;500(7461):222-6. PMID: 23812591
6. Heuser M, Yun H, Berg T, Yung E, Argiropoulos B, Kuchenbauer F, Park G, Hamwi I, Palmqvist L, Lai CK, Leung M, Lin G,
Chaturvedi A, Thakur BK, Iwasaki M, Bilenky M, Thiessen N, Robertson G, Hirst M, Kent D, Wilson NK, Göttgens B, Eaves C,
Cleary ML, Marra M, Ganser A, Humphries RK. Cell of Origin in AML: Susceptibility to MN1-Induced Transformation Is
Regulated by the MEIS1/AbdB-like HOX Protein Complex. Cancer Cell. 2011 Jul 12;20(1):39-52. PMID: 21741595
7
Epithelial polarity in tumour invasion and metastasis
Terry Fox New Investigator Operating Grant (2011-2015)
Investigator: Luke McCaffrey, McGill University
Mentoring Program: Unraveling metabolic adaptations associated with disease progression and therapeutic response in
metastatic breast cancer
Scientific Summary: Under the mentorship of the Preclinical Models and Therapeutic Targets for Metastatic Breast Cancer
Program, our objective is to build a molecular understanding of how cell polarity regulates invasive and metastatic breast
cancer with the goal of identifying novel prognostic markers and therapeutic targets. Metastatic progression correlates
strongly with loss of tissue structure and organization; accordingly, cell polarity proteins are frequently disrupted in tumours,
but the role of these key regulators in breast cancer progression is not understood. Accumulating evidence indicates that
atypical protein kinase C (aPKC) isoforms, which are key transducers of polarity signalling, are disrupted in breast cancer and
may have either oncogenic or tumour suppressive functions. Furthermore, we have found that the Par3 polarity protein is a
key regulatory of aPKC in normal and tumourigenic epithelial cells. In this project we are further exploring the contribution of
Par3 and aPKC in breast tumour progression. To address the objectives of our project, we have three primary questions:
 What are the mechanisms by which Par3/aPKC polarity organizes the mammary epithelium, and how this is disrupted in
breast cancer?
 How do aPKC isoforms promote or suppress breast cancer progression?
 Does aPKC polarity mediate cancer stem cell activity to control the differentiation state of breast tumours?
Key Findings: We found that Par3 silencing dramatically reduced tumour latency in breast cancer mouse models and
produced invasive and metastatic tumours that retained epithelial marker expression. Par3 depletion was associated with
induction of matrix metalloproteinases, destruction of the extracellular matrix, and invasion, all mediated by atypical PKCdependant JAK/Stat3 activation. Importantly, we found that Par3 expression is significantly reduced in human breast cancers,
which correlates with active aPKC and Stat3. These data identify Par3 and aPKC as regulators of signaling pathways relevant
to invasive breast cancer.
List of Key Publications:
1.
McCaffrey L, Macara IG. The Par3/aPKC interaction is essential for end bud remodelling and progenitor differentiation during
mammary gland morphogenesis. Genes Dev: 23(12) 1450-60 (2009).
2.
McCaffrey L, Macara IG. Widely conserved signalling pathways in the establishment of cell polarity. Cold Spring Harb Perspect
Biol: 1(2) 1-17 (2009).
3.
McCaffrey L, Macara IG. Epithelial organization, cell polarity, and Tumourigenesis. Trends Cell Biol: 21(12) 625-680 (2011).
4.
McCaffrey L, Montalbano J, Mihai C, Macara IG. Loss of the Par3 Polarity Protein Promotes Breast Tumourigenesis and
Metastasis. Cancer Cell: 22, 601-614 (2012).
5.
McCaffrey L, Macara IG. Signaling Pathways in Cell Polarity. Cold Spring Harb Perspect Biol. 4(6): 1-15 (2012).
6.
Macara IG, McCaffrey L. Cell Polarity in Morphogenesis and Metastasis. Phil Trans R Soc B (In Press).
8
Exploring clonal evolution in non-Hodgkin lymphomas using serial tumour sampling and
liquid biopsies
Terry Fox New Investigator Operating Grant (2015-2017)
Investigator: Ryan Morin, Simon Fraser University
Mentoring Program: Molecular correlates in treatment failure in lymphoid cancers
Funding Partner: BC Cancer Foundation
Scientific Summary: We are studying the patterns of clonal evolution in aggressive non-Hodgkin lymphomas (NHLs)
including diffuse large B-cell lymphoma (DLBCL) and mantle cell lymphoma (MCL). Substantial effort to date has been
placed on understanding the genes that are commonly mutated in these two deadly cancers but little is currently
known regarding clonal evolution patterns under the selective pressure applied by the standard treatments, such as
chemotherapeutic agents and immunotherapy. We are performing genomic analysis of serial tumour biopsies to infer
these changes and patterns of clonal evolution in individual patients. We are also seeking evidence for genes that are
more commonly mutated at relapse due to clonal selection. In parallel, we are implementing methods for detecting
and tracking somatic mutations non-invasively using a method known as “liquid biopsies” that involves sequencing
genomic DNA shed from tumour cells that is present in blood plasma.
Aims: We aim to identify genes that are more commonly mutated at relapse in MCL and DLBCL and to determine the
means by which these become enriched at relapse by more general clonal analysis of these tumours. Clonal
architecture will be determined through computational analysis of bulk tumour sequencing data and singl- cell analysis
of disassociated tumour tissue. Liquid biopsies have shown utility for identifying clonal mutations in a variety of solid
tumours but little is known regarding their utility in detecting and accurately representing mutations present at
different clonal frequencies. We aim to firmly establish the extent to which clonal and subclonal mutations can be
detected and differentiated via liquid biopsies in these NHLs.
Updates: We have analyzed 38 uniformly treated DLBCL tumour biopsies collected at relapse using exome sequencing
and have identified the commonly mutated genes. For a subset of these cases, we sequenced matched diagnostic
tumour DNA and observed patterns of clonal evolution in some of these patients. By comparing mutation prevalence
to published cohorts, three genes were found significantly enriched for mutations at relapse. We found evidence for
clonal enrichment of mutations in several of these genes in one or more patient, suggesting that aggressive subclones
that contribute to relapse exist in some patients at the time of diagnosis. We are also studying liquid biopsies from
each of these patients to determine the correspondence of mutation abundance in the plasma and tumour. In parallel,
we have begun characterizing additional MCL patients using exome sequencing and single-cell analysis and await
subsequent blood and biopsy samples from these patients for liquid biopsy applications.
List of Key Publications:
1.
Ryan D Morin, Sarit Assouline, Miguel Alcaide, Arezoo Mohajeri, Rebecca L Johnston, Lauren Chong, Jasleen Grewal,
Stephen Yu, Daniel Fornika, Kevin Bushell, Torsten H Nielsen, Tina Petrogiannis-Haliotis, Michael Crump, Axel Tosikyan,
Bruno Grande, David Macdonald, Caroline Rousseau, Maryam Bayat, Pierre Sesques, Remi Froment, Marco Albuquerque,
Yuri Monczak, Kathleen Klein-Oros, Celia Greenwood, Yasser Riazalhosseini, Madeleine Arseneault, Errol Camlioglu, Andre
Constantin, Pan Qiang-Hammarstrom, Qiang Roujun Peng, Koren K Mann, and Nathalie A Johnson. Genetic landscapes of
relapsed or refractory diffuse large B cell lymphomas. Blood (Submitted).
2.
Sarit Assouline, Michael Crump, Torsten Holm Nielsen, Lauren Chong, MIGUEL ALCAIDE, David MacDonald, Axel
Tosikyan, Abbas Kezouh, Vishal Kukreti, Tina Petrogiannis-Haliotis, Rémi Froment, Celia Greenwood, Kathleen Klein Oros,
Errol Camlioglu, Ayushi Sharma, Wilson H. Miller, Rosa Christodoulopoulos, Caroline Rousseau, Nathalie A Johnson, Ryan
D Morin, and Koren Kathleen Mann. A randomized, phase II study with biomarker analysis of panobinostat +/- rituximab in
relapsed diffuse large B cell lymphoma. Blood (Submitted).
3.
Lim EL, Trinh DL, Scott DW, Chu A, Krzywinski M, Robertson G, Mungall AJ, Schein J, Boyle M, Johnson NA, Steidl C,
Connors JM, Morin RD, Gascoyne RD and Marra MA. Comprehensive miRNA Sequence Analysis Reveals Survival
Differences in Diffuse Large B-cell Lymphoma Patients. Genome Biology. 2015, 16:18.
9
4.
David D W Twa, Anja Mottok, Fong Chun Chan, Susana Ben-Neriah, Bruce W Woolcock, King L Tan, Andrew J Mungall,
Helen McDonald, Yongjun Zhao, Raymond S Lim, Brad H Nelson, Katy Milne, Sohrab P Shah, Ryan D Morin, Marco A
Marra, David W Scott, Randy D Gascoyne and Christian Steidl. Genomic rearrangements involving programmed death
ligands are recurrent in primary mediastinal large B-cell lymphoma. Blood 123, 2062–2065 (2014).
5.
Morin, R. D. & Gascoyne, R. D. Newly identified mechanisms in B-cell non-Hodgkin lymphomas uncovered by nextgeneration sequencing. Semin Hematol 50, 303–313 (2013).
6.
Trinh, D. L. et al. Analysis of FOXO1 mutations in diffuse large B-cell lymphoma. Blood (2013).
7.
Morin, R. D. et al. Mutational and structural analysis of diffuse large B-cell lymphoma using whole genome sequencing.
Blood 122, 1256–1265 (2013).
10
Understanding cancer stem cell heterogeneity and dynamics: Implications for therapy in
human colorectal cancer
Terry Fox New Investigator Operating Grant (2013-2016)
Investigator: Catherine A. O’Brien, Princess Margaret Cancer Centre
Collaborator: Jason Moffat, University of Toronto
Mentoring Program: Genetic dissection of actionable targets required for maintenance of metastatic disease
Scientific Summary: We are developing and applying methods to better understand functional tumour dynamics at the
level of the single cell. In the cancer stem cell (CSC), field experiments typically involve isolating CSC and non-CSC
fractions and injecting them separately into immunocompromised mice at limiting dilution. Our hypothesis is that
current experimental models in solid tumour CSC research, although widely accepted, do not provide a complete
picture because they do not assess how CSC subsets interact with each other and with non-CSCs. The ability to track
and isolate viable individual clones provides a powerful tool to study the clonal structure of tumours and how the
clones interact with each other in the context of tumour formation. This model also provides a means to better
understand the clonal composition of metastatic lesions, as compared to the primary tumour site. Finally, we are using
lentiviral barcoding of individual cells to study response to therapeutic intervention, including standard-of-care
chemotherapies and “stem cell” directed therapies at the clonal level. This work will provide unprecedented insight
into how tumours recur following treatment, at the level of the individual cell.
Aims: Our work uses shRNA lentiviral barcode libraries to uniquely label individual cancer stem cells and follow them in
the context of the bulk tumour, using human colorectal cancer (CRC) xenograft models.
In our first aim we are working towards demonstrating how clonal co-operation and competition influence overall
tumour structure and growth patterns. In our second aim we are using the lentiviral barcoding system to study the
effect of therapeutic intervention on the clonal structure of tumours at the level of the single cell. Another advantage
of our model is that it will allow us to viably isolate, characterize, and test the therapy resistant clones we identify. In
our final aim, we are studying clonal composition in the context of an orthotopic xenograft metastasis model of
colorectal cancer. This work will allow us to identify patterns of clonal spread and help us to determine if there is a
subset of CRC stem cells that are predisposed to metastasize. If we can identify clones that preferentially metastasize,
we can viably isolate and characterize these specific CSCs.
Updates: We have successfully labeled and tracked individual cancer cells in vivo using three colorectal cancer
xenograft models. We started by carrying out the gold standard test to enumerate CSCs, the in vivo serial passage
limiting dilution assay. At limiting dilution we expected tumours to arise from a single CSC; however, we found that this
is not always the case. Instead we are detecting clonal structures that suggest the existence of clonal co-operation and
competition. We have isolated and are currently combining individual clones in vivo and in vitro to functionally define
how individual clones interact with and influence each other’s growth.
The focus of the first aim is to better understand cancer cell co-operation and competition, at the level of the individual
cells. The ability to understand how individual clones functionally interact is key to developing novel strategies aimed
at targeting the tumour as a whole.
In our second aim we are treating barcoded CRC xenografts with standard of care chemotherapies and stem cell
directed therapies. As a first step we are testing the efficacy of treatment strategies that decrease the relative
abundance of all clones versus those strategies that decrease the overall clonal complexity. There is an assumption that
less complexity would be favourable; however, this remains to be experimentally proven. The results obtained from
our work will have important implications for the field.
11
In our final aim we have lentivirally barcoded CRC cells that have been orthotopically injected into
immunocompromised mice and are currently awaiting the formation of liver metastases, the plan being to viable
isolate tumour cells from both the primary tumour and liver metastases and sequence the barcodes to determine the
clonal structures of each tumour site. Our aim is to determine whether metastatic spread is random or if we can use
the barcode labeling to predict the subset of clones that is predisposed to metastasize.
List of Key Publications:
1. Roulois D, Loo Yau H, Singhania R, Wang Y, Danesh A, Shen SY, Han H, Liang G, Pugh TJ, Jones PA, O’Brien CA, De Carvalho DD.
DNA-demethylating agents target colorectal cancer-initiating cells by activation of MDA5/MAVS/IRF7 pathway. Cell: 2015 Aug
27;162(5):961-73. PubMed PMID:26317465.
2. Chen EC, Karl TA, Kalisky T, Gupta SK, O'Brien CA, Longacre TA, van de Rijn M,Quake SR, Clarke MF, Rothenberg ME. KIT
Signaling Promotes Growth of Colon Xenograft Tumors in Mice and is Upregulated in a Subset of Human Colon Cancers.
Gastroenterology. 2015 May 27. PubMed PMID: 26026391.
3. Belmont PJ, Jiang P, McKee TD, Xie T, Isaacson J, Baryla NE, Roper J, Sinnamon MJ, Lee NV, Kan JL, Guicherit O, Wouter BG,
O’Brien CA, Shields D, Olson P, Vanarsdale T, Weinrich SL, Rejto P, Christensen JG, Fantin VR, Hung KE, Martin ES. Resistance to
dual blockage of the kinases PI3K and mTOR in KRAS mutant colorectal cancer models results in combined sensitivity to inhibition of
the receptor tyrosine kinase EGFR. Sci Signaling 2014 Nov 11; 7(351) PubMed PMID: 25389372.
4. Kreso A, van Galen P, Pedley NM, Lima-Fernandes E, Frelin C, Davis T, Cao L,Baiazitov R, Du W, Sydorenko N, Moon YC, Gibson
L, Wang Y, Leung C, Iscove NN, Arrowsmith CH, Szentgyorgyi E, Gallinger S, Dick JE, O'Brien CA. Self-renewal as a therapeutic target
in human colorectal cancer. Nat Med. 2014 Jan;20(1):29-36. PubMed PMID: 24292392.
5. Kreso A, O'Brien CA, van Galen P , Gan O , Notta F , Brown AM , Ng K , Ma J , Wienholds E , Dunant C , Pollett A , Gallinger S ,
McPherson J , Mullighan CG , Shibata D, Dick JE. Variable Clonal Repopulation Dynamics Influence Chemotherapy Response in
Colorectal Cancer. Science (New York, N.Y.), 2013 Feb 1;339(6119):543-8. PubMed PMID: 23239624
6. Vizeacoumar FJ, Arnold R, Vizeacoumar FS, Chandrashekhar M, Buzina A, Young JT, Kwan JH, Sayad A, Mero P, Lawo S, Tanaka
H, Brown KR, Baryshnikova A, Mak AB, Fedyshyn Y, Wang Y, Brito GC, Kasimer D, Makhnevych T, Ketela T, Datti A, Babu M, Emili
A, Pelletier L, Wrana J, Wainberg Z, Kim PM, Rottapel R, O'Brien CA, Andrews B, Boone C, Moffat J. A negative genetic interaction
map in isogenic cancer cell lines reveals cancer cell vulnerabilities. Mol Syst Biol. 2013 Oct 8;9:696. PubMed PMID: 24104479;
7. O’Brien CA, Kreso A, Ryan P, Hermans K, Gibson L, Wang Y, Tsatsanis A, Gallinger S, Dick JE. Id1 and Id3 regulate the selfrenewal capacity of human colon cancer-initiating cells through p21. Cancer Cell 2012: 21(6):777-792. PubMed 22698409
8. O'Brien CA, Pollett A, Gallinger S, Dick JE. A human colon cancer cell capable of initiating tumour growth in immunodeficient mice.
Nature 2007.Jan 4;445 (7123):106-110: 445: pp 7123-7129. PubMed 17122772
12
Understanding the impact of cancer cell fate decisions during ovarian cancer treatment
Terry Fox New Investigator Operating Grant (2014-2016)
Investigator: Francis Rodier, Université de Montréal Collaborators: David Huntsman, BC Cancer Agency;
Anne-Marie Mes-Masson, Université de Montréal
Mentoring Program: A pan-Canadian program for the development of biomarker-driven, sub-type specific
management of ovarian cancer
Scientific Summary: This project aims to develop new tools that will help select the best treatment options for ovarian
cancer (OvCa) patients. OvCa is one of the most lethal malignancies affecting women with poor five-year survival rates,
mostly because it is often detected late, at an aggressive stage. The success of any cancer treatment is usually
calculated based on long-term parameters such as patient survival and increased quality of life. Surprisingly, while
general health effects in response to cancer treatments are well-documented, very little is known about the cellular
biology behind tumour regression. For example, the immediate responses of cancer cells to damages initiated by
radiotherapy or chemotherapy remain unknown. In reality, damaged cells have many options: they can repair the
lesions (very often cancer therapy causes DNA lesions), they can die, or they can enter a state of permanent growth
arrest termed senescence. Every cancer is unique, and theoretically, cancer cells may select any of these decisions.
Whether these cellular decisions taken during treatment will influence the outcome of cancer treatment is currently
unknown.
Aims: Our hypothesis is that decisions taken by single OvCa cells in response to treatment will impact long-term
patient survival. In this research program, we propose to extensively characterize OvCa cell responses to therapy and
to define whether each different response has a clinical impact on patient survival. This knowledge could help us to
detect early whether a cancer is susceptible or is responding appropriately to a treatment and would improve our
ability to select the appropriate treatment. We also think that this knowledge could benefit treatment outcomes by
generating novel innovative pharmacological targets.
Updates: We have now determined that two particular subsets of OvCa, the diseases called high-grade serous and
clear cell carcinoma, both prefer to use a response to treatment termed “cellular senescence”, which is a phenomena
akin to accelerated cellular aging. Moreover, we observed that the occurrence of senescence in OvCa tumours is
apparently beneficial for the patient. We are now designing senescence-specific biomarkers to predict whether a
particular OvCa tumour can undergo senescence and to follow in real-time the occurrence of senescence during
treatment. As a long-term goal, we propose that pharmaceutically enhancing OvCa senescence during treatment could
improve overall treatment success.
List of Key Publications:
1.
2.
3.
4.
5.
6.
Gonzalez LC, Ghadaouia S, Martinez A, Rodier F. Premature aging/senescence in cancer cells facing therapy: good or bad?
Biogerontology. 2015 Sep 2. [Epub ahead of print] PubMed PMID: 26330289.
Cheng S, Rodier F. Manipulating senescence in health and disease: emerging tools. Cell Cycle. 2015;14(11):1613-4. doi:
10.1080/15384101.2015.1039359. PubMed PMID: 25928330.
Malaquin N, Carrier-Leclerc A, Dessureault M, Rodier F. DDR-mediated crosstalk between DNA-damaged cells and their
microenvironment. Front Genet. 2015 Mar 12;6:94. doi: 10.3389/fgene.2015.00094. eCollection 2015. Review. PubMed
PMID: 25815006.
Demaria M, Ohtani N, Youssef SA, Rodier F, Toussaint W, Mitchell JR, Laberge RM, Vijg J, Van Steeg H, Dollé ME,
Hoeijmakers JH, de Bruin A, Hara E, Campisi J. An essential role for senescent cells in optimal wound healing through
secretion of PDGF-AA. Dev Cell. 2014 Dec 22;31(6):722-33. doi: 10.1016/j.devcel.2014.11.012. Epub 2014 Dec 11. PubMed
PMID: 25499914.
Laberge RM, Adler D, DeMaria M, Mechtouf N, Teachenor R, Cardin GB, Desprez PY, Campisi J, Rodier F. Mitochondrial
DNA damage induces apoptosis in senescent cells. Cell Death Dis. 2013 Jul 18;4:e727. doi: 10.1038/cddis.2013.199. PubMed
PMID: 23868060.
Rodier F. Detection of the senescence-associated secretory phenotype (SASP). Methods Mol Biol. 2013;965:165-73. doi:
10.1007/978-1-62703-239-1_10. PubMed PMID: 23296657.
13
7.
8.
9.
Rodier F, Campisi J. Four faces of cellular senescence. J Cell Biol. 2011 Feb 21;192(4):547-56. doi: 10.1083/jcb.201009094.
Epub 2011 Feb 14. Review. PubMed PMID: 21321098.
Rodier F, Muñoz DP, Teachenor R, Chu V, Le O, Bhaumik D, Coppé JP, Campeau E, Beauséjour CM, Kim SH, Davalos AR,
Campisi J. DNA-SCARS: distinct nuclear structures that sustain damage-induced senescence growth arrest and inflammatory
cytokine secretion. J Cell Sci. 2011 Jan 1;124(Pt 1):68-81. doi: 10.1242/jcs.071340. Epub 2010 Nov 30. PubMed PMID:
21118958.
Rodier F, Coppé JP, Patil CK, Hoeijmakers WA, Muñoz DP, Raza SR, Freund A,Campeau E, Davalos AR, Campisi J.
Persistent DNA damage signalling triggers senescence-associated inflammatory cytokine secretion. Nat Cell Biol. 2009
Aug;11(8):973-9. doi: 10.1038/ncb1909. Epub 2009 Jul 13. PubMed PMID: 19597488.
14
Are genomic instability and clonal diversity prognostic indicators of high-grade serous
ovarian cancer?
Terry Fox New Investigator Operating Grant (2012-2015)
Investigator: Sohrab Shah, BC Cancer Agency
Mentoring Program: A pan-Canadian program for the development of biomarker-driven, sub-type specific
management of ovarian cancer
Scientific Summary: In North America, ovarian cancer is the leading cause of death due to gynecological malignancies.
The majority of women diagnosed with this disease are not expected to survive beyond five years. This project focuses
on the most common subtype, high-grade serous ovarian cancers which account for 70% of ovarian cancers. The
current standard of treatment involves platinum-based chemotherapies, however, though they are effective in treating
the primary tumour, in almost all instances, the cancer will reform and are resistant to any of the currently available
therapies. Current clinical markers are ineffective in determining which patients will respond more effectively to
chemotherapy and live longer without evidence of disease. High-grade serous carcinomas are genomically diverse and
heterogeneous, i.e., their genetic make-up vary from tumour to tumour and within the same tumour, populations of
cells can be very different from one another.
Our research group recently observed that there are, in fact, global patterns of diversity that exist amongst this group
of tumours. This proposal will examine whether these global patterns of genomic diversity can be used to segregate
patient populations and predict which patients fare better than others. We have assembled a highly productive and
multidisciplinary team to undertake this research. State-of-the-art genome sequencing technology and novel
bioinformatic and algorithmic approaches developed in our lab will be used to decipher the entire DNA sequence of
the tumours which will be compared to the patient’s normal DNA to identify global patterns of change. In addition,
using state-of-the art single-cell microfluidic devices developed by our group, we will be able to sequence the DNA in
individual cells to determine whether there are subpopulations of cells within the primary tumour that are resistant to
platinum-based therapy and emerge as dominant clones in relapsed patients. All the tools, infrastructure and tissue
specimens required for the discovery phase of this project are available within the BC Cancer Agency and UBC.
We will validate our findings in a much larger patient population (>500 samples) using a national ovarian cancer
resource (COEUR) funded by the Terry Fox Research Institute. The ultimate goal is to develop biomarkers or
parameters that can be used clinically to predict a patient’s response to chemotherapy. The discoveries from this
research can be immediately translatable to other cancer types that follow similar patterns of evolution and
progression.
15
The role of the CD73-adenosinergic pathway in prostate cancer
Terry Fox New Investigator Operating Grant (2015-2017)
Investigator: John Stagg, PhD, Centre de Recherche du Centre Hospitalier de l’Université de Montréal
Mentoring Program: The Canadian Prostate Cancer Biomarker Network
Funding Partner: Fonds de Recherhce du Québec-Sante
Scientific Summary: Immunotherapy is a promising approach for treatment of prostate cancer (PC). Nevertheless,
clinical benefits from immunotherapy have traditionally been modest in PC, due at least in part to the ability of PC to
successfully evade immune control. The general objective of this proposal is to validate CD73 as a therapeutic target in
PC. Previous studies from our group have highlighted the importance of the CD73-adenosine axis for tumour immune
escape (1-5). Adenosine produced by CD73 activates four adenosine receptors (A1, A2A, A2B and A3), each possessing
variable tissue distribution and affinity to adenosine. A2A adenosine receptor plays a dominant role in the suppression
of T-cell responses against cancer. Through this proposal, we hope a better understanding of the role and place of the
adenosinergic pathway in PC will lead to the development of new therapeutic agents able to synergize with existing
immunotherapies.
Aims: We will investigate the prognostic value of CD73 in human PC and its association with immune infiltrates and
tissue hypoxia. Using mouse models of PC, we will investigate the role of CD73 in the prostate tumour
microenvironement. Finally, we will perform pre-clinical studies to test whether targeted blockade of CD73, or
downstream A2A adenosine receptors, can synergize with immune-checkpoint inhibitors and cell-based prostate
vaccines.
Updates: By crossing TRAMP transgenic mice, which spontaneously develop PC, with CD73 knockout mice, we
established the first proof-of-concept that CD73 promotes PC in mice. CD73 deficiency was associated with a significant
reduction of PC growth and an increased infiltration of CD8+ T-cells (3). Moreover, anti-CD73 mAb therapy effectively
suppressed the growth and metastasis of transplanted TRAMP-C1 tumours. Our preliminary data thus suggest that
CD73 is a potential target in PC.
List of Key Publications:
1.
Stagg J, et al. Anti-CD73 antibody therapy inhibits breast cancer tumor growth and metastasis and induces antitumor
immunity. 2010. Proc Natl Acad Sci USA. 107(4):1547-1552.
2.
Stagg J, et al. CD73-deficient mice have increased anti-tumor immunity and are resistant to experimental metastasis. Cancer
Res. 2011 Apr 15;71(8):2892-900.
3.
Stagg J, Beavis PA, Divisekera U, Liu MC, Möller A, Darcy PK, Smyth MJ. CD73-deficient mice are resistant to
carcinogenesis. Cancer Res. 2012 May 1;72(9):2190-6.
4.
Loi S, Pommey S, Haibe-Kains B, Beavis PA, Darcy PK, Smyth MJ, Stagg J. CD73 promotes anthracycline resistance and poor
prognosis in triple negative breast cancer. Proc Natl Acad Sci U S A. 2013 Jul 2;110(27):11091-6.
5.
Allard B1, Turcotte M, Stagg J. Targeting CD73 and downstream adenosine receptor signaling in triple-negative breast
cancer. Expert Opin Ther Targets. 2014 May 6.
16
Transcriptional and epigenetic consequences of MLL-AF9 translocations
Terry Fox New Investigator Operating Grant (2014-2017)
Investigator: Brian Wilhelm, University of Montreal
Mentoring Program: Improved assignment of best-available therapy for patients with MDS/AML
Funding Partners: Fonds de Recherche du Québec- Sante; The Cole Foundation; L’Institut de Recherche en
immunologie et cancérologie
Scientific Summary: Translocations of the mixed-lineage leukemia (MLL) gene are particularly frequent in acute
myeloid leukemia (AML) in infants and young adults, and are associated with a poor prognosis. While some MLL
translocations have been studied in a murine setting, the analysis of pediatric MLL-AML patient samples is complicated
by their scarcity and very high genetic heterogeneity. To overcome these limitations, we have used a novel model
system that allows multiple leukemias to be generated from individual human CD34+ cord blood (CB) samples. This is
the equivalent of having a single patient developing multiple independent MLL-AF9 (MA9) leukemias, removing the
problem of patient genetic heterogeneity. As a result, the data from our model AML system gives us a novel and
unique advantage when compared to RNA-seq data from MA9 AML patients. The step-wise analysis of AML
development has enabled us to clearly identify expression and splicing changes that are specific to MA9 leukemias but
which could not have been identified using the “noisy” patient data alone. By filtering our data to identify genes
expressed specifically in MA9 AML but which are not expressed in normal blood cells, we have discovered 34 MA9specific candidate genes that are biomarkers of this disease. We now wish to exploit this system to interrogate the
changes highlighted by our model leukemias, to elucidate the mechanisms of MA9 tumorigenesis in human cells.
Aims: The overall aim of this project is to study the mechanisms by which MLL-AF9 forces leukemia development and
to use this information to develop more effective treatments for patients. The first specific aim is to characterize which
of our candidate genes may be essential for leukemia development, through a small-scale shRNA screen. The second
aim is to perform a structure/function study of one of the candidates, a receptor tyrosine kinase, where we already
have evidence for its importance for this type of leukemia. Lastly, because our model leukemia data suggests
collaborating mutations are not required for MLL-AF9 AML, we will perform in vitro functional studies of the gene
NCOR2. We have identified this nuclear co-repressor protein as consistently losing a critical interaction domain as a
result of alternative splicing in our model and patient MLL-AF9 AMLs.
Updates: To date, we have generated and performed RNA-seq on 22 independent leukemia samples from 4 single CB
donors and we have also analyzed the patterns of DNA methylation at each stage of development. Initial shRNA knockdown experiments have highlighted several genes whose loss causes profound growth defects, and additional
experiments are now underway to try and better define the role of these proteins. In addition, several of our candidate
genes have been validated for use in a diagnostic test, which will improve patient diagnosis compared to standard
cytogenetics. Lastly, our work studying the function of NCOR2 has uncovered connections to another protein that
regulates the differentiation of cells, potentially implicating it in the development of AML. We will test this hypothesis
to validate whether or not this protein may also represent a novel therapeutic target in MA9-AML.
List of Key Publications:
1.
Lavallée VP, Baccelli I, Krosl J, Wilhelm B, Barabé F, Gendron P, Boucher G, Lemieux S, Marinier A, Meloche S, Hébert J,
Sauvageau G. The transcriptomic landscape and directed chemical interrogation of MLL-rearranged acute myeloid
leukemias. Nat Genet. 2015 Sep;47(9):1030-7.
2.
Celton M, Forest A, Gosse G, Lemieux S, Hebert J, Sauvageau G, Wilhelm BT. Epigenetic regulation of GATA2 and its impact
on normal karyotype acute myeloid leukemia. Leukemia. 2014 Aug;28(8):1617-26.
3.
Wilhelm BT, Briau M, Austin P, Faubert A, Boucher G, Chagnon P, Hope K, Girard S, Mayotte N, Landry JR, Hébert J,
Sauvageau G. RNA-seq analysis of 2 closely related leukemia clones that differ in their self-renewal capacity. Blood. 2011 Jan
13;117(2):e27-38.
17
4.
Gosse G, Celton M, Lamontagne V, Forest A, Wilhelm BT. Whole genome and transcriptome analysis of a novel AML cell line
with a normal karyotype. Leuk Res. 2015 Jul;39(7):709-18.
5.
Nocq J, Celton M, Gendron P, Lemieux S, Wilhelm BT. Harnessing virtual machines to simplify next-generation DNA
sequencing analysis. Bioinformatics. 2013 Sep 1;29(17):2075-83.
6.
Deneault E, Wilhelm BT, Bergeron A, Barabé F, Sauvageau G. Identification of non-cell-autonomous networks from
engineered feeder cells that enhance murine hematopoietic stem cell activity. Exp Hematol. 2013 May;41(5):470-478.e4.
7.
Shifman AR, Johnson RM,Wilhelm BT, Cascade: An RNA-seq visualization tool for cancer genomics, (submitted, BMC
Genomics)
8.
Wilhelm BT, Gil L, Celton M, Bergeron A, Lamontagne V, Roques E, Forest A, Johnson RM, Pécheux L, Simard J, Pelloux J,
Bellemare-Pelletier A, Gagnon E, Hebert J, Cellot S, Barabé F,Integrated analysis of MLL-AF9 AML patients and model
leukemias highlights novel therapeutic targets, (submitted, Science).
18
Exploring novel mechanisms of tumour vascularization in malignant brain tumours
Terry Fox New Investigator Operating Grant (2012-2015)
Investigator: Gelareh Zadeh, UHN
Mentoring Program: Genetic analysis of signaling pathways for vascular development and tumour angiogenesis
Scientific Summary: Angiogenesis involves a highly regulated and co-ordinated interaction of multiple angiogenic
factors and is critical for both embryonal development and physiological vessel formation in adults. Angiogenesis is also
proven to significantly contribute to the progression of various disease processes, including cancer.
Glioblastoma (GBM) is among the most angiogenic tumors, and it therefore makes sense from an investigative
perspective to understand the mechanisms of angiogenesis in these tumours as a means for identifying new
therapeutic opportunities. However, despite initial positive early response to anti-angiogenic therapy in GBMs, the
clinical benefits of anti-angiogenic treatment is limited and GBM recurrence within a few months remains inevitable.
This is, in part, due to unidentified mechanisms of neo-vascularization that can evade radiation and targeted therapy.
Therefore, recent research interest has focused on the possibility of new vessel formation through progenitor cells that
are replenished at a constant and continuous level and as a result they can avoid therapeutics.
Neovascularization has traditionally been considered to occur through two distinct processes, angiogenesis and
vasculogenesis. Angiogenesis is the process by which new vessels form from sprouting and branching of pre-existing
vessels, whereas vasculogenesis or de novo vessel formation, occurs by differentiation of endothelial precursor cells
(EPC). It has long been thought that vasculogenesis was restricted to embryonal vessel development while post-natal
vessel formation occurs primarily through angiogenesis. However, emerging evidence suggests that vasculogenesis can
occur in adult life and it has been argued to provide a potential mechanism for cancer neovascularization through
mobilization of EPCs from the bone marrow (BM) or circulation. This hypothesis remains highly controversial and there
is debate as to whether bone marrow derived progenitor cells (BMDCs) actually differentiate to endothelial cells (ECs)
or contribute to formation of vascular channels in neoplastic processes.
Additional open questions include whether BMDC vasculogenesis is influenced by tumor type, microenvironmental
factors, tumour growth stage and response to therapy. To do this, however, we needed a model that could examine
the process of vasculogenesis in vivo, and in real time. This is the impetus for us to establish the experimental
strategies outlined above in (Ai), using a two-photon laser microscopy (2PLM) system, coupled with an intra-cranial
window in mouse models of GBM to obtain in-vivo real-time longitudinal imaging of normal brain and GBM associated
vasculature. This strategy complements traditional immunohistochemical (IHC) and immunofluorescent (IF) analysis,
as it allows visualization at single cell resolution of fluorescent bone marrow derived progenitor cells, and fluorescent
GBM cells. It then goes beyond the traditional IHC and IF techniques by the examination of these processes in real
time, in a living mammalian organism. In a series of carefully controlled experiments, we observed that bone marrowderived cells (BMDCs) are recruited to the brain in response to cranial radiation (CR).
We demonstrated that BMDCs are recruited specifically to the site of CR, in a radiation dose and temporal-spatial
manner. We showed that BMDCs do not form endothelial cells but rather they differentiate predominantly into
inflammatory cells and microglia. Using a GBM in vivo model, we show three distinct patterns of BDMC activity in
response to tumour growth. While these cells do support GBM neo-vascularization, we definitively show that there is
no evidence of direct differentiation of BMDCs into endothelial cells and, moreover, no contribution to vasculogenesis
or to de novo vessel formation. This result is in contrast to prior studies by other groups, and we strongly believe that
the careful use of our 2PLM high-resolution in vivo optical imaging single-cell resolution system in real time was critical
in resolving this controversial topic in the field of GBM angiogenesis. Additionally, these findings support the concept
that a disruption of the region-dependent contribution of BMDCs to vasculogenesis may be an important therapeutic
opportunity.
19
The Terry Fox New Frontiers Program Project Grant: Canadian Oncolytic Virus Consortium
(COVCo)
Terry Fox New Frontiers Program Project (2012-2017)
Project Leader: John Bell, OHRI
Investigators: Harold Atkins, Jean-Simon Diallo, OHRI; David Stojdl, Children’s Hospital of Eastern Ontario; Brad Nelson,
Deeley Research Institute/BC Cancer Agency; Patrick Lee, Dalhousie University; Nahum Sonenberg, McGill University;
Brian Lichty, Dr. Jonathan Bramson, Dr. Yonghong Wan, Dr. Karen Mossman, McMaster University; Andrea McCart,
UHN
Scientific Summary: Our program is directed toward the discovery and testing of novel replicating anti-cancer viruses,
and complimentary biotherapeutic strategies for the treatment of cancer. We are focused on the most promising
oncolytic therapeutics including three that are currently in clinical development. We are also exploring cutting-edge
immunotherapies including genetically modified immune cell platforms and personalized anti-tumour vaccines, and
how best to combine OVs with immune-based therapies for cancer. We are carrying out fundamental studies to
understand how viral therapeutics interact with the host, the tumour and tumour microenvironment. Small-molecule
screening and functional genomics approaches are being used to identify therapeutic targets that can be modified to
enhance virus killing of tumour cells.
Our ongoing studies include:







Understanding how best to manipulate or engineer the host immune system to both facilitate virus delivery
and potentiate anti-tumour immune responses. 
Delineating signaling pathways that sensitize tumour vascular endothelium and cancer-associated fibroblasts
to virus infection and destruction, and developing methods to modulate these pathways in favour of enhanced
therapeutic activity. 
Characterization of the innate anti-viral response in normal and tumour cells that determine virus selectivity. 
Identification of tumour-specific pathways that can sensitize cancer cells to virus killing. 
Mechanism of action of small molecules that uniquely inactive the anti-viral interferon response. 
Role of protein translation regulation in determining virus therapeutic activity. 
Optimization of combination strategies using viruses and immunotherapies for synergistic anti-cancer activity
in preclinical models.
Highlights:
We have made substantial gains in understanding how to manipulate
the host immune system to maximize the therapeutic impact of oncolytic viruses, how to target, select and/or
optimize OV vectors for activity in resistant tumours, and revealed the influence of the normal cell component of
the tumour microenvironment, in the therapeutic action of OVs. Notably, adipocytes render tumours significantly
less sensitive to virotherapy, a key observation considering the prominence of fat as a risk factor for cancer. It is
also clear that combination approaches addressing barriers in the tumour microenvironment, or enhancing the
anti-tumour immune responses will have a dramatic impact on OV therapeutic efficacy. In 2015 our investigators
opened a first in man phase I/II clinical trial of an oncolytic rhabdovirus (Maraba MG1) in combination with antiMAGE A3 vaccination, which has arisen from the heterologous prime-boost approach in subproject 5. Also in this
past year, we have established pre-clinically that this oncolytic vaccination approach is readily adaptable to HPV
associated cancers, and relevant prostate cancer antigens. This first trial has paved the way for follow-on trials
targeting HPV associated cancers and prostate cancer over the next 2-4 years. Importantly, we plan to follow this
phase I/IIb trial with a subsequent trial evaluating the prime-boost approach in combination with checkpoint
inhibition, in partnership with BioCanRx, Turnstone Biologicals, OICR, and Merck.
20
List of Key Publications:
1.
2.
3.
4.
5.
6.
7.
8.
9.
10.
11.
12.
13.
14.
15.
16.
17.
18.
19.
20.
21.
22.
23.
Ottolino-Perry K, Tang N, Head R, Ng C, Arulanandam R, Angarita FA, Acuna SA, Chen Y, Bell J, Dacosta RS, McCart JA.
Tumour vascularization is critical for oncolytic vaccinia virus treatment of peritoneal carcinomatosis. Int J Cancer. 2013 Jul
24 [epub ahead of print].
Heo J, Reid T, Ruo L, Breitbach CJ, Rose S, Bloomston M, Cho M, Lim HY, Chung HC, Kim CW, Burke J, Lencioni R,
Hickman T, Moon A, Lee YS, Kim MK, Daneshmand M, Dubois K, Longpre L, Ngo M, Rooney C, Bell JC, Rhee BG, Patt R,
Hwang TH, Kirn DH. Randomized dose-finding clinical trial of oncolytic immunotherapeutic vaccinia JX-594 in liver cancer.
Nat Med. 2013 Mar; 19(3):329-36.
Breitbach CJ, Arulanandam R, De Silva N, Thorne SH, Patt R, Daneshmand M, Moon A, Ilkow C, Burke J, Hwang TH, Heo J,
Cho M, Chen H, Angarita FA, Addison C, McCart JA, Bell JC, Kirn DH. Oncolytic vaccinia virus disrupts Tumour-associated
vasculature in humans. Cancer Res. 2013 Feb 15; 73(4):1265-75.
Bridle BW, Chen L, Lemay CG, Diallo JS, Pol J, Nguyen A, Capretta A, He R, Bramson JL, Bell JC, Lichty BD, Wan Y. HDAC
inhibition suppresses primary immune responses, enhances secondary immune responses, and abrogates autoimmunity
during Tumour immunotherapy. Mol Ther. 2013 Apr; 21(4):887-94.
Russell JC, Peng KW, Bell JC. Oncolytic virotherapy. Nat Biotechnol. 2012 Jul 10; 30(7):658-70.
CG Lemay, Rintoul JL, Kus A, Paterson JM, Garcia V, Falls TJ, Ferreira L, Bridle BW, Conrad DP, Tang VA, Diallo JS,
Arulanandam R, Le Boeuf F, Garson K, Vanderhyden BC, Stojdl DF, Lichty BD, Atkins HL, Parato KA, Bell JC, Auer RC.
Harnessing oncolytic virus-mediated antiTumour immunity in an infected cell vaccine. Mol Ther. 2012 Sep; 20(9):1791-9.
Le Boeuf F, Batenchuk C, Vähä-Koskela M, Breton S, Roy D, Lemay C, Cox J, Abdelbary H, Falls T, Waghray G, Atkins H,
Stojdl D, Diallo JS, Kærn M, Bell JC. Model- based rational design of an oncolytic virus with improved therapeutic potential.
Nat Commun. 2013; 4:1974.
McGray AJ, Bernard D, Hallett R, Kelly R, Jha M, Gregory C, Bassett JD, Hassell JA, Pare G, Wan Y, Bramson JL. Combined
vaccination and immunostimulatory antibodies provides durable cure of murine melanoma and induces transcriptional
changes associated with positive outcome in human melanoma patients. Oncoimmunology. 2012 Jul 1; 1(4):419-31.
Pan D, Marcato P, Ahn DG, Gujar S, Pan LZ, Shmulevitz M, Lee PW. Activation of p53 by chemotherapeutic agents enhances
reovirus oncolysis. PLoS One. 2013; 8(1):e54006.
Gujar S, Dielschneider R, Clements D, Helson E, Shmulevitz M, Marcato P, Pan D, Pan LZ, Ahn DG, Alawadhi A, Lee PW.
Multifaceted therapeutic targeting of ovarian peritoneal carcinomatosis through virus-induced immunomodulation. Mol
Ther. 2013 Feb; 21(2):338-47.
West NR, Kost SE, Martin SD, Milne K, Deleeuw RJ, Nelson BH, Watson PH. Tumour-infiltrating FOXP3(+) lymphocytes are
associated with cytotoxic immune responses and good clinical outcome in oestrogen receptor-negative breast cancer. Br J
Cancer. 2013 Jan 15; 108(1):155-62. 7
Pol JG, Zhang L, Bridle BW, Stephenson KB, Rességuier J, Hanson S, Chen L, Kazdhan N, Bramson JL, Stojdl DF, Wan Y,
Lichty BD. Maraba virus as a potent oncolytic vaccine vector. Mol Ther. 2014 22(2): 420-9.
Lichty BD, Breitbach CJ, Stojdl DF & Bell JC. Going viral with cancer immunotherapy. Nature Reviews Cancer. 2014
Aug;14(8):559-67.
Arulanandam R, Batenchuk C, Varette O, Zakaria C, Forbes NE, Davis C, Krishnan R, Garcia V, Karmacharya R, Cox J, Sinha
S, Babawy A, Waite K, Weinstein E, Falls T, Chen A, Hamill J, Da Silva N, Conrad DP, Atkins H, Garson K, Ilkow C, Kaern M,
Vanderhyden B, Sonenberg N, Alain T, Le Boeuf F, *Bell JC, *Diallo JS. Microtubule Destabilizers Disrupt Interferon
Production and Sensitize to Rhabdovirus Bystander Killing. Nat Commun. 2015 Mar 30;6:6410.
Ilkow CS, Marguerie M, Batenchuk C, Mayer J, Ben Neriah D, Cousineau S, Falls T, Jennings V, Boileau M, Bellamy D, Bastin
D, Tanese de Souza C, Alkayyal A, Zhang J, Le Boeuf F, Arulanandam R, Stubbert L, Sampath P, Thorne S, Paramanthan P,
Chatterjee A, Strieter RM, Burdick M, Addison C, Stojdl DF, Atkins HL, Auer R, Diallo JS, Lichty B, and Bell JC. Reciprocal
cellular cross-talk within the tumor microenvironment promotes oncolytic virus activity. Nat Med. 2015 May;21(5):530-6.
Arulanandam R, Batenchuk C, Angarita FA, Ottolino-Perry K, Cousineau S, Mottashed A, Burgess E, Falls TJ, De Silva N,
Tsang J, Howe GA, Bourgeois-Daigneault MC, Conrad DP, Daneshmand M, Breitbach CJ, Kirn DH, Raptis L, Sad S, Atkins
H, Huh MS, Diallo JS, Lichty B, Ilkow CS, Le Boeuf F, Addison CL, McCart JA and Bell JC. VEGF-mediated induction of
PRD1-BF1/Blimp1 expression sensitizes tumour vasculature to oncolytic virus infection. Cancer Cell. 2015 Jul 22 pii: S15356108(15)00219-6.
Forbes N, Krishnan R, Diallo JS. Pharmacological modulation of anti-tumor immunity induced by oncolytic viruses. Front.
Oncol. 23 July 2014 | doi: 10.3389/fonc.2014.00191
Nehdi A, Sean P, Linares I, Colina R, Jaramillo M, Alain T. Deficiency in either 4E-BP1 or 4E-BP2 augments the innate antiviral immune response. PLoS One. 2014 Dec 22;9(12):e114854.
Bhat M, Robichaud N, Hulea L, Sonenberg N, Pelletier J, Topisirovic I. Targeting the translation machinery in cancer. Nat
Rev Drug Discov. 2015 Apr;14(4):261-78. Review. PMID:25743081
Pelletier J, Graff J, Ruggero D, Sonenberg N. Targeting the eIF4F translation initiation complex: a critical nexus for cancer
development. Cancer Res. 2015 Jan 15;75(2):250-63. Review. PMID:25593033
Faller WJ, Jackson TJ, Knight JR, Ridgway RA, Jamieson T, Karim SA, Jones C, Radulescu S, Huels DJ, Myant KB, Dudek
KM, Casey HA, Scopelliti A, Cordero JB, Vidal M, Pende M, Ryazanov AG, Sonenberg N, Meyuhas O, Hall MN, Bushell M,
Willis AE, Sansom OJ. mTORC1-mediated translational elongation limits intestinal tumour initiation and growth. Nature.
2015 Jan 22;517(7535):497-500. PMID:25383520
Piccirillo CA, Bjur E, Topisirovic I, Sonenberg N, Larsson O. Translational control of immune responses: from transcripts to
translatomes. Nat Immunol. 2014 Jun;15(6):503-11. Review. PMID:24840981
VanSeggelen H, Tantalo D, Afsahi A, Hammill J, and Bramson JL. Chimeric antigen receptor-engineered T cells as oncolytic
virus carriers. 2015 Mol Ther Oncol, in press.
21
The Terry Fox New Frontiers Program Project Grant in ultrasound and MRI for cancer
therapy (2014-2017)
Investigators: Gregory J. Czarnota and Gregory J. Stanisz, Sunnybrook Research Institute; Michael C. Kolios, Ryerson
University.
Scientific Summary: We are proposing to develop and enhance the use of ultrasound and MRI techniques to improve
cancer treatments. Our focus is on making chemotherapy and radiation therapy, two of the most common cancer
treatments, significantly better. We will continue our highly productive track record of developing quantitative
ultrasound and MRI methods to detect and track the progression of the effects of cancer therapies on cell death. In
addition we will continue to develop ultrasound (with a goal to MRI guidance) to significantly enhance the effects of
radiation.
The research here firstly focuses on using ultrasound to track responses to chemotherapy. We have demonstrated that
quantitative ultrasound can be used one week into a 4-6 month course of chemotherapy to determine as a new
function imaging method whether it is working or not. This will be combined with new MRI methods and photoacoustic
imaging as new methods tor tumour response tracking. For therapy we have also recently demonstrated that
ultrasound-stimulated microbubbles can be used to increase the efficacy of radiation treatments whereby a 2 Gy dose
of radiation combined with these treatments. We will scale up that research here to large animal models and clinically
compatible MRI systems with a view to having clinical impact through the introduction of these new methods in the
near term.
Specifically, there are four highly interrelated projects proposed which complement each other and are critical to
bringing these technologies ultimately to the clinic. The first will see the continued development of quantitative
ultrasound methods and new photoacoustic methods for the detection of tumour responses to cancer therapies at
high and low frequency, for preclinical and clinical applications, respectively. We will specifically expand this
component to include new work on photoacoustics to obtain functional measurements based on blood content and
oxygenation. The second will focus on correlative analyses which will be integrated with the quantitative ultrasound
approaches and will focus on evaluating new MRI methods and correlating these with whole-mount, three-dimensional
histopathological data and ultrasound data. We will be integrating these into our analyses as they are rapidly becoming
clinical standards. The third project will be centered about evaluating quantitative ultrasound data from patients
receiving cancer therapy and will draw on background established methods in quantitative ultrasound and be guided
by ongoing developments from the first two projects. The last project will further develop recent innovations in using
ultrasound as an enhancing agent for cancer therapy based on our discovery of bubble-enhanced ultrasound
potentiation of tumour response and as targeting method to deliver radio-sensitizers. The effects of these will be
tracked using our new imaging methods as they are prepared for clinical implementation.
List of Key Publications:
1.
El Kaffas A, Sadeghi-Naini A, Falou O, Giles A, Czarnota GJ. (2015). Assessment of tumour responseto radiation and vascular
targeting therapy using quantitative ultrasound spectroscopy. Med Phys. 42(8): 4965.
2.
Sadeghi-Naini A, Zhou S, Gangeh M, Jahedmotlagh Z, Falou O, Ranieri S, Azrif M, GIles A, CzarnotaGJ. (2015). Quantitative
evaluation of cell death in vitro and in vivo using conventional-frequency ultrasound. Oncoscience. In Press
3.
Merino T, Tran WT, Czarnota GJ. (2015). Re-irradiation for locally recurrent refractory breast cancer. Oncotarget. In Press
4.
Czarnota GJ. (2015). Ultrasound innovations in therapy response monitoring. Med Phys. 42(6): 3683.
5.
Czarnota GJ. (2015). Ultrasound-stimulated microbubble enhancement of radiation response. Biological Chemistry. 396(6-7):
645-57.
6.
Sadeghi-Naini A, Sofroni E, Papanicolau N, Falou O, Sugar L, Morton G, Yaffe M, Nam R, Sadeghian A, Kolios MC, Chung HT,
22
Czarnota GJ. (2015). Quantitative Ultrasound Spectroscopic Imaging for Characterization of Disease Extent in Prostate Cancer
Patients . Translational Oncology. 8(1):25-34.
7.
Sannachi K, Taddayon H, Sadgehi-Naini A, Tran W, Gandhi S, Wright F, Oelze M, Czarnota GJ. (2015). Non-invasive evaluation
of breast cancer response to chemotherapy using quantitative ultrasonic backscatter parameters. Medical Image Analysis. 20(1):
224-36.
8.
Tadayyon H, Sadegh-Naini A, Czarnota GJ. (2014). Non-invasive characterization of locally advanced breast cancer using textural
analysis of ultrasound spectral parametric images. Trans Oncol. 7(6):759-67.
9.
Briggs K, Al Mahrouki A, Nofiele J, El-Falou A, Stanisz M, Kim HC, Kolios MC, Czarnota GJ. (2014).Non-invasive Monitoring of
Ultrasound-Stimulated Microbubble Radiation Enhancement Using Photoacoustic Imaging. Technology in Cancer Research &
Treatment. 13(5): 435-44.
10. Kim HC, Al-Mahrouki A, Gorjizadeh A, Sadeghi-Naini A, Karshafian R, Czarnota GJ. (2014).Quantitative ultrasound
characterization of tumor cell death: ultrasound-stimulated microbubbles for radiation enhancement. PLoS One. 9(7): e102343.
11. Gangeh M, Sadeghi-Naini A, Diu M, Tadayyon H, Kamel M, Czarnota GJ. (2014). Categorizing extent of tumour cell death
response to cancer therapy using quantitative ultrasound spectroscopy and maximum mean discrepancy. IEEE Transactions on
Medical Imaging. 33(6): 1390-400.
12. Sadeghi-Naini A, Sannachi L, Pritchard K, Trudeau M, Ghandi S, Wright F, Zubovits J, Yaffe M, Kolios M, Czarnota GJ. (2014).
Early prediction of therapy responses and outcomes in breast cancer patients using quantitative ultrasound spectral texture.
Oncotarget. 5(11): 3497-511.
13. Ahmed El Kaffas, Joris Nofiele, Anoja Giles, Song Cho, Stanley K. Liu, Gregory J. Czarnota. (2014).DLL4-Notch Signalling
Blockade Synergizes Combined Ultrasound-Stimulated Microbubble and Radiation Therapy in Human Colon Cancer Xenografts.
PLoS One. 9(4): e93888.
14. Al-Mahrouki AA, Iradji S, Tran WT, Czarnota GJ. (2014). Cellular characterization of ultrasound stimulated microbubble
radiation enhancement. Disease Models & Mechanisms. 7(3): 363-372.
15. El Kaffas A, Al-Mahrouki A, Tran WT, Giles A, Czarnota GJ. (2014). Sunitinib effects on the radiation response of endothelial and
breast tumor cells. Microvascular Research. 92: 1-9
16. Nofiele JT, Czarnota GJ, Cheng HL.(2014). Noninvasive manganese-enhanced magnetic resonance imaging for early detection of
breast cancer metastatic potential. Mol Imaging.
17. Sannachi L, Tadayyon H, Sadeghi-Naini A, Kolios MC, Czarnota GJ. (2014). Personalization of breast cancer chemotherapy using
noninvasive imaging methods to detect tumor cell death responses. Breast Cancer Management. 3(1): 31-35.
18. Tadayyon H, Sadeghi-Naini A, Wirtzfeld L, Wright FC, Czarnota G. (2014). Quantitative ultrasound characterization of locally
advanced breast cancer by estimation of its scatterer properties. Medical Physics. 41(1): 012903.
19. Kwok SJ, El Kaffas A, Lai P, Al Mahrouki A, Lee J, Iradji S, Tran WT, Giles A, Czarnota GJ. (2013). Ultrasound-Mediated
Microbubble Enhancement of Radiation Therapy Studied Using Three-Dimensional High-Frequency Power Doppler Ultrasound.
Ultrasound in Medicine & Biology. 39(11): 1983-90.
20. Tran WT, El Kaffas A, Al-Mahrouki A, Gillies C, Czarnota GJ. (2013). A review of vascular disrupting agents as a concomitant
anti-tumour modality with radiation. Journal of Radiotherapy in Practice. 12(3): 255-262.
21. Kim HC, Al-Mahrouki A, Gorjizadeh A, Karshafian R, Czarnota GJ. (2013). Effects of biophysical parameters in enhancing
radiation responses of prostate tumors with ultrasound-stimulated microbubbles. Ultrasound in Medicine & Biology. 39(8): 137687.
22. Sadeghi-Naini A, Papanicolau N, Falou O, Tadayyon H, Lee J, Zubovits J, Sadeghian A, Karshafian R, Al-Mahrouki A, Giles A,
Kolios MC, Czarnota GJ. (2013). Low-frequency quantitative ultrasound imaging of cell death in vivo. Medical Physics. 40(8):
082901.
23. Sadeghi-Naini A, Falou O, Tadayyon H, Al-Mahrouki A, Tran W, Papanicolau N, Kolios MC, Czarnota GJ. (2013). Conventional
frequency ultrasonic biomarkers of cancer treatment response in vivo. Translational Oncology. 6(3): 234-43.
24. Kolios MC, Berndl ES, Wirtzeld LC, Strohm EM, Czarnota GJ. (2013). Acoustic and photoacoustic imaging of spheroids. The
Journal of the Acoustical Society of America. 133(5)
25. Sadeghi-Naini A, Falou O, Czarnota GJ. (2013). Characterizing tumor heterogeneous response to chemotherapy using lowfrequency ultrasonic spectroscopy. The Journal of the Acoustical Society of America. 133(5)
23
The Terry Fox New Frontiers Program Project Grant in the development of stemness-based
prognostic biomarkers and therapeutic targets (2015-2020)
Investigators: John Dick, Norman Iscove, and Rodger Tiedemann, Princess Margaret Cancer Centre, University Health
Network; Gary Bader, University of Toronto; and Peter Dirks, The Hospital for Sick Children.
Scientific Summary: Tumour heterogeneity plays a major role in therapy failure resulting in disease progression and
recurrence. Our team is guided by the central hypothesis that both sub-clonal genetic diversity and the existence of
cellular hierarchies contributes to tumour heterogeneity and it is the unified effect of both that influences the
stemness properties of individual tumour cells; ultimately, stemness is the key biological property that governs patient
outcomes.
We are focused on defining the determinants of stemness for high-risk cancers including acute myeloid leukemia
(AML), myeloma and glioblastomas multiforma (GBM) that have poor outcome and urgently require effective
therapies. Our vision is translating the unique knowledge we have gained into a new generation of clinically relevant
biomarkers and therapeutics that target the vulnerabilities of CSCs, reducing therapy failure and increasing patient
survival.
Each project is built upon state-of-the-art CSCs assays, using either primary human cancer cells or engineered murine
models, and novel approaches to screen for vulnerabilities of cancer cells in general, and CSCs specifically. We will also
define the genetic basis for subclonal diversity in myeloma and GBM. Our focus is to deploy specific functional assays
that measure self-renewal, the key hallmark of the stemness state, to gain mechanistic insight into how selected
genetic, epigenetic or metabolic targets govern CSCs function for each cancer type.
The vast amount of data will be integrated into a single bioinformatic core to gain insight into stemness at a pathway
level, which will be central to therapeutic target identification. The specific aims for each project coalesce around three
major translational outcomes: the development of stemness-based biomarkers for predicting clinical features that will
enable improved clinical cancer management; the identification of new therapeutic targets that ensure eradication of
bulk tumour cells as well as the CSCs that lie at the root of the cancer; and pre-clinical development of targets using
state-of-the-art primary cancer xenografts that our team pioneered.
24
The Terry Fox New Frontiers Program Project in killing the hydra: Genetic dissection of
actionable targets required for maintenance of metastatic disease (2014-2019)
Investigators: Sean Egan, Michael Taylor, The Hospital for Sick Children, UofT; James Woodgett, Samuel Lunenfeld
Research Institute, Mount Sinai Hospital, UofT; Eldad Zacksenhaus, Toronto General Research Institute, UofT
Scientific Summary: The majority of cancer deaths occur secondary to metastatic disease, as primary tumours are often
controllable with a combination of surgery and radiotherapy. Despite this, most studies are focused on primary tumours.
In 2012, the Taylor lab reported on results of a Sleeping Beauty transposon-based screen that revealed substantial
divergence between metastatic medulloblastoma and matching primary tumour. The Egan and Zacksenhaus labs have
performed similar screens in mouse models of breast cancer associated with mutations in p53, Pik3ca and Rb. Indeed,
the metastatic genes identified so far are very different from mutations associated with primary tumour formation in
these models. Finally, the Woodgett lab has defined novel tumour suppressor functions associated with a Wnt pathway
kinase. As a group, we have developed a series of innovative tools and assays to define the role of specific signaling
proteins and pathways in metastatic breast cancer and medulloblastoma. These include doxycycline-repressible
transgenics that can be used to probe the function of PI3K and ʙ-catenin in circulating tumour cells and disseminated
disease. In addition, we have established “Lazy Piggy” transposon-containing mice that can be used to screen for genes
that are required for continued survival of primary and disseminated tumour cells.
We hypothesize that the best targets for rationale therapy to treat metastatic disease is the subset of mutational
events that is required for tumour maintenance.
We will define critical maintenance genes for metastatic cancers of the brain and breast through the following projects
and cores:
 Targeting novel transcriptional pathways in metastatic BC
 Targeting metastatic maintenance pathways in medulloblastoma
 Contributions of Wnt and PI3K signaling in breast & brain tumour dissemination
 Defining and targeting metastatic events in RB1/p53 tumour suppressor pathway-driven breast and brain cancer
 Bioinformatics and genomics core
List of Key Publications:
1.
Wright KL, Adams JR, Liu JC, Loch AJ, Wong RG, Jo CEB, Beck LA, Santhanam DR, Weiss L, Mei X, Lane TF, Koralov SB,
Done SJ, Woodgett JR, Zacksenhaus E, Hu P and Egan, SE. Ras signaling is a key determinant for metastatic dissemination
and poor survival of luminal breast cancer patients. In press.
2.
Shih DJH, Northcott PA, Remke M, Korshunov A, Ramaswamy V, Kool M, Luu B, Yao Y, Wang X, Dubuc A, Garzia L,
Peacock J, Mack S, Wu X, Rolider A, Morrissey S, Cavalli F, Jones DTW, Zitterbard K, Faria CC, Schuller U, Kren L, Kumabe
T, Tominage T, Shin Ra Y, Garami M, Hauser P, Chan JA, Robinson S, Bognar L, Klekner A, Saad AG, Liau LM, Albrecht S,
Fontebasso A, Cinalli G, De Antonellis P, Zollo M, Cooper MK, Thompson RC, Bailey S, Lindsey JC, Di Rocco C, Massimi L,
Michiels EMC, Scherer SW, Phillips JJ, Gupta N, Fan X, Muraszko KM, Vibhaker R, Eberhart CG, Fouladi M, Lach B, Jung S,
Wechsler-Reya RJ, Fevre-Montagne M, Jouvet A, Jabado N, Pollack IF, Weiss WA, Lee JY, Cho BK, Kim SK, Wang KC,
Leonard JR, Rubin JB, de Torres C, Lavarino C, Mora J, Cho YJ, Tabori U, Olson JM, Gajjar A, Packer RJ, Rutkowski S,
Pomeroy SL, French PJ, Kloosterhof NK, Kros JM, Van Meir RG, Clifford SC, Bourdeaut F, Delattre O, Doz F, Hawkins CE,
Malkin D, Grajkowska WA, Perk-Polnik M, Bouffet E, Rutka JT, Pfister SM, Taylor MD. 2014 Cytogenetic Prognostication
Within Medulloblastoma Subgroups. J Clin Oncology. 20;32(9):886-96.
3.
Dembowy J, Adissu HA, Liu JC, Zacksenhaus E and Woodgett, JR. (2015) Effect of glycogen synthase kinase-3 inactivation
on mouse mammary gland development and oncogenesis. Oncogene 34, 3514-3526.
4.
Liu JC, Voisin V, Wang S, Lehal R, Wang D-Y, Datti A, Uehling D, Al-awar R, Egan SE, Bader GD, Tsao M, Mak TW,
Zacksenhaus E. Combined deletion of murine Pten and p53 induces mammary tumors with dependency on eEF2K. EMBO
Molecular Medicine. 2014 Oct 20;6(12):1542-60
25
The Terry Fox New Frontiers Program Project Grant in molecular correlates of treatment
failure in lymphoid cancers (2013-2016)
Investigator: Randy Gascoyne, BC Cancer Agency
Co-Investigators: Marco Marra, BC Cancer Agency, Genome Science Centre; Sohrab Shah, BC Cancer Agency; Christian
Steidl, Joseph Connors, BC Cancer Agency
Scientific Summary: Most lymphoid cancers are easily treated and, in specific subtypes, treatment is given with
curative intent. Diffuse large B-cell lymphoma (DLBCL) accounts for 30 to 40% of all non-Hodgkin lymphomas (NHL) and
is curable even when widely disseminated at the time of diagnosis. Seminal work from our group has recently
characterized the mutational landscape of these tumours and now the current renewal of this TFRI New Frontiers
Program Project Grant will investigate at unprecedented resolution, the molecular correlates of treatment failure
through the study of clinical samples from patients who were not cured with state-of-the-art therapy. Similar studies
will also be conducted in follicular lymphoma (FL), the second most common form of NHL.
The BC Cancer Agency is recognized as a world leader in using next-generation sequencing technologies of lymphoid
cancers to understand the fundamental biology and identify the recurrent genetic abnormalities (so-called driver
mutations) that represent the underpinnings of these cancers. Building on previous work from our group, we will
leverage prior discoveries to determine if tumours from patients with primary treatment failure are different from
those experiencing complete remissions through the study of relapsed or recurrent disease. We hypothesize that a
limited number of cellular pathway perturbations underlie the biology of treatment failure and by studying these cases
we will identify the candidate molecules and pathways that could be used to explore novel, targeted therapies. We
plan to study the functional consequences of these genetic alterations and develop a small suite of tests that could be
used to recognize at the time of diagnosis those patients destined to not be cured with current treatments. We
strongly believe that these studies will ultimately improve the outcome for patients with both DLBCL and FL and fulfill
our mandate of delivering precision medicine for patients with lymphoid cancers.
List of Key Publications:
1.
Pastore A, Jurinovic V, Kridel R, Hoster E, Staiger AM, Szczepanowski M, Pott Cm Kopp N, Murakami M, Horn H, Leich E,
Moccia AA, Mottok A, Sunkavalli A, van Hummelen P, Ducar M, Ennishi D, Shulha HP, Hother C, Connors JM, Sehn LH,
Dreyling M, Neuberg D, Moller P, Feller AC, Hansmann ML, Stein H, Rosenwald A, Ott G, Klapper W, Unterhalt M, Hiddemann
W, Gascoyne RD, Weinstock DM, Weigert O. Integration of gene mutations improves risk prognostication in follicular lymphoma.
Lancet Oncol (on-line August 7, 2015). PMID: 26256760
2.
Scott DW*, Mottok A*, Ennishi D, Wright GW, Farinha P, Ben-Neriah S, Kridel R, Barry GS, Hother C, Abrisqueta P, Boyle M,
Meissner B, Telenius A, Savage KJ, Sehn LH, Slack GW, Steidl C, Staudt LM, Connors JM, Rimsza LM, Gascoyne RD. Prognostic
significance of diffuse large B-cell lymphoma cell-of-origin determined by digital gene expression in formalin-fixed paraffinembedded tissue biopsies. *Authors contributed equally to the work. J Clin Oncol. 2015 Aug 3. pii: JCO.2014.60.2383. [Epub
ahead of print]. PMID:26240231
3.
Twa DDW, Mottok A, Chan FC, Ben-Neriah S, Woolcock BW, Tan KL, Mungall AJ, McDonald H, Zhao Y, Lim RS, Nelson BH,
Milne K, Shah SP, Morin RD, Marra MA, Scott DW, Gascoyne RD, Steidl C. Recurrent genomic rearrangements in primary
testicular lymphoma. Journal of Pathology, 2015 Jun;236(2):136-41. PMID: 25712539
4.
Chan FC, Telenius A, Healy S, Ben-Neriah S, Mottok A, Lim R, Drake M, Hu S, Ding J, Ha G, Scott DW, Kridel R, Bashashati A,
Rogic S, Johnson N, Morin RD, Rimsza LM, Sehn L, Connors JM, Marra MA, Gascoyne RD, Shah SP, Steidl C. An RCOR1 lossassociated gene expression signature identifies a prognostically significant DLBCL subgroup. Blood. 2015 Feb 5;125(6):959-966.
PMID: 25395426
5.
Lim EL, Trinh DL, Scott DW, Chu A, Krzywinski M, Zhao YJ, Robertson AG, Mungall AJ, Schein J, Boyle M, Mottok A, Ennishi D,
Johnson NA, Steidl C, Connors JM, Morin RD, Gascoyne RD, Marra MA. Comprehensive miRNA Sequence Analysis Reveals
Survival Differences in Diffuse Large B-cell Lymphoma Patients. Genome Biol. 2015 Jan 29;16(1):18. (This article made it to the
cover of the journal.) PMID: 25723320.
26
6.
Ha G, Roth A, Khattra J, Ho J, Yap D, Prentice LM, Melnyk N, McPherson A, Bashashati A, Laks E, Biele J, Ding J, Le A, Rosner
J, Shumansky K, Marra MA, Gilks CB, Huntsman DG, McAlpine JN, Aparicio S, Shah SP. TITAN: Inference of copy number
architectures in clonal cell populations from tumor whole genome sequence data. Genome Res. 2014 Nov;24(11):1881-1893.
PMID: 25060187
7.
Morin RD, Mungall K, Pleasance E, Mungall AJ, Goya R, Huff R, Scot DW, Ding J, Roth A, Chiu R, Corbett RD, Chan FC, MendezLago M, Trinh DL, Bolger-Munro M, Taylor G, Hadj Khodabakhshi A, Ben-Neriah S, Pon J, Meissner B, Woolcock B, Farnoud N,
Rogic S, Lim E, Johnson NA, Shah S, Jones S, Steidl C, Holt R, Birol I, Moore R, Connors JM, Gascoyne RD, Marra MA.
Mutational and structural analysis of diffuse large B cell lymphoma using whole genome sequencing. Blood (epub May 22, 2013)
(PMID: 23699601).
8.
Trinh DL, Scott DW, Morin RD, Mendez-Lago M, An J, Jones SJM, Mungall AJ, Zhao Y, Schein J, Steidl C, Connors JM,
Gascoyne RD, Marra MA. Analysis of FOXO1 mutations in diffuse large B cell lymphoma. Blood 121(18): 3666-74 (2013) (PMID:
23460611).
9.
Johnson NA, Slack GW, Savage KJ, Connors JM, Ben-Neriah S, Rogic S, Scott DW, Tan KL, Steidl C, Hosrman DE, Sehn LH,
Chan WC, Iqbal J, Meyer P, Lenz G, Wright G, Rimsza LM, Valentino C, Brunhoeber P, Grogan TM, Braziel RM, Cook JR, Tubbs
RR, Weisenburger DD, Campo E, Rosenwald A, Ott G, Delabie J, Jaffe ES, Staudt LM, Gascoyne RD. Concurrent expression of
MYC and BCL2 in R-CHOP treated diffuse large B cell lymphoma. J Clin Oncol 28: 3452-3459 (2012) (PMID: 22851565).
27
The Terry Fox New Frontiers Program Project Grant in oncometabolism and the molecular
pathways that fuel cancer (2015-2019)
Investigators: Vincent Giguère, Julie St-Pierre, Russell Jones, Arnim Pause, Nahum Sonenberg, William Muller, Peter
Siegel, Goodman Cancer Research Centre; Ivan Topisirovic, Michael Pollak, Lady Davis Research Institute.
Funding Partners: McGill University Rosalind and Morris Goodman Cancer Centre; The Quebec Breast Cancer
Foundation
Scientific Summary: The purpose of this program project grant is to foster a comprehensive research platform to study
the links between metabolic reprogramming and cancer progression. The overall goals are to identify and integrate
regulatory pathways and metabolic networks that impact the cancer phenotype with a focus on metastatic progression
and mechanisms of therapeutic resistance. Long-term goals are to discover and develop novel therapeutic strategies
to induce or target metabolic vulnerabilities leading to inability of tumours to overcome metabolic stresses and
reprogram their biosynthetic pathways to support aberrant growth, ultimately sensitizing them to anti-cancer drugs.
Poor-outcome breast cancer will constitute the central focus of the proposed studies as our group has developed
strong expertise in mouse models of human breast cancers, thus providing a unifying theme for the group. However,
other cancer types, such as prostate cancer, will be incorporated in certain studies to test specific hypotheses.
Recent Discoveries and Accomplishments:
 Established that the LKB1-AMPK energy-sensing pathway mediates tumour suppression and metabolic
reprogramming in cancer.
 Showed that loss of LKB1 enhances the development ErbB2-driven breast cancer through metabolic
reprogramming of breast cancer cells.
 Identified the tumour suppressor FLCN as an evolutionary-conserved negative regulator of AMPK.
 Showed that constitutive AMPK activation promotes PGC-1-mediated mitochondrial biogenesis, which stimulates
HIF transcriptional activity and metabolic reprogramming in cancer cells.
 Discovery that miR-378* is both a component and a regulator of the PGC-1β/ERRγ axis, and that ERBB2-activated
breast cancer cells can specifically exploit this pathway to reprogram their energy metabolism and thus increase
their growth potential.
 Demonstrated that PGC-1α, along with ERRα, is a positive regulator of the expression of glutamine metabolism
genes in ERBB2+ breast cancer cells.
 The biological relevance of the control of glutamine metabolism genes by the PGC-1α/ERRα axis was demonstrated
by consequent regulation of glutamine flux through the TCA.
 Showed that PGC-1α expression is positively correlated with that of the glutamine pathway in ERBB2+ breast
cancer patients, and high expression of this pathway is associated with reduced patient survival.
 Demonstrated that metformin exerts its cell-autonomous effects by directly inhibiting mitochondrial complex I.
 Established that the expression of ERR is restored in lapatinib-resistant breast cancer cells through constitutive
re-instatement of mTOR signalling, thus providing the optimal metabolic settings required for cell survival in the
presence of the lapatinib insult.
 Demonstrated that loss of ERRα promotes hepatocarcinogenesis development via metabolic and inflammatory
disturbances in the tumour cells and resident macrophages.
 Determined that mTOR inhibitors, as well as biguanides exert their biological effects by altering translation of a
specific subset of mRNAs encoding proteins known to promote neoplastic growth such as cyclins.
 Demonstrated that the anti-neoplastic effects of biguanides are attenuated by a compensatory activation of
glycolysis and that the anti-proliferative effects of biguanides are potentiated in cells that are deprived of serine
 Showed that the mTORC1/4E-BP pathway maintains cellular energy homeostasis by modulating translation of
mRNAs encoding mitochondria-related genes
 Determined the role of LKB1 in promoting ErbB2-mediated breast cancer progression and metastasis.
28


Validated claudin-2, a tight junctional protein, as a clinically relevant and functionally important mediator of breast
cancer liver metastasis.
The Metabolomic Core Facility contributed to the advancement of all research projects through the development
of novel technologies related to the study of metabolomics.
List of Key Publications:
1.
2.
3.
4.
5.
6.
7.
8.
9.
10.
11.
12.
13.
14.
15.
16.
17.
18.
19.
20.
21.
22.
23.
24.
25.
26.
Andrzejewski, S., Gravel, S.P., Pollak M.N. and St-Pierre J. (2014) Metformin directly acts on mitochondria to alter cellular
bioenergetics. Cancer & Metabolism 2:12.
Chan, M., Gravel, M., Bramoullé, A., Bridon, G., Avizonis, D., Shore, G.C. and Roulston, A. (2014) Synergy between the NAMPT
Inhibitor GMX1777(8) and Pemetrexed in non-small cell lung cancer cells Is mediated by PARP activation and enhanced NAD
consumption. Cancer Res. 74:5948-5954.
Chang, C.H., Curtis, J.D., Maggi, L.B. Jr, Faubert, B., Villarino, A.V., O'Sullivan, D., Huang, S.C., van der Windt, G.J., Blagih, J.,
Qiu, J., Weber, J.D., Pearce, E.J., Jones, R.G. and Pearce, E.L. (2013) Posttranscriptional control of T cell effector function by
aerobic glycolysis. Cell 153:1239-1251.
Chaveroux, C., Eichner, L.J., Dufour, C.R., Shatnawi, A., Khoutorski, A., Bourque, G., Sonenberg, N. and Giguère, V. (2013)
Molecular and genetic crosstalks between mTOR and ERR are key determinants of rapamycin-induced non-alcoholic fatty lever.
Cell Metabolism 17:586-598
Deblois, G., Charhour, G., Perry, M.-C., Sylvain-Drolet, G., Muller, W.J. and Giguère, V. (2010) Transcriptional control of the
ERBB2 amplicon by ERR and PGC-1 promotes mammary gland tumorigenesis. Cancer Research 70:10277-10287.
Deblois, G. and Giguère, V. (2012) Cancer: Reprogramming clinical outcome. Nature 481:275-266.
Deblois, G. and Giguère, V. (2013) Estrogen-related receptors (ERRs) in breast cancer: control of cellular metabolism and beyond.
Nature Reviews | Cancer 13:27-36.
Deblois, G., St-Pierre, J. and Giguère, V. (2013) The PGC-1/ERR axis in cancer. Oncogene 32:3483-3490.
Dupuy, F., Dong, Z., Avizonis, D., Ling, C., Griss, T., Siwak, D.R., Mills, G.B., Muller, W.J., Siegel, P.M., and Jones, R.G. (2013).
LKB1 loss promotes ErbB2-driven breast cancer development but impairs the growth of lung metastases. Cancer & Metabolism 1:
18.
Eichner, L.J., Dufour, C.R., Perry, M.C., Bertos, N., Park, M., St-Pierre, J. and Giguère, V. (2010) miR-378* mediates metabolic
shift in breast cancer cells via the PGC-1/ERR transcriptional pathway. Cell Metab. 12:352-361.
Everts, B., Amiel, E., Huang, S.C., Smith, A.M., Chang, C.H., Lam, W.Y., Redmann, V., Freitas, T.C., Blagih, J., van der Windt, G.J.,
Artyomov, M.N., Jones, R.G., Pearce, E.L. and Pearce, E.J. (2014) TLR-driven early glycolytic reprogramming via the kinases
TBK1-IKKɛ supports the anabolic demands of dendritic cell activation. Nat Immunol. 2014 15:323-332.
Faubert, B., *Boily, B., *Izreig, S., Griss, T., Samborska, B., Dong, Z., Dupuy, F., Chambers, C., Fuerth, B.J., Viollet, B., Mamer,
O.A., Avizonis, D., DeBerardinis, R.J., Siegel, P.M., and **Jones, R.G. (2013). AMPK is a negative regulator of the Warburg Effect
and suppresses tumor growth in vivo. Cell Metab. 17:113-124.
Faubert, B., Vincent, E.E., Griss, T., Svensson, R., Mamer, O.A., Avizonis, D., Shaw, R.J., and **Jones, R.G. (2014). Loss of LKB1
promotes metabolic reprogramming of cancer cells via HIF1. Proc. Nat. Acad. Sci.U.S.A. 111:2554-2559.
Faubert, B., Vincent, E.E., Poffenberger, M.C., and **Jones, R.G. (2014). The AMP-activated protein kinase (AMPK) and cancer:
Many faces of a metabolic regulator. Cancer Letters. pii: S0304-3835(14)00054-8.
Foretz, M., Guigas, B., Bertrand, L., Pollak, M.N. and Viollet, B. (2014) Metformin: from mechanisms of action to therapies. Cell
Metab. 20:953-966.
Giguère, V. (2014) Estrogen receptor mutations in breast cancer - an anticipated "rediscovery?". Mol. Endocrinol. 28:427-428.
Gravel, S.-P., Andrzejewski, S., Avizonis, D. and St-Pierre, J. (2014). Stable isotope tracer analyses in isolated mitochondria from
mammalian systems. Metabolites 4:166-183.
Gravel, S.P., Hulea, L., Toban, N., Birman, E., Blouin, M.-J., Zakikhani, M., Zhao, Y., Topisirovic, I., St-Pierre, J. and Pollak, M.N.
(2014). Serine deprivation enhances anti-neoplastic activity of biguanides. Cancer Research 74:7521-7533.
Javeshghani, S., Zakikhani, M., Austin, S., Bazile, M., Blouin, M. J., Topisirovic, I., St-Pierre, J., Pollak, M.N. (2012) Carbon source
and myc expression influence the antiproliferative actions of metformin Cancer Res. 72:6257-6267.
Klimcakova, E., Chénard, V., McGuirk, S., Germain, D., Avizonis, D., Muller, W.J. and St-Pierre, J. (2012). PGC-1α promotes the
growth of ErbB2/Neu-induced mammary tumors by regulating nutrient supply. Cancer Res. 72:1538-1546.
Larsson, O., Masahiro, M., Topisirovic, I., Alain, T., Blouin, M.J., Pollak, M.N. and Sonenberg, N. (2012) Distinct perturbation of
the translatome by the antidiabetic drug metformin. Proc Natl Acad Sci USA 109:8977-8982.
Leprivier, G., Remke, M., Rotblat, B., Dubuc, A., Mateo, A., Kool, M., Agnihotri, S., El-Naggar, A., Yu, B., Somasekharan, S.P.,
Faubert, B., Bridon, G., Tognon, C.E., Mathers, J., Thomas, R., Li, A., Barokas, A., Bowden, M., Smith, S., Wu, X., Korshunov, A.,
Hielscher, T., Northcott, P.A., Galpin, J.D., Ahern, C.A., Wang, Y., McCabe, M.G., Collins, P., Jones, R.G., Pollak, M.N., Delattre,
O., Gleave, M.E., Jan, E., Pfister, S.M., Proud, C.G., Derry, W.B., Taylor, M.D., and Sorensen, P.H.B. (2013). The eEF2 kinase
confers resistance to nutrient deprivation by blocking translation elongation. Cell 153:1064-1079.
McGuirk, S., Gravel, S.-P., Deblois, G., Papadopoli, D., Faubert, B., Wegner, A., Hiller, K., Avizonis, D., Akavia, U.-D., Jones, R.G.,
Giguère, V. and St-Pierre, J. (2013) PGC-1α supports glutamine-mediated lipogenesis in breast cancer. Cancer & Metabolism 1:22.
Morita, M., Gravel, S.-P., Chénard, V., Sikström, K., Zheng, L., Alain, T., Gandin, V., Avizonis, D., Arguello, M., Zakaria, C.,
McLaughlan, S., Nouet, Y., Pause, A., Pollak, M., Gottlieb, E., Larsson, O., St-Pierre, J., Topisirovic, I. and Sonenberg, N. (2013).
Translational control of the mitochondrion. Cell Metabolism 18:698-711.
Pearce, E.L., Poffenberger, M., Chang, C.-H., and Jones, R.G. (2013). Fueling immunity: New insights on metabolism and
lymphocyte function. Science 342(6155): 1242454.
Pollak, M.N. (2012) Investigating metformin for cancer prevention and treatment: the end of the beginning. Cancer Discovery
2:778-790.
29
The Terry Fox New Frontiers Program Project Grant in Prostate Cancer Progression
(2011-2016)
Investigators: Martin Gleave, Colin Collins, Emma Guns, Michael Cox, Chris Ong, Paul Rennie, Amina Zoubeidi, YZ
Wang, VPC,UBC; Kim Chi, Shoukat Dedhar, Poul Sorensen, BC Cancer Agency
Scientific Summary: Progression to castrate resistance following androgen ablation is the main obstacle to improving
survival for men with advanced prostate cancer and the central focus of our Terry Fox New Frontiers Program Project
Grant, comprised of a multidisciplinary team of 20 scientists and clinicians. Androgen ablation precipitates a cascade of
changes in transcriptional and signalling networks that provide a selective survival and growth advantage for subpopulations of the tumour cells, thereby accelerating progression and rendering cells more resistant to therapy.
Objectives:
 Elucidate genomic, molecular and cellular mechanisms responsible for progression to castrate resistance.
 Use this information to develop new therapies aimed at biologically relevant and tumour-specific targets and
pathways to delay progression of late stage disease.
 Partner with national clinical trials networks and industry to accelerate bench-to-bedside translation of our
discovery science.
 Since our ongoing TF program cycle renewal in December 2011, we have published over 530 papers, applied for or
obtained >130 patents, initiated 65 clinical trials and enrolled 814 patients. Since the beginning of support of our
Terry Fox New Frontiers Program on Prostate Cancer Progression we have outlicensed 6 novel therapeutics and
completed 8 Phase I/II trials of novel agents discovered as a direct consequence of laboratory research performed
under this program. Two more novel drug products that target SEMA3C and the DNA binding domain of the AR are
ready for clinical development; these novel agents were discovered as a direct consequence of pre-clinical
laboratory research performed under the auspices of the program.
Leading in this regard is OGX-011 (Custirsen), now in global Phase III trials. The AFFINITY trial (second-line metastatic
Castrate-Resistant Prostate Cancer (CRPC)) and ENSPIRIT trial (non-small-cell lung cancer (NSCLC)) are ongoing, while
the SYNERGY trial failed to achieve its primary end point as a first-line therapy in CRPC. We also led the bench-bedside
translation of a second novel inhibitor targeting Hsp27, OGX-427, which is currently in six randomized Phase II studies
in CRPC, bladder, NSCLC and pancreatic cancer. The Borealis-1 TM Phase II Trial of OGX-427 in metastatic bladder
cancer has been completed and data recently presented showing 50% reduction in risk of death in patients with low
performance status. This direct translation of basic science to the clinic is how our team works and is in keeping with
the mandate of the Terry Fox Foundation.
Our program consists of six individual but highly integrated research projects grouped into four relational areas:
 Target Discovery: Project #1 is using next-generation sequencing technologies to identify changes in the genomes
and transcriptomes of tumours mechanistically linked to castrate resistance, with a particular focus on androgen
receptors and androgen biosynthetic pathways.
 Cell Biology Mechanisms: Project #2 is continuing studies on the stress response and cytoprotective chaperones in
treatment resistance, focusing on the role of clusterin in endoplasmic reticular stress and autophagy. Project #3 is
investigating mechanisms of ERG-mediated prostatic carcinogenesis and progression.
 Molecular & Cellular Targets: Projects #4 and #5 are investigating the anti-apoptosis proteins, BIRC6, and
Semaphorin 3C, respectively, in treatment resistance of prostate cancer and as potential therapeutic targets.
 Clinical Evaluation: Project #6 is performing correlative measures to test biologic activity for a combination of a
potent anti-androgen in combination with an inhibitor of clusterin (OGX-011) or Hsp27 (OGX-427, both developed
in our Program) in a Phase II clinical trial in CRPC patients. In this regard we have initiated a multi-center
randomized trial of OGX-427 +/- abiraterone in post ABI CRPC. All sub-projects within the Terry Fox program are
supported by a shared core facility with five major components: Advanced Genomics & Bioinformatics, Pathology &
Molecular Imaging, Animal Models, Analytical Pharmacology, and Translational Trials.
30
In summary, our Program Project Grant on Prostate Cancer Progression is a major catalyst for translational research
that has enabled us to have already brought several new therapies from bench to bedside. The program will help
further accelerate discovery and validation of novel cellular and molecular targets and uncover mechanisms for
treatment resistance.
By pooling our talents and resources into a single co-operative effort, we maximize our potential for solving the
problem of prostate cancer progression in the most efficient manner. This program is an ideal example of how team
science enables the discovery of underlying mechanisms of prostate cancer progression, facilitates the development of
new multimodality therapies, and accelerates translation of research into clinical practice. The many publications
related to this program listed below are evidence of our team-based interactions and productivity.
List of Key Publications:
1.
Akamatsu S, Wyatt AW, Lin D, Lysakowski S, Zhang F, Kim S, Tse C, Wang K, Mo F, Haegert A, Brahmbhatt S, Bell B,
Adomat H, Xue H, Dong X, Fazli L, Tsai H, Lotan TL, Kossai M, Mosquera JM, Rubin MA, Beltran H, Zoubeidi A, Wang Y,
Gleave ME, Collins CC. The placental gene PEG10 promotes progression of neuroendocrine prostate cancer. 2015 Cell
Reports [epub July 30] 07/2015; DOI:10.1016/j.celrep. 2015.07.012 IF 7.21
2.
Azad AA, Volik S, Wyatt A, Haegert A, Bell R, McConeghy B, Shukin R, Paris P, Anderson S, Bazov J, Youngren J, Thomas G,
Small EJ, Gleave ME, Collins CC, Chi KM. (2015). Androgen receptor gene aberrations in circulating cell-free DNA:
biomarkers of therapeutic resistance and response in castration-resistant prostate cancer Clinical Cancer Research. 21:231524. doi: 10.1158/1078-0432. PMID: 25712683 IF 8.193.
3.
Beltran H, Eng K, Mosquera JM, Sigaras A, Romanel A, Rennert H, Kossai M, Pauli C, Faltas B, Fontugne J, Park K,
Banfelder J, Prandi D, Madhukar N, Zhang T, Padilla J, Greco N, McNary TJ, Herrscher E, Wilkes D, MacDonald TY, Xue H,
Vacic V, Emde AK, Oschwald D, Tan AY, Chen Z, Collins C, Gleave ME, Wang Y, Chakravarty D, Schiffman M, Kim R,
Campagne F, Robinson BD, Nanus DM, Tagawa ST, Xiang JZ, Smogorzewska A, Demichelis F, Rickman DS, Sboner A,
Elemento O, Rubin MA.Whole-Exome Sequencing of Metastatic Cancer and Biomarkers of TreatmentResponse. JAMA
Oncol. 2015 Jul 1;1(4):466-74. doi: 10.1001/jamaoncol.2015.1313. PMID:26181256
4.
Boutros, P.C., Fraser, M. et al. (2015). Spatial genomic heterogeneity within localized, multifocal prostate cancer. Nature
Genetics 47: 736–745. IF 29.64
5.
Choi SYC, Lin D, Gout PW, Collins CC, Xu Y, Wang YZ. (2014). Lessons from patient-derived xenografts for better in vitro
modeling of human cancer” Advanced Drug Delivery Reviews. 2014 Dec 15;79-80C:222-237. PMID 25305336 IF 12.71
6.
Clermont PL, Lin D, Crea F, Wu R, Xue H, Wang Y, Thu KL, Lam WL, Collins CC, Wang YZ, Helgason CD. Polycombmediated silencing in neuroendocrine prostate cancer. Clinical Epigenetics 2015, 7:40 doi:10.1186/s13148-015-0074-4. IF
6.219.
7.
Crea F, Saidy NRN, Collins C, Wang YZ. The epigenetic/non-coding origin of tumour dormancy. Trends Mol Med 21(4): 206211, 2015. IF 10.11
8.
Eigl BJ, North S, Winquist E, Finch D, Wood L, Sridhar SS, Powers J, Good J, Sharma M, Squire JA, Bazov J, Jamaspishvili
T, Cox ME, Bradbury PA, Eisenhauer EA, Chi KN. A Phase II Study of SB939 in Patients with Castration Resistant Prostate
Cancer. Investigational New Drugs, 2015 May 19. [Epub ahead of print] . PMID: 25983041
9.
Gleave M, Chi K. Toward predictive signatures of enzalutamide response and resistance. Eur Urol. 2015 Jan;67(1):61-3. doi:
10.1016/j.eururo.2014.08.012. Epub 2014 Aug 20. PMID: 25151015
10. Lalonde E, Ishkanian AS, Sykes J, Fraser M, Ross-Adams H, Erho N, Dunning MJ, Halim S, Lamb AD, Moon NC, Zafarana G,
Warren AY, Meng X, Thoms J, Grzadkowski MR, Berlin A, Have CL, Ramnarine VR, Yao CQ, Malloff CA, Lam LL, Xie H,
Harding NJ, Mak DY, Chu KC, Chong LC, Sendorek DH, P'ng C, Collins CC, Squire JA, Jurisica I, Cooper C, Eeles R, Pintilie
M, Dal Pra A, Davicioni E, Lam WL, Milosevic M, Neal DE, van der Kwast T, Boutros PC, Bristow RG. (2014). Tumour
genomic and microenvironmental heterogeneity for integrated prediction of 5-year biochemical recurrence of prostate
cancer: a retrospective cohort study. Lancet Oncology 15:1521-32. PMID: 25456371 IF 24.725
11. Lamoureux F, Baud'huin M, Ory B, Guiho R, Zoubeidi A, Gleave M, Heymann D, Rédini F. Clusterin inhibition using OGX011 synergistically enhances zoledronic acid activity in osteosarcoma. Oncotarget. 2014 Sep 15;5(17):7805-19. PMID:
25138053
12. Lin D, Wyatt AW, Xue H, Wang Y, Dong X, Haegert A, Wu R, Brahmbhatt S, Mo F, Jong L, Bell RH, Anderson S, HurtadoCull A, Fazli L, Sharma M, Beltran H, Rubin MA, Cox ME, Gout PW, Morris J, Goldenberg L, Volik SV, Gleave ME, Collins
CC, Wang Y. (2014). High fidelity patient-derived xenografts for accelerating prostate cancer discovery and drug development
Cancer Res. 74(4):1272-83. PMID: 24356420 IF 9.284
31
The Terry Fox New Frontiers Program Project Grant in core pathogenic pathways in human
leukemia (2012-2017)
Investigators: R. Keith Humphries, Connie J. Eaves, Aly Karsan and Andrew Weng, BC Cancer Agency; Martin Hirst, UBC
Scientific Summary: Acute leukemias remain one of the most devastating and costly cancers with less than 1 in 5 adult
patients surviving 10 years and some childhood patients failing current treatments. Research into how normal blood
cells are formed and perturbations in leukemia have provided major insights into why cures are so hard to achieve,
with many seminal contributions provided by this longstanding program project. These indicate that most human
leukemias are sustained by a rare subset of “leukemia stem cells” which are often resistant to currently used drugs.
Cures, thus, require treatments that effectively target these leukemia stem cells, ideally with little toxicity for normal
cells.
This now seems possible with the advent of modern tools that can identify every change in every gene in cells, and that
can also determine whether and why every gene is being expressed. In addition, vast libraries of naturally occurring
and synthetic chemicals that can target specific molecules in cells are now available. Our group now brings powerful
genetic engineering tools to enable human models of aggressive leukemia to be rapidly created in the lab so that
mechanisms of treatment resistance and new drugs and biomarkers can be efficiently analyzed and tested directly and
repeatedly in human cells that mimic, but do not rely on, patients’ cells. Our projects focus on examples of the worst
types of leukemia known, with the goals to develop human models of these and, in concert with two groups of world
experts in the molecular analysis of cells, to use these models to search for common therapeutic targets.
Our research findings are providing powerful new platforms to study the process of initiation and progression of
several aggressive leukemias and are leading to the identification of relevant genes and pathways, many linked to the
epigenome ((e.g. Ikaros in CML, IGF-R in MDS, HIF1alpha in T-ALL). Insights gained promise new therapeutic targets and
biomarkers.
List of Key Publications:
1.
2.
3.
4.
5.
6.
7.
8.
9.
10.
11.
12.
13.
14.
Lai CK, Moon Y, Kuchenbauer F, Starzcynowski DT, Argiropoulos B, Yung E, Beer P, Schwarzer A, Sharma A, Park G, Leung M, Lin G, Vollett
S, Fung S, Eaves CJ, Karsan A, Weng AP, Humphries RK & Heuser M. Cell fate decisions in malignant hematopoiesis: leukemia phenotype is
determined by distinct functional domains of the MN1 oncogene. PLoS One 19: e112671 2014.
Imren S, Heuser M, Gasparetto M, Beer PA, Norddahl GL, Xiang P, Chen L, Berg T, Rhyasen GW, Rosten P, Park G, Moon Y, Weng AP, Eaves
CJ & Humphries RK. Modeling de novo leukemogenesis from human cord blood with MN1 and NUP98HOXD13. Blood 124: 3608-3612,
2014.
Nguyen LV, Cox C, Eirew P, Knapp DJHF, Pellacani D, Kannan N, Carles A, Moksa M, Balani S, Shah S, Hirst M, Aparicio S & Eaves CJ. DNA
barcoding reveals diverse growth kinetics of human breast tumour subclones in serially passaged xenografts. Nature Comm 5:5871, 2014
Mingay M & Hirst M. The Epigenomic Toolkit. Drug Discovery Today: Disease Models. DOI: 10.1016/j.ddmod.2014.05.004. Epub 2014
August 22.
Deveau A, Forrester A, Coombs A, Wagner G, Grabher C, Chute I, Léger D, Mingay M, Alexe G, Rajan V, Liwski R, Hirst M, Stegmaier K,
Lewis S, Look T, and Berman J. Epigenetic therapy restores normal hematopoiesis in a zebrafish model of NUP98-HOXA9-induced myeloid
disease. Leukemia, Leukemia. 2015 May 28. doi: 10.1038/leu.2015.126.
Sharma A, Yun H, Jyotsana N, Chaturvedi A, Schwarzer A, Yung E, Lai CK, Kuchenbauer F, Argiropoulos B, Gorlich K, Ganser A, Humphries
RK & Heuser M. Constitutive Irf8 expression inhibits AML by activation of repressed immune response signaling. Leukemia 29: 57-68, 2015.
Giambra V, Jenkins C, Lam SH, Hoofd C, Belmonte M, Wang X, Gusscott S, Gracias D & Weng AP. Leukemia stem cells in T-ALL require
active Hif1α and Wnt signaling. Blood 125:3917-27, 2015.
Zhang X, Ma W, Cui J, Yao H, Zhou H, Ge Y, Xiao L, Hu X, Liu BH, Yang J, Li YY, Chen S, Eaves CJ, Wu D & Zhao Y. Regulation of p21 by
TWIST2 contributes to its tumor-suppressor function in human acute myeloid leukemia. Oncogene 34: 3000-10, 2015.
Beer PA, Knapp DJ, Miller PH, Kannan N, Sloma I, Heel K, Babovic S, Bulaeva E, Rabu G, Terry J, Druker BJ, Loriaux MM, Loeb KR, Radich
JP, Erber WN & Eaves CJ. Disruption of IKAROS activity in primitive chronic phase CML cells mimics myeloid disease progression. Blood
125:504-515, 2015.
Eaves CJ. Hematopoietic stem cells: concepts, definitions, and the new reality. Blood 125:2605-2613, 2015.
Patenaude A, Woerher S, Umlandt P, Wong F, Ibrahim R, Kyle A, Unger S, Fuller M, Parker J, Minchinton A, Eaves CJ & Karsan A. A Novel
Population of Local Pericyte Precursor Cells in Tumor Stroma that Require Notch Signaling for Differentiation. Microvasc Res. 101:38-47,
2015.
Cole A, Wang Z, Coyaud E, Voisin V, Gronda M, Jitkova Y, Mattson R, Hurren R, Babovic S, Maclean N, Restall I, Wang X, Jeyaraju DV,
Sukhai MA, Prabha S, Bashir S, Ramakrishnan A, Leung E, Qia YH, Zhang N, Combes KR, Ketela T, Lin F, Houry WA, Aman A, Al-Awar R,
Zheng W, Wienholds E, Xu CJ, Dick J, Wang JC, Moffat J, Minden MD, Eaves CJ, Bader GD, Hao Z, Kornblau SM, Raught B & Schimmer
AD. Inhibition of the mitochondrial protease ClpP as a therapeutic strategy for human acute myeloid leukemia. Cancer Cell. 27:864-76, 2015.
Sikorski DJ, Caron NJ, VanInsberghe M, Zahn H, Eaves CJ, Piret JM, Hansen CL. Clonal analysis of individual human embryonic stem cell
differentiation patterns in microfluidic cultures. Biotechnol J. [Epub head of print, Jun 9, 2015].
von Palffy S, Bulaeva E, Babovic S, Kannan N, Knapp D, Wei L, Eaves CJ, Beer P. Dominant negative IKAROS enhances IL-3-stimulated
signaling in wild-type but not BCR-ABL1+ mouse BA/F3 cells. Exp Hematol. 43:514-523, 2015.
32
The Terry Fox New Frontiers Program Project Grant in the genomics of forme fruste
tumours: new vistas on cancer biology and treatment (2013-2018)
Investigators: David Huntsman, Samuel Aparicio, Carl Hansen, Martin Hirst, Chenghan Lee, Marco Marra, Gregg Morin,
Ryan Morin, Torsten Nielsen, Sohrab Shah, Poul Sorensen, T. Michael Underhill, Stephen Yip
Co-applicants: Paul Clarkson, Jessica McAlpine, David Schaeffer, Anna Tinker
Scientific Summary
This PPG is an innovative approach to making clinically meaningful cancer discoveries. Forme fruste tumours are
clinically and pathologically homogenous tumour types that we believe our driven by a limited number of genetic
events. This property makes them ideal for research, but equally important, these tumours are in critical need of
improved diagnostics and treatments.
Additionally, discoveries made from the study of forme fruste tumours often have broader clinical relevance. Our
research program addresses the objective in three ways: (1) Attacking the cancer problem through the unique
perspective gained from the study of rare forme fruste tumour types; (2) Using state-of-the-art technology including
next generation sequencing and microfluidics to achieve our objectives; and (3) Addressing commonly overlooked
areas of cancer biology such as RNA editing and non-coding alterations, and alterations in epigenetic signatures. Our
team has been studying forme fruste tumours since 2010, and we have had an exceptional track record of identifying
mutations that are characteristic of forme fruste tumours and uncovering how these mutations drive tumour biology.
We have developed four inter-related sub-projects and one core facility that will work together to study these cancers:
Sub-project one is the main sequencing/discovery project and it will generate a complete catalogue of genomic
alterations in forme fruste tumours using a comprehensive suite of next-generation sequencing techniques.
Sub-project two will combine some of the deep sequencing data from sub-project one with newly generated
sequencing data for selected forme fruste tumours with known alterations to comprehensively describe the clonal subpopulations within these tumour types. This deep sequencing data will be analyzed to characterize the genotype of
individual cell populations within the tumours and how the different clonal mutation profiles have evolved. As part of
this sub-project, model xenograft systems will be studied, pre- and post- treatment, to determine how these tumours
evolve under the pressure of targeted therapeutics.
Sub-project three will use isogenic tumour cell line models to validate how genomic alterations identified in subprojects one and two will affect the epigenome, mutant protein expression, protein interaction networks, and
tumourigenic cellular phenotypes. Sequence based methods will query the epigenome, and proteomic methods will
identify effects on protein networks and the translatome. Drug screens will be used to confirm therapeutically
actionable targets and processes.
Sub-project four will optimize methods for measuring levels of circulating tumour DNA in forme fruste cancers as a
novel diagnostic and tumour monitoring tool. The circulating tumour DNA levels will be correlated with clinical
parameters.
These four sub-projects will be supported by a Data Analysis Core for bioinformatics analysis, statistical analysis, and
data analysis. The discoveries from this project will be translated into the clinic through our collaboration with the
SMART (Shared Access Medicine: An Approach to Rare Tumours), through the Center for Drug Research and
Development, and through collaborations with clinical trials groups. The work described in this program will help
improve the management and treatment for forme fruste tumours and provide general insights into the biology of
cancer.
33
List of Key Publications:
1.
2.
3.
4.
5.
6.
7.
8.
9.
10.
11.
12.
13.
14.
15.
16.
17.
18.
19.
Wang, Y., et al., The oncogenic toles of DICER1 RNase IIIb domain mutations in ovarian Sertoli-Leydig cell tumors. Neoplasia,
accepted.
Karnezis, A.N., et al., Dual loss of the SWI/SNF complex ATPases SMARCA4/BRG1 and SMARCA2/BRM is highly sensitive and
specific for small cell carcinoma of the ovary, hypercalcemic type. J Pathol, accepted.
Somasekharan, S.P., et al., YB-1 regulates stress granule formation and tumor progression by translationally activating G3BP1. J
Cell Biol, 2015. 208(7): p. 913-29.
Nielsen, T.O., N.M. Poulin, and M. Ladanyi, Synovial sarcoma: recent discoveries as a roadmap to new avenues for therapy. Cancer
Discov, 2015. 5(2): p. 124-34.
Jamshidi, F., T.O. Nielsen, and D.G. Huntsman, Cancer genomics: why rare is valuable. J Mol Med (Berl), 2015. 93(4): p. 369-81.
El-Naggar, A.M., et al., Translational Activation of HIF1alpha by YB-1 Promotes Sarcoma Metastasis. Cancer Cell, 2015. 27(5): p.
682-97.
Eirew, P., et al., Dynamics of genomic clones in breast cancer patient xenografts at single-cell resolution. Nature, 2015. 518(7539): p.
422-6.
Chu, Q.S., et al., A phase II study of SB939, a novel pan-histone deacetylase inhibitor, in patients with translocation-associated
recurrent/metastatic sarcomas-NCIC-CTG IND 200dagger. Ann Oncol, 2015. 26(5): p. 973-81.
Chen, J., et al., Recurrent DICER1 hotspot mutations in endometrial tumours and their impact on microRNA biogenesis. J Pathol,
2015.
Wiegand, K.C., et al., ARID1A/BAF250a as a prognostic marker for gastric carcinoma: a study of 2 cohorts. Hum Pathol, 2014.
45(6): p. 1258-68.
Twa, D.D., et al., Genomic rearrangements involving programmed death ligands are recurrent in primary mediastinal large B-cell
lymphoma. Blood, 2014. 123(13): p. 2062-5.
Roth, A., et al., PyClone: statistical inference of clonal population structure in cancer. Nat Methods, 2014. 11(4): p. 396-8.
Ramos, P., et al., Small cell carcinoma of the ovary, hypercalcemic type, displays frequent inactivating germline and somatic
mutations in SMARCA4. Nat Genet, 2014. 46(5): p. 427-9.
Jamshidi, F., et al., Diagnostic value of next-generation sequencing in an unusual sphenoid tumor. Oncologist, 2014. 19(6): p. 62330.
Ha, G., et al., TITAN: inference of copy number architectures in clonal cell populations from tumor whole-genome sequence data.
Genome Res, 2014. 24(11): p. 1881-93.
Witkowski, L., et al., DICER1 hotspot mutations in non-epithelial gonadal tumours. Br J Cancer, 2013. 109(10): p. 2744-50.
White, A.K., et al., High-throughput microfluidic single-cell digital polymerase chain reaction. Anal Chem, 2013. 85(15): p. 7182-90.
Sheffield, B.S. and T.O. Nielsen, Myxoid liposarcoma in a 91-year-old patient. Mol Cytogenet, 2013. 6(1): p. 50.
Anglesio, M.S., et al., Type-specific cell line models for type-specific ovarian cancer research. PLoS One, 2013. 8(9): p. e72162
34
The Terry Fox New Frontiers Program Project Grant in unraveling metabolic adaptations
associated with disease progression and therapeutic response in metastatic breast
cancer (2014-2015)
Investigators: Russell Jones, Nahum Sonenberg, Vincent Giguère, William Muller, Peter Siegel, Morag Park, McGill
University
Scientific Summary: Tumour cells reprogram a variety of their central metabolic and bioenergetic pathways to fuel
growth, survival and metastatic progression. In the context of breast cancer, these metabolic adaptations allow cancer
cells to deal with ever-changing conditions in the primary tumour (hypoxia) and foreign metastatic microenvironments.
Oncogenes and tumour suppressor genes encode proteins that regulate cellular metabolic pathways, causing a shift from
oxidative phosphorylation to aerobic glycolysis (“Warburg effect”). However, the mechanisms that induce and control
these metabolic adaptations remain largely undefined. Also, the impact that metabolic reprogramming has on metastatic
progression and therapeutic resistance, two key challenges in breast cancer management, remains to be elucidated.
Goals: The overall goals of this program are to identify and integrate key regulatory signaling and metabolic networks
that impact on poor outcome in breast cancers with a focus on metastatic progression and mechanisms of resistance of
HER2 and basal breast cancer.
Each of the individual projects addresses a unique issue regarding metabolic adaptation in breast cancer. Project # 1 (Dr.
Jones) investigates the impact of AMPK signaling as a key metabolic checkpoint in basal and HER2+ breast cancers.
Project # 2 (Dr. Sonenberg) unravels mTOR and translational regulation of metabolism during the development of
therapeutic resistance to targeted and chemotherapeutic agents. Project # 3 (Dr. Giguère) will define transcriptional
networks governed by the ERR/PGC-1 axis that regulate metabolic programming in breast cancer. Project # 4 (Dr. Muller)
will investigate how receptor tyrosine kinase (HER2) signaling modulates metabolic networks and therapeutic resistance.
Project # 5 (Dr. Siegel) explores how mitochondrial dysfunction drives the acquisition of aggressive breast cancer
phenotypes and how tumour/stromal interactions with the metastatic microenvironment alter tumour cell metabolism
in basal breast cancers.
List of Key Publications:
1. ERBB2 deficiency alters an E2F-1-dependent adaptive stress response and leads to cardiac dysfunction.
Perry MC1, Dufour CR2, Eichner LJ1, Tsang DW1, Deblois G1, Muller WJ3, Giguère V4.
Mol Cell Biol. 2014 Dec 1;34(23):4232-43. doi: 10.1128/MCB.00895-14. Epub 2014 Sep 22.
2. Serine deprivation enhances antineoplastic activity of biguanides.
Gravel SP1, Hulea L2, Toban N3, Birman E4, Blouin MJ4, Zakikhani M4, Zhao Y4, Topisirovic I5, St-Pierre J6, Pollak M7.
Cancer Res. 2014 Dec 15;74(24):7521-33. doi: 10.1158/0008-5472.CAN-14-2643-T. Epub 2014 Nov 6.
3. Lyn modulates Claudin-2 expression and is a therapeutic target for breast cancer liver metastasis.
Tabariès S1,2, Annis MG1,2, Hsu BE1,2, Tam CE1,2, Savage P1,2, Park M1,2,3,4, Siegel PM1,2,3.
Oncotarget. 2015 Apr 20;6(11):9476-87.
4. PDK1-Dependent Metabolic Reprogramming Dictates Metastatic Potential in Breast Cancer.
Dupuy F1,Tabariès S2, Andrzejewski S1, Dong Z2, Blagih J3, Annis MG2, Omeroglu A4, Gao D5, Leung S5, Amir E6, Clemons M7, AguilarMahecha A8, Basik M8, Vincent EE3, St-Pierre J1, Jones RG9, Siegel PM10.
Cell Metab. 2015 Oct 6;22(4):577-89. doi: 10.1016/j.cmet.2015.08.007. Epub 2015 Sep 10.
5. Mitochondrial Phosphoenolpyruvate Carboxykinase Regulates Metabolic Adaptation and Enables Glucose-Independent Tumor
Growth.
Vincent EE1, Sergushichev A2, Griss T1, Gingras MC3, Samborska B1, Ntimbane T4, Coelho PP1, Blagih J1, Raissi TC1, Choinière L4,
Bridon G4, Loginicheva E5, Flynn BR1, Thomas EC6, Tavaré JM6, Avizonis D4, Pause A3, Elder DJ6, Artyomov MN7, Jones RG8.Mol
Cell. 2015 Oct 15;60(2):195-207. doi: 10.1016/j.molcel.2015.08.013.
35
The Terry Fox New Frontiers Program Project Grant in discovery and therapeutic
development of antibody-based targets in oncology (2015-2018)
Investigators: Steven Jones, John Babcook, François Bénard, Kuo-Shyan Lin, Gregg Morin, Paul Schaffer, Tomas Hudlicky
Funding Partner: BioCanRx
Scientific Summary: This proposal brings together a multidisciplinary team of experts from academia and industry
focused on developing therapeutic antibody-based diagnostics, theranostics and therapeutics for newly discovered
tumour-associated targets. It stands to bring together world-class genomics, bioinformatics, and proteomics capabilities
(Genome Sciences Centre, GSC) with both an established and a novel antibody-drug conjugate platform (Centre for Drug
Research and Development, CDRD) and a clinically integrated imaging platform (BC Cancer Agency, BCCA). Underlying all
of these individual programs is the cutting-edge antibody generation platform established at the CDRD which will
generate panels of antibodies and derivatives to validate the targets identified from the bioinformatics program, and
become the basis of therapeutics for the antibody-drug conjugate program and companion theranostics for the imaging
program. Together this team will generate a suite of fully validated antibody-based therapeutics with matched
theranostics which could be used individually or combined for next-generation targeted therapy.
Targets
Deep Sequencing for
mAb-Specific Target
Discovery & Validation
Project 1
•RNAseq & Bioinformatics
•Splice variants, gene fusions
•Multiple tumour types
•Protein target validation
•mAb characterization
mAbs
Therapeutic
Antibody Generation
(Core)
mAbs
Antibody-Drug
Conjugate Platform
Development
Project 2
•Potent cytotoxic conjugates
•Novel cleavable Linkers
•Non-conventional Toxins
Radioimmunoconjugate
Platform Development
Project 3
•Diagnostics
•Theranostics
•Radioimmunotherapeutics
Project Outcome
Suite of Multi-Modal Therapeutics with Matched Theranostics
List of Key Publications:
Beck A, Wurch T, Bailly C, Corvaia N. 2010. Strategies and challenges for the next generation of therapeutic antibodies. Nat Rev
Immunol 10(5): 345-352.
Carter PJ, Senter PD. 2008. Antibody-drug conjugates for cancer therapy. Cancer J 14(3): 154-169.
Dargahi D, Swayze RD, Yee L, Bergqvist PJ, Hedberg BJ, Heravi-Moussavi A, Dullaghan EM, Dercho R, An J, Babcook JS, Jones SJ.
2014. A pan-cancer analysis of alternative splicing events reveals novel tumor-associated splice variants of matriptase. Cancer Inform.
13: 167-177.
Hughes B. 2010. Antibody-drug conjugates for cancer: poised to deliver? Nat Rev Drug Discov 9(9): 665-667.
Nelson AL, Dhimolea E, Reichert JM. 2010. Development trends for human monoclonal antibody therapeutics. Nat Rev Drug Discov
9(10): 767-774.
Webb S. 2011. Pharma interest surges in antibody-drug conjugates. Nat Biotechnol 29(4): 297-298
36
The Terry Fox New Frontiers Program Project in Li-Fraumeni Syndrome: Applying genetic
determinants of cancer risk to cancer surveillance and prevention (2015-2018)
Investigators: David Malkin, Adam Shlien, Anna Goldenberg, Andrea Doria, Hospital for Sick Children, University of
Toronto; Jason Berman, IWK Health Centre, Dalhousie University
Scientific Summary: Li-Fraumeni Syndrome (LFS) is a highly penetrant autosomal dominantly inherited predisposition
syndrome associated with a remarkably heterogeneous presentation of early onset cancers. In 1990, the PI (Malkin)
discovered that germline TP53 mutations cause >80% of LFS. Since then, this group has demonstrated that TP53
mutations occur with striking frequency in a wide spectrum of patients with component LFS tumours, with or without a
family history of cancer.
In addition, epigenetic, genomic and genetic events have been found to modify the phenotypic effects of an underlying
germline TP53 mutation. We have elucidated a mechanistic model to explain tumour initiation/progression in LFS in
which a constitutional (or possibly early somatic) TP53 mutation favours accumulation of epigenetic or genetic events
that facilitate accelerated telomere attrition, chromothripsis, and subsequent somatic cell transformation. We
developed a clinical surveillance protocol that takes advantage of innovative imaging techniques such as rapid sequence
whole body MRI (WB-MRI), ultrasonography and biochemical tests to detect occult malignancy. This approach improves
survival and reduces treatment-related morbidity. Our protocol has been rapidly adopted worldwide.
Notwithstanding this extraordinary progress, patients with LFS continue to face seemingly insurmountable challenges:
1) It is impossible to prevent cancers from developing;
2) It is impossible to prevent therapy-induced cancers from developing;
3) It is impossible to predict what types of cancer will develop and at what age; and
4) It is extremely difficult to effectively treat these patients who face dismal survival rates with devastating treatmentrelated toxicities.
We propose to address these challenges through the following interwoven projects:
Projects 1-2: We will define the epigenetic and genetic modifiers that confer specific phenotypes in TP53 mutation
carriers and create multi-level algorithms merging genetic and clinic-pathologic data to refine tumour type and age of
onset risk estimates in TP53 mutation carriers.
Project 3: We will use this information to refine and implement novel MRI-based surveillance strategies, and to create
molecular surveillance techniques for early tumor detection.
Project 4: We will explore potential chemoprevention strategies for TP53 mutation carriers using a powerful zebrafish
p53 model ().
Each project complements the others and addresses a key element of the continuum from molecular genotyping through
risk stratification to translational strategies for early tumour detection and cancer prevention in Li-Fraumeni Syndrome.
We anticipate that these studies will lead to a better understanding the role of early p53 alterations in cancer generally,
and to transform the care of patients with Li-Fraumeni Syndrome.
37
The Terry Fox New Frontiers Program Project Grant in the genetic analysis of signaling
pathways for vascular development and tumour angiogenesis (2010-2015)
Investigators: Andras Nagy, Anthony Pawson, Jeff Wrana, Susan Quaggin; Samuel Lunenfeld Research Institute, Mount
Sinai Hospital, Hao Ding; University of Manitoba, Janet Rossant; Hospital for Sick Children
Scientific Summary: Anti-angiogenic therapy is one of the most promising cancer treatments. It prevents the
formation of new blood vessels and thereby hinders tumours from growing. This treatment, however, comes with a
long list of severe side effects, including high blood pressure and kidney failure, which limits the full utilization of this
powerful approach to fighting cancer. In this program project, we join the expertise of six laboratories to overcome
these limitations.
 The Nagy Lab is committed to obtain mechanistic insights into the broad range of consequences of anti-angiogenic
therapies on normal physiology including the immune system.
 The Quaggin Lab focuses on finding ways to protect the kidney and other organs from damage during antiangiogenic therapy.
 The Rossant Lab uses stem cells to increase the quality of blood vessels so that the delivery of chemotherapy can
be enhanced. They also develop robust cell-based screening tools to test new anti-angiogenic therapies.
 The Wrana Lab has developed a system to measure the movement of cancer cells. They use this to define novel
pathways mediating cancer metastasis via stromal tumour interactions.
 The Ding Lab investigates how the PDGF gene, that plays an important role in vessel formation, is involved in
medulloblastomas, the most common type of brain tumour in children.
 The Pawson Lab studies how disruptions of cell-cell interactions can cause cancer.
 There are two core facilities serving our laboratories; one maintains and distributes genetically modified mouse
lines for cancer modeling and the other provides high throughput screens.
List of Key Publications:
1.
Adipose Vascular Endothelial Growth Factor Regulates Metabolic Homeostasis through Angiogenesis. Sung HK, Doh KO, Son JE,
Park JG, Bae Y, Choi S, Nelson SML, Cowling R, Nagy K, Michael IP, Koh GY, Adamson SL, Pawson A, Nagy A. (2013) Cell
Metabolism. Volume 17, Issue 1, 61-72, 8 January 2013. PMID: 23312284
2.
Soluble FLT1 Binds Lipid Microdomains in Podocytes to Control Cell Morphology and Glomerular Barrier Function. Jin J, Sison
K, Li C, Tian R, Wnuk M, Sung HK, Jeansson M, Zhang C, Tucholska M, Jones N, Kerjaschki D, Shibuya M, Fantus IG, Nagy A,
Gerber HP, Ferrara N, Pawson T, Quaggin SE. (21012) Cell. 2012 Oct 12;151(2):384-99. PMID: 23063127
3.
Interaction domains of Sos1/Grb2 are finely tuned for cooperative control of embryonic stem cell fate. Findlay GM, Smith MJ,
Lanner F, Hsiung MS, Gish GD, Petsalaki E, Cockburn K, Kaneko T, Huang H, Bagshaw RD, Ketela T, Tucholska M, Taylor L,
Bowtell DD, Moffat J, Ikura M, Li SS, Sidhu SS, Rossant J, Pawson T. Cell. 2013 Feb 28;152(5):1008-20. PMID:23452850
4.
The adaptor protein Grb2 is not essential for the establishment of the glomerular filtration barrier. Bisson N, Ruston J, Jeansson
M, Vanderlaan R, Hardy WR, Du J, Hussein SM, Coward RJ, Quaggin SE, Pawson T. PLoS One. 2012;7(11):e50996. Epub 2012
Nov 30. PMID:23226445
5.
Exosomes mediate stromal mobilization of autocrine Wnt-PCP signaling in breast cancer cell mtility. Luga, V.*, Zhang, L.*,
Viloria-Petit, A., Ogunjimi, A., Inanlou, M., Chiu, E., Nasser Hosein, A., Buchanan, M., Basik, M. and Wrana, J.L. (2012). Cell, 151,
1542-1556. PMID: 23260141
38
Molecular and cellular differentiation: New targets and treatments (2009-2015)
Investigators: Christopher Paige, (Project Leader), Norman Iscove, John Dick, Robert Rottapel, Ben Neel, Juan Carlos
Zuniga-Pflucker (OCI ,UHN); Tak Mak and Pam Ohashi, (Campbell Family Institute for Breast Cancer Research, OCI,
UHN).
Scientific Summary: Fully functional mature cells differentiate from progenitors through a series of stages regulated by
genes and proteins. Our Terry Fox Program Project group studies both the normal process of differentiation and the
changes that occur when malignancies arise. We define the molecular differences that serve to drive the
transformation and progression of cancer cells. Based on this information we develop and use novel technologies to
find targets for therapeutic intervention. We study both solid and dispersed cancers using both mouse and human
models with a particular emphasis on ovarian cancer, as an example of solid tumors, and leukemia. In addition to
molecules which might be targets of drug or biological therapy, we also use the latest understanding of the cells and
cytokines which drive immunity to develop novel protocols to harness the power of the immune system to recognize
and eliminate cancer cells.
List of Key Publicatons:
1.
Sriskanthadevan S, Jeyaraju DV, Chung TE, Prabha S, Xu W, Skrtic M, Jhas B, Hurren R, Gronda M, Wang X, Jitkova Y, Sukhai
MA, Lin FH, Maclean N, Laister R, Goard CA, Mullen PJ, Xie S, Penn LZ, Rogers IM, Dick JE, Minden MD, Schimmer AD. AML
cells have low spare reserve capacity in their respiratory chain that renders them susceptible to oxidative metabolic stress. Blood.
2015 125(13):2120-30.
2.
Laurenti E, Frelin C, Xie S, Ferrari R, Dunant CF, Zandi S, Neumann A, Plumb I, Doulatov S, Chen J, April C, Fan JB, Iscove N,
Dick JE. CDK6 Levels Regulate Quiescence Exit in Human Hematopoietic Stem Cells. Cell Stem Cell. 2015 16(3):302-13.
3.
Nguyen LT and Ohashi, PS. Clinical blockade of PD1 and LAG3 – potential mechanisms of action. Nature Reviews Immunol.
2015. (15):45-56.
Nelles ME, Paige CJ. CD4+ T cell plasticity engenders robust immunity in response to cytokine therapy. OncoImmunology. 2015
4(3).
4.
5.
Kreso A, van Galen P, Pedley NM, Lima-Fernandes E, Frelin C, Davis T, Cao L, Baiazitov R, Du W, Sydorenko N, Moon Y-C,
Gibson, Wang Y, Leung C, Iscove NN, Arrowsmith CH, Szentgyorgyi E, Gallinger S, Dick JE, O’Brien CA. Self-renewal as a
therapeutic target in human colorectal cancer. Nat. Med. 2014 20(1):29–36.
6.
Laurenti E, Frelin C, Xie S, Ferrari R, Dunant CF, Zandi S, Neumann A, Plumb I, Doulatov S, Chen J, April C, Fan JB, Iscove NN,
Dick JE. CDK6 levels regulate quiescence exit in human hematopoietic stem cells. Cell Stem Cell. 2015 16(3):302-13.
7.
van Galen P, Kreso A, Mbong N, Wienholds E, Xie S, Laurenti E, Eppert K, Wouters BG, Dick JE The unfolded protein response
governs integrity of the human hematopoietic stem cell pool during stress. Nature 2014 (7504):268-72.
8.
Theocharides APA, Dobson SM, Laurenti E, Notta F, Voisin V, Cheng PY, Yuan JS, Guidos CJ, Minden, MD, Mullighan, CG,
Torlakovic E and Dick JE Dominant-negative Ikaros cooperates with BCR-ABL1 to induce human acute myeloid leukemia in
xenografts. Leukemia 2015 29(1):177-87.
9.
Moti N, Malcolm T, Hamoudi R, Mian S, Garland G, Hook CE, Burke GA, Wasik MA, Merkel O, Kenner L, Laurenti E, Dick JE,
Turner SD. Anaplastic large cell lymphoma-propagating cells are detectable by side population analysis and possess an expression
profile reflective of a primitive origin. Oncogene. 2014 34(14):1843-52.
10. Dick JE. Tumor archaeology: tracking leukemic evolution to its origins. Sci Transl Med. 2014 (238):238fs23.
11. Qiao W, Wang W, Laurenti E, Turinsky AL, Wodak SJ, Bader GD, Dick JE, Zandstra PW. Intercellular network structure and
regulatory motifs in the human hematopoietic system. Mol Syst Biol. 2014 10(7):741.
12. Nelles ME, Moreau JM, Furlonger CL, Berger A, Medin JA, Paige CJ. 2014. Murine Splenic CD4+ T cells, induced by innate
immune cell interactions and secreted factors, develop anti-Leukemia cytotoxicity. Cancer Immunol. Res. 2014 2(11):1113-1124.
13. Liu TW, Stewart JM, MacDonald TD, Chen J, Clarke B, Shi J, Wilson BC, Neel BG, Zheng G, Biologically-targeted detection of
primary and micro- metastatic ovarian cancer Theranostics. 2013 3(6):420-427.
14. Frelin C, Herrington R, Janmohamed S, Barbara M, Tran G, Paige CJ, Benveniste P, Zuñiga-Pflücker J-C, Souabni A, Busslinger
M, Iscove NN. Gata3 regulates the self-renewal of long-term hematopoietic stem cells. Nat. Immunol. 2013 10:1037-44.
39
15. Wei LZ, Xu Y, Nelles ME, Furlonger C, Wang JCM, Di Grappa MA, Khokha R, Medin JA, Paige CJ. Localized interleukin-12
delivery for immunotherapy of solid tumors. Localized interleukin-12 delivery for immunotherapy of solid tumors. J. Cell Mol
Med. 2013 11:1465-74.
16. Lin AE, Ebert G, Ow Y, Preston SP, Toe JG, Cooney JP, Scott HW, Sasaki M, Saibil SD, Dissanayake D, Kim RH, Wakeham A,
You-Ten A, Shahinian A, Duncan G, Silvester J, Ohashi PS, Mak TW, Pellegrini M. AR1H2 is essential for embryogenesis, and its
hematopoietic deficiency causes lethal activation of the immune system. Nat. Immunol. 2013 14: 27-33.
17. Johnson DJ, Pao L, Dhanji S, Murakami K, Ohashi PS, Neel BG. Shp1 regulates T cell homeostasis by limiting IL-4 signals. J.
Exp. Med. 2013 209: 77-9.
18. Berger A, Frelin C, Shah DK, Benveniste P, Herrington R, Gerard NP, Zuniga-Pflucker J-C, Nscove NN, Paige CJ. Neurokinin-1
receptor signalling impacts bone marrow repopulation efficiency. PLoS One 2013 8:e58787.
19. Laurenti E, Doulatov S, Zandi S, Plumb I, Chen J, April C, Fan JB, Dick JE. The transcriptional architecture of early human
hematopoiesis identifies multilevel control of lymphoid commitment. Nat Immunol. 2013 14(7):756-63.
20. Liu TW, Stewart JM, Macdonald TD, Chen J, Clarke B, Shi J, Wilson BC, Neel BG, Zheng G. Biologically-targeted detection of
primary and micro-metastatic ovarian cancer. Theranostics. 2013 3(6):420-7.
21. Marcotte R, Brown KR, Suarez F, Sayad A, Karamboulas K, Krzyzanowski PM, Sircoulomb F, Medrano M, Fedyshyn Y, Koh JL,
van Dyk D, Fedyshyn B, Luhova M, Brito GC, Vizeacoumar FJ, Vizeacoumar FS, Datti A, Kasimer D, Buzina A, Mero P, Misquitta
C, Normand J, Haider M, Ketela T, Wrana JL, Rottapel R, Neel BG, Moffat J. Essential gene profiles in breast, pancreatic, and
ovarian cancer cells. Cancer Discov. 2012 2(2):172-8.
22. Chen G, Dimitriou I, Milne L, Lang KS, Lang PA, Fine N, Ohashi PS, Kubes P, Rottapel R. The 3BP2 adapter protein is required
for chemoattractant-mediated neutrophil activation. 2012 J Immunol. 2012 189(5):2138-50.
23. Chio C, Sasaki M, Ghazarian D, Moreno J, Done S, Ueda, T Inoue S, Chang Y-L, Chen NJ, Mak TW. TRADD contributes to tumour
suppression by regulating ULF-dependent p19Arf ubiquitylation. Nature Cell Biol. 2012 14: 625-633.
24. Dervovic DD, Ciofani M, Kianizad K, Zuniga-Pflucker JC. Comparative and functional evaluation of in vitro generated to ex vivo
CD8 T cells. J Immunol 2012 189:3411-3420.
25. Brustle A, Brenner D, Knobbe CB, Lang PA, Virtanen C, Hershenfield BM, Reardon C, Lacher SM, Ruland J, Ohashi PS, Mak TW.
The NF-B regulator MALT1 determines the encephalitogenic potential of Th17 cells. J. Clin. Invest. 2012 122: 4698-709.
26. Weinacht KG, Brauer PM, Felgentreff K, Devine A, Gennery AR, Giliani S, Al-Herz W, Schambach A, Zuniga-Pflucker JC,
Notarangelo LD. The role of induced pluripotent stem cells in research and therapy of primary immunodeficiencies. Curr Opin
Immunol. 2012.
27. Kennedy, M., G. Awong, C. M. Sturgeon, A. Ditadi, R. LaMotte-Mohs, J. C. Zuniga-Pflucker, and G. Keller. T lymphocyte potential
marks the emergence of definitive hematopoietic progenitors in human pluripotent stem cell differentiation cultures. Cell reports
2012. 2:1722-1735.
28. McPherson AJ, Snell LM, Mak TW, Watts TH. Opposing Roles for TRAF1 in the Alternative versus Classical NF-kappaB Pathway
in T Cells. J. Biol. Chem. 2012 287: 23010-9.
29. Levaot N, Voytyuk O, Dimitriou I, Sircoulomb F, Chandrakumar A, Deckert M, Krzyzanowski PM, Scotter A, Gu S, Janmohamed
S, Cong F, Simoncic PD, Ueki Y, La Rose J, Rottapel R. Loss of Tankyrase-mediated destruction of 3BP2 is the underlying
pathogenic mechanism of cherubism. Cell 2011 147(6):1324-39.
30. Lewis M, Meza-Avina ME, Wei L, Crandall IE, Bello AM, Poduch E, Liu Y, Paige CJ, Kain KC, Pai EF, Kotra LP. Novel
interactions of fluorinated nucleotide derivatives targeting orotidine 5’-monophosphate decarboxylase. J. Med. Chem. 2011
54(8):2891-901.
31. Rosas-Ballina M, Olofsson PS, Ochani M, Valdés-Ferrer SI, Levine YA, Reardon C, Tusche MW, Pavlov VA, Andersoon U, Chavan
S, Mak TW, Tracey KJ. Acetylcholine-synthesizing T cells relay neural signals in a vagus nerve circuit. Science 2011 334: 98-10
40
The Terry Fox New Frontiers Program Project Grant in nanoparticle-enhanced
photoacoustic imaging for cancer localization and therapeutic guidance (2013-2017)
Investigators: Brian Wilson, Princess Margaret Cancer Center (Project Leader and Sub-project 3 Leader); Stuart Foster,
Sunnybrook Research Institute (Sub-project 1 Leader); Gang Zheng, UHN (Sub-project 2 Leader); John Trachtenberg,
Ralph DaCosta, Princess Margaret Cancer Centre; Norman Marcon, Cathy Streutker, Maria Cirocco, St. Michael’s
Hospital; Theo van der Kwast, Robert Weersink, UHN; Linda Sugar, Masoom Haider, Sunnybrook Research Institute
Scientific Summary: This new project aims to develop a novel technology platform to be applied to two significant
unmet clinical needs. The first technology component is photoacoustic imaging, combining the molecular specificity of
light with the deep imaging of ultrasound, and which Dr Foster has pioneered. The second is all-organic nanoparticles
(“porphysomes”) discovered in Dr Zheng’s lab that have an exceptional properties for imaging and therapeutics. Here,
they are to be used primarily for their high photoacoustic image contrast. The first unmet need is to treat patients with
low/intermediate-risk “focal” prostate cancer (PCa) with minimal risk to normal tissues. This builds on previous work
using laser photothermal therapy. The nanoparticle-enhanced photoacoustic imaging platform is intended to improve
the efficacy and safety of this approach and to facilitate its cost-effective dissemination and clinical adoption. The
second unmet need is to detect high-grade dysplasia in patients with Barrett’s Esophagus (BE) and to ensure absence
of submucosal invasion so that minimally-invasive endoscopic mucosal resection can be performed, reducing the need
for esophagectomy and associated morbidity. In both applications, the long term goal is to change the balance
between achieving effective tumor control and the side effects of radical therapies that significantly impact quality of
life. The project comprises three subprojects, as follows:
Sub-project 1: “Photoacoustic Imaging Technology” focuses on the design, fabrication, testing and optimization of
hardware and software for photoacoustic imaging. The technology is based on prior development of high-resolution)
ultrasound imaging,[1] extending this to photoacoustic mode.[2] Two different photoacoustic probes are under
development. The first is a transrectal device for imaging of the prostate, while the second is intended for endoscopic
use. These two probes will be validated in animal models in vivo before translating to the first-in-human studies in Subproject 3. The transrectal probe is under constructed in collaboration with industry partners. The endoscopic probe has
been designed to incorporate a miniaturized ultrasound transducer using a novel fabrication technique.
Sub-project 2: “Porphysomes for Photoacoustic Imaging” comprises further development and optimization for the
clinical applications of two different forms of porphysomes.[3,4] The first, pyro-porphysomes, are intended as
photoacoustic image contrast agents and have been validated for intrinsic tumor targeting in various tumor models.[5]
Both photoacoustic and fluorescence[6] imaging are possible with the same nanoparticles, which is particularly valuable
for endoscopic applications. An animal model of BE has been established to enable pre-clinical testing and optimization
of porphysomes for this specific application. The second, J-porphysomes, are designed to enable real-time 3-D imaging
of the temperature distribution during photothermal treatment of focal prostate cancer, enabling optimal treatment
delivery. The principle of this approach has been demonstrated.[7] In order to translate porphysomes into clinical trials
(Sub-project 3), scale-up, gmp (good manufacturing practice) and toxicity testing are required. These are in progress,
with additional internal and external support.
Sub-project 3: “First-in-Human Studies of PAI and Porphysomes in PCa and GI” comprises a series of small-scale studies
in patients, piggybacking on current clinical practice or ongoing clinical trials, with the intent to obtain first-in-human
data on safety, technical feasibility and performance of the different photoacoustic/porphysome combinations. The
first human ex vivo tissue studies of intrinsic photoacoustic contrast are in progress: a) in intact prostatectomy
specimens to assess tumor delineation and b) in endoscopic mucosal resection specimens to assess altered
microvascular patterns.[8] Investigations of quantitative imaging with topically-applied porphysomes is also in progress.
Additional planned PCa studies comprise: intrinsic and porphysome-enhanced photoacoustic imaging in combination
with MRI for tumor localization; photoacoustic imaging during and post photothermal treatment to assess response;
and thermal mapping using J-porphysomes during treatment. In the GI track, the transrectal system will also be used
41
to image rectal cancer, while the endoscopic system will be used for in vivo imaging in BE patients. These studies will
inform subsequent definitive clinical trials of the new technologies.
References:
1. Pavlovich CP, et al., High-resolution transrectal ultrasound: Pilot study of a novel technique for imaging clinically localized
prostate cancer. Urol Oncol. 34: e27-32, 2014.
2. Castelino RF, Lee H and Foster FS, Multi-frequency intravascular imaging probe for ultrasound and frequency domain
photoacoustic imaging, Proc. SPIE, 2015
3. Ng KK and Zheng G, Molecular Interactions in Photonic Organic Nanoparticles: Principles and Theranostic Applications,
4.
5.
6.
7.
8.
Chemical Reviews 2015, in press. doi 10.1021/acs.chemrev.5b00140.
Shakiba M, Chen J and Zheng G, Porphyrin Nanoparticles in Photomedicine, in Applications of Nanoscience to Photomedicine,
Ch 26, Chandros Publishing, 2015.
Muhanna N, MacDonald TD, Chan H, Jin CS, Burgess L, Cui LY, Chen J, Irish JC and Zheng G, Multimodal Nanoparticle for
Primary Tumor Delineation and Lymphatic Metastasis Mapping in A Head-and-Neck Cancer Rabbit Model, Adv Healthcare
Materials 2015, in press. doi: 10.1002/adhm.201500363.
Ng KK, Takada M, Jin C and Zheng G, Self-Sensing FRET-Porphysomes for Fluorescence-Guided Photothermal Therapy,
Bioconjugate Chemistry 26: 345-51, 2015.
Ng KK, Shakiba M, Huynh E, Weersink RA, Roxin Á, Wilson BC and Zheng G, Stimuli-responsive photoacoustic nanoswitch
for in vivo sensing applications, ACS Nano.8: 8363-73, 2014.
Lim L, Streutker CJ, Marcon NE, Cirocco M, Iakovlev VV, DaCosta R, Foster, FS and Wilson BC, Clinical study of ex vivo
photoacoustic imaging in endoscopic mucosal resection tissues, Proc. SPIE, 9323-06, 2015
42
The Terry Fox New Frontiers Program Project Grant: A research pipeline for hypoxiadirected precision cancer medicine (2014-2019)
Investigators: Robert Bristow, Bradley Wouters, Marianne Koritzinsky, Michael Milosevic, Anthony Fyles, and
David Jaffray
Scientific Summary: Cancer cells sense and respond to hypoxia through complex biological pathways that also
adversely affect patient prognosis. These include changes in cell signaling, proliferation, metabolism, angiogenesis,
DNA repair, and metastasis. However, these pathways may also be amenable to precise targeting to offset aggressive
hypoxia-based phenotypes.
Our new TFRI program constitutes a research pipeline with short-, medium-, and long-term interactive goals aimed
directly at improving patient outcome by targeting or exploiting hypoxic cell phenotypes in tumours. This program
consists of five projects that include novel mechanistic studies to understand how hypoxia influences protein
expression relevant to metastasis, identification of new therapeutic targets and development of new biologics,
understanding the relationship between hypoxia and genetic instability, investigating the therapeutic potential of
targeting hypoxia and IFP-driven chemokine and bone marrow cell recruitment in tumours, and implementing imaging
and genomic-based personalized medicine approaches in hypoxia-directed clinical trials with new agents.
Our team will focus on using these innovative and combined approaches to improve cures in cervix, head and neck,
prostate, and pancreatic cancer using a “pipeline of basic science to clinical trials”.
Recent Discoveries and Accomplishments:
 Secretion of hypoxia-induced proteins is supported by superior disulfide bond formation in hypoxia.
 APEX2-mediated affinity tag purification facilitates cargo-specific interactions in the secretory pathway.
 Fumarate hydratase mediates metabolic reprogramming during hypoxia through PKM2.
 Expression of OCT3 is important for metformin uptake and response in cancer cells.
 Metformin as a novel biological modifier of radiotherapy.
 Flavoprotein POR is shown as a key determinant of sensitivity to the hypoxia-activated prodrug SN30000.
 Targeting tumour hypoxia to prevent cancer metastasis.
 Showed chromosomal instability as a prognostic marker in cervical cancer.
 Developing a prognostic micro-RNA signature for human cervical carcinoma.
 Establishment of an orthotopic primary cervix cancer xenograft model.
 Completed functional genomic screens on 29 head and neck cell lines
 Identified Notch3 as a therapeutic target in head and neck cancer
 Characterized the dynamic interactions among the tumor microenvironment, chemokines, BMDCs and
radiotherapy in curable cervix cancers.
 Developed a unique, image-guided targeted radiation protocol in mice to administer localized beams
specifically to the cervix tumour xenograft.
 Production of FAZA for hypoxia
 PET imaging is produced on site for clinical trials and pre-clinical studies.
 Initiation of Phase II study: metformin as a novel personalized biologic therapy in women with hypoxic cervix
cancer.
 Demonstrated that Plerixafor with combined standard treatment of radiochemotherapy was successful in
reducing tumour growth and metastasis.
List of Key Publications:
1.
Tan Q, Joshua A, Saggar J, Yu M, Wang M, Kanga N, Zhang J, Chen E, Wouters BG, Tannock I. Effect of Pantoprazole to enhance activity of docetaxel against
human tumor xenografts by inhibiting autophagy. Br J Cancer. 2015 Feb 3.
43
2.
Rupaimoole R, Wu SY, Pradeep S, Ivan C, Pecot CV, Gharpure KM, Nagaraja AS, Armaiz-Pena GN, McGuire M, Zand B, Dalton HJ, Filant J, Miller JB, Lu C,
Sadaoui NC, Mangala LS, Taylor M, van den Beucken T, Koch E, Rodriguez-Aguayo C, Huang L, Bar-Eli M, Wouters BG, Radovich M, Ivan M, Calin GA, Zhang
W, Lopez-Berestein G, Sood AK: Hypoxia-mediated downregulation of miRNA biogenesis promotes tumour progression. Nat Commun. 29; 5:5202 (2014)
3.
Chan N, Ali M, McCallum GP, Kumareswaran R, Koritzinsky M, Wouters BG, Wells PG, Gallinger S, Bristow RG: Hypoxia provokes base excision repair changes
and a repair-deficient, mutator phenotype in colorectal cancer cells. Mol Cancer Res.; 12(10): 1407-15 (2014)
4.
Edgar L, Vellanki R, Halupa A, Hedley D, Wouters BG, Nitz M. Identification of Hypoxic Cells Using an Organotellurium Tag Compatible with Mass Cytometry,
Angew Chem Int Ed Engl. 53(43):11473-7, 2014.
5.
Mujcic H, Hill RP, Koritzinsky M, Wouters BG. Hypoxia Signaling and the Metastatic Phenotype. Curr Mol Med. 14(5):565-79, 2014.
6.
Van Galen P, Kreso A, Mbong N, Wienholds E, Kent DG, Fitz-Maurice T, Chambers JE, Xie S, Laurenti E, Hermans K, Eppert K, Marciniak SJ, Goodall JC, Green
AR, Wouters BG, Dick JE, The unfolded protein response governs integrity of the HSC pool during stress. Nature. 510(7504),268-72, 2014.
7.
Gedye CA, Hussain A, Paterson J, Smrke A, Saini H, Sirskyj D, Pereira K, Lobo N, Stewart J, Go C, Ho J, Medrano M, Hyatt E, Yuan J, Lauriault S, Kondratyev
M, van den Beucken T, Jewett M, Dirks P, Guidos CJ, Danska J, Wouters BG, Neel B, Rottapel R, Ailles LE, Cell surface profiling using high-throughput flow
cytometry: a platform for biomarker discovery and analysis of cellular heterogeneity. PLoS One. 9(8): e105602, 2014.
8.
Joshua A, Zannella VE, Downes M, Bowes B, Hersey K, Koritzinsky M, Schwab M, Evans A, van der Kwast T, Trachtenberg J, Finelli A, Fleshner N, Sweet J,
Pollak M. A pilot “window of opportunity” neoadjuvant study of Metformin in Localised Prostate Cancer. Prostate cancer and Prostatic Diseases 2014,
Sep;17(3):252-8.
9.
Milosevic MF, Pintilie M, Hedley DW, Bristow RG, Wouters BG, Oza AM, Laframboise S, Hill RP, Fyles AW: High tumor interstitial fluid pressure identifies
cervical cancer patients with improved survival from radiotherapy plus cisplatin versus radiotherapy alone. Int J Cancer. 1; 135(7): 1692-9 (2014).
10.
van den Beucken T, Koch E, Chu K, Rupaimoole R, Prickaerts P, Adriaens M, Voncken JW, Harris AL, Buffa FM, Haider S, Starmans MH, Yao CQ, Ivan M, Ivan
C, Pecot CV, Boutros PC, Sood AK, Koritzinsky M, Wouters BG: Hypoxia promotes stem cell phenotypes and poor prognosis through epigenetic regulation of
DICER. Nat Commun. 29; 5:5203 (2014)
11.
Liu FF, Shi W, Done SJ, Miller N, Pintilie M, Voduc D, Nielsen TO, Nofech-Mozes S, Chang MC, Whelan TJ, Weir LM, Olivotto IA, McCready DR, Fyles AW:
Identification of a Low-Risk Luminal A Breast Cancer Cohort That May Not Benefit From Breast Radiotherapy. J Clin Oncol. (2015)
12.
Pettersen EO, Ebbesen P, Gieling RG, Williams KJ, Dubois L, Lambin P, Ward C, Meehan J, Kunkler IH, Langdon SP, Ree AH, Flatmark K, Lyng H, Calzada MJ,
Peso LD, Landazuri MO, Görlach A, Flamm H, Kieninger J, Urban G, Weltin A, Singleton DC, Haider S, Buffa FM, Harris AL, Scozzafava A, Supuran CT, Moser I,
Jobst G, Busk M, Toustrup K, Overgaard J, Alsner J, Pouyssegur J, Chiche J, Mazure N, Marchiq I, Parks S, Ahmed A, Ashcroft M, Pastorekova S, Cao Y,
Rouschop KM, Wouters BG, Koritzinsky M, Mujcic H, Cojocari D: Targeting tumour hypoxia to prevent cancer metastasis. From biology, biosensing and
technology to drug development: the METOXIA consortium. J Enzyme Inhib Med Chem. 27: 1-33 (2014)
13.
Tan Q, Joshua AM, Saggar JK, Yu M, Wang M, Kanga N, Zhang JY, Chen X, Wouters BG, Tannock IF: Effect of pantoprazole to enhance activity of docetaxel
against human tumour xenografts by inhibiting autophagy. Br J Cancer. 112(5): 832-40
(2015)
14.
Edgar LJ, Vellanki RN, Halupa A, Hedley D, Wouters BG, Nitz M. Identification of hypoxic cells using an organotellurium tag compatible with mass cytometry.
Angew Chem Int Ed Engl. 53(43): 11473-7 (2014)
15.
Boutros PC, Fraser M, Harding NJ, de Borja R, Trudel D, Lalonde E, Meng A, Hennings-Yeomans PH, McPherson A, Sabelnykova VY, Zia A, Fox NS, Livingstone
J, Shiah YJ, Wang J, Beck TA, Have CL, Chong T, Sam M, Johns J, Timms L, Buchner N, Wong A, Watson JD, Simmons TT, P'ng C, Zafarana G, Nguyen F, Luo
X, Chu KC, Prokopec SD, Sykes J, Dal Pra A, Berlin A, Brown A, Chan-Seng-Yue MA, Yousif F, Denroche RE, Chong LC, Chen GM, Jung E, Fung C, Starmans
MH, Chen H, Govind SK, Hawley J, D'Costa A, Pintilie M, Waggott D, Hach F, Lambin P, Muthuswamy LB, Cooper C, Eeles R, Neal D, Tetu B, Sahinalp C, Stein
LD, Fleshner N, Shah SP, Collins CC, Hudson TJ, McPherson JD, van der Kwast T, Bristow RG: Spatial genomic heterogeneity within localized, multifocal
prostate cancer. Nat Genet. (2015)
16.
Berlin A, Cho E, Kong V, Howell KJ, Lao B, Craig T, Bayley A, Chung P, Gospodarowicz M, Warde P, Catton C, Bristow RG, Ménard C: Phase 2 trial of guidelinebased postoperative image guided intensity modulated radiation therapy for prostate cancer: Toxicity, biochemical, and patient-reported health-related qualityof-life outcomes. Pract Radiat Oncol. (2015)
17.
Rupaimoole R, Wu SY, Pradeep S, Ivan C, Pecot CV, Gharpure KM, Nagaraja AS, Armaiz-Pena GN, McGuire M, Zand B, Dalton HJ, Filant J, Miller JB, Lu C,
Sadaoui NC, Mangala LS, Taylor M, van den Beucken T, Koch E, Rodriguez-Aguayo C, Huang L, Bar-Eli M, Wouters BG, Radovich M, Ivan M, Calin GA, Zhang
W, Lopez-Berestein G, Sood AK: Hypoxia-mediated downregulation of miRNA biogenesis promotes tumour progression. Nat Commun. (2014)
18.
Dodbiba L, Teichman J, Fleet A, Thai H, Starmans MH, Navab R, Chen Z, Girgis H, Eng L, Espin-Garcia O, Shen X, Bandarchi B, Schwock J, Tsao MS, ElZimaity H, Der SD, Xu W, Bristow RG, Darling GE, Boutros PC, Ailles LE, Liu G: Appropriateness of using patient-derived xenograft models for pharmacologic
evaluation of novel therapies for esophageal/gastro-esophageal junction cancers. PLoS One. (2015)
19.
Lalonde E, Ishkanian AS, Sykes J, Fraser M, Ross-Adams H, Erho N, Dunning MJ, Halim S, Lamb AD, Moon NC, Zafarana G, Warren AY, Meng X, Thoms J,
Grzadkowski MR, Berlin A, Have CL, Ramnarine VR, Yao CQ, Malloff CA, Lam LL, Xie H, Harding NJ, Mak DY, Chu KC, Chong LC, Sendorek DH, P'ng C, Collins
CC, Squire JA, Jurisica I, Cooper C, Eeles R, Pintilie M, Dal Pra A, Davicioni E, Lam WL, Milosevic M, Neal DE, van der Kwast T, Boutros PC, Bristow RG:
Tumour genomic and microenvironmental heterogeneity for integrated prediction of 5-year biochemical recurrence of prostate cancer: a retrospective cohort
study. Lancet Oncol. 15(13):1521-32 (2014)
20.
Cui L, Tse K, Zahedi P, Harding SM, Zafarana G, Jaffray DA, Bristow RG, Allen C: Hypoxia and cellular localization influence the radiosensitizing effect of gold
nanoparticles (AuNPs) in breast cancer cells. Radiat Res. 182(5):475-88 (2014)
44
STP collaboration with the Centre for Drug Research and Development (CDRD)
Terry Fox Research Institute Translational Cancer Research Project (2013-2015)
Investigators: John Babcook, CDRD; Rob Rottapel, Ben Neel, Brad Wouters, OCI; David Andrews, McMaster University;
Peter Dirks, Hospital for Sick Children; Daniel Durocher, Frank Sicheri, Samuel Lunenfeld Research Institute, Mount
Sinai Hospital; Jason Moffat, Sachdev Sidhu, UofT
Scientific Summary: The Selective Therapies Program (STP) is a translational program whose objective is to identify
novel cancer targets for which new anti-cancer therapeutics can be developed with heightened selective properties.
The STP has been able to identify novel cancer targets using high-throughput RNA interference (RNAi) screening
technologies and new high-throughput screening strategies. Dr. Sidhu has led the development of novel synthetic
antibodies to promising cancer drug targets identified by the STP. To further exploit the therapeutic potential of these
novel targets and antibodies, the Centre for Drug Research and Development (CDRD) will select antibodies generated
by the STP and conjugate their novel, potent cytotoxins to generate antibody-drug conjugates (ADCs). These ADCs will
be assessed for their ability to specifically deliver the toxin payloads and kill target-expressing tumour cells. Lead ADC
candidates will then be selected for further therapeutic development.
45
Canadian Colorectal Cancer Consortium (C4) (2012-2017):
Investigators: Gerald Batist, Jewish General Hospital, McGill University; and Steven Gallinger, Mount Sinai Hospital,
UHN
Scientific Summary: Colorectal cancer (CRC) is the most common malignancy of the gastrointestinal tract, and the
second leading cause of cancer death among Canadians. The more advanced the disease is at the time of diagnosis, the
greater the risk of metastases. The main strategies proven to ameliorate the health of Canadians with colorectal cancer
are: 1.) reducing risk, by improving outcomes through earlier diagnosis; and 2.) increasing therapeutic responses for
those with more advanced disease.
The uptake of CRC screening in most Canadian provinces remains too low for the potential impacts of screening to be
achieved across the population, even after recent efforts to increase screening rates. Although a variety of innovations
in treatment have dramatically improved the outcome for patients with Stage III and IV disease, the inevitable
development of therapeutic resistance remains the major obstacle to improving survival.
The overall objective of this project is to establish a molecular-based approach to translational cancer care that will
improve the outcome of CRC patients by:
1.) Increasing the impact of early diagnosis, decreasing mortality and the cost of managing CRC through targeted
screening of families stratified by risk (Screening Axis).
2.) Improving the life expectancy and reducing the cost of the management of advanced CRC through the study of drugresistant metastatic disease and the development of a biomarker panel to predict drug resistance (Therapeutic Axis).
This will be achieved by creating the C4, a Canadian multidisciplinary and inter-institutional network. A major outcome
of the C4 will be an integrated infrastructure for the development of a large-scale, molecular based approach to
translational cancer care for CRC. The C4 aims to use genetic data for the breadth of clinical challenges faced from the
time of diagnosis to the time of treatment. The C4, supported by TFRI, will put Canada at the forefront of translation al
cancer research allowing us to establish a unique and high-impact program.
List of Key Publications:
1. Identification of novel variants in colorectal cancer families by high-throughput exome sequencing. DeRycke MS, Gunawardena SR,
Middha S, Asmann YW, Schaid DJ, McDonnell SK, Riska SM, Eckloff BW, Cunningham JM, Fridley BL, Serie DJ, Bamlet WR, Cicek
MS, Jenkins MA, Duggan DJ, Buchanan D, Clendenning M, Haile RW, Woods MO, Gallinger SN, Casey G, Potter JD, Newcomb PA, Le
Marchand L, Lindor NM, Thibodeau SN, Goode EL. Cancer Epidemiol Biomarkers Prev. 2013 Jul;22(7):1239-51. doi: 10.1158/10559965.EPI-12-1226. Epub 2013 May 1.
2. Identification of Lynch syndrome among patients with colorectal cancer. Moreira L, Balaguer F, Lindor N, de la Chapelle A, Hampel
H, Aaltonen LA, Hopper JL, Le Marchand L, Gallinger S, Newcomb PA, Haile R, Thibodeau SN, Gunawardena S, Jenkins MA, Buchanan
DD, Potter JD, Baron JA, Ahnen DJ, Moreno V, Andreu M, Ponz de Leon M, Rustgi AK, Castells A; EPICOLON Consortium. JAMA.
2012 Oct 17;308(15):1555-65. doi: 10.1001/jama.2012.13088.
3.Biopsies: next-generation biospecimens for tailoring therapy. Basik M, Aguilar-Mahecha A, Rousseau C, Diaz Z, Tejpar S, Spatz A,
Greenwood CM, Batist G. Nat Rev Clin Oncol. 2013 Aug;10(8):437-50.
4.Next-generation biobanking of metastases to enable multidimensional molecular profiling in personalized medicine. Diaz Z, AguilarMahecha A, Paquet ER, Basik M, Orain M, Camlioglu E, Constantin A, Benlimame N, Bachvarov D, Jannot G, Simard MJ, Chabot B,
Gologan A, Klinck R, Gagnon-Kugler T, Lespérance B, Samson B, Kavan P, Alcindor T, Dalfen R, Lan C, Chabot C, Buchanan M,
Przybytkowski E, Qureshi S, Rousseau C, Spatz A, Têtu B, Batist G. Mod Pathol. 2013 Jun 7.
5.Physician recruitment of patients to non-therapeutic oncology clinical trials: ethics revisited. Black L, Batist G, Avard D, Rousseau C,
Diaz Z, Knoppers BM.Front Pharmacol. 2013;4:25.
46
Development of new treatment and biomarker for hepatocellular carcinoma: From
woodchuck to human
NSC-TFRI International Collaborative Research (2013-2016)
Investigators: John Bell, OHRI; Pei-Jer Chen, National Taiwan University College of Medicine
Scientific Summary: Lack of sensitive biomarkers for timing diagnosis and effective therapeutics for advanced tumours
are the two main reasons for the poor outcome of hepatocellular carcinomas (HCC). Therefore, there is a pressing
demand to develop new diagnosis and treatment strategies for HCC. Chronic hepatitis B virus (HBV) infection is one of
the major causes of HCC and the woodchuck (Marmota monax) chronically infected with woodchuck hepatitis virus
(WHV), a virus with high similarity to human HBV, recapitulates the complex liver milieu and natural course from
chronic HBV infection to HCC. It represents, and has been used, as an ideal preclinical model for HBV-related
translational studies. Despite the approval of the molecular targeted agent, sorafenib, for advanced HCC treatment, its
efficacy has been modest. New regimens, such as oncolytic viruses, are promising anti-cancer agents that deserve
further investigation to optimizing efficacy and safety in the relevant woodchuck model.
Therefore, the objectives of this project are:
 To adapt oncolytic virus-based therapeutics based on vaccinia virus or rhabdovirus platforms for testing against
liver cancer in the woodchuck hepatitis B model;
 To use surgical explants from woodchuck or human HCC subjects to study their susceptibilities to oncolytic virus
infection and to identify transcriptional or genomic markers that predict animal/patient HCC permissiveness for
oncolytic virus infection and treatment responses;
 To develop unique viral integration cellular junction DNA as a biomarker for follow-up of HCC growth and
treatment in woodchucks.
List of Key Publications:
1.
Tseng TC, Liu CJ, Chen CL, Yang HC, Su TH, Wang CC, Yang WT, Kuo SF, Liu CH, Chen PJ, Chen DS, Kao JH. Risk stratification of hepatocellular carcinoma in
hepatitis B virus e antigen-negative carriers by combining viral biomarkers. J Infect Dis. 2013 Aug; 208(4):584-93.
2.
Kim MK, Breitbach CJ, Moon A, Heo J, Lee YK, Cho M, Lee JW, Kim SG, Kang DH, Bell JC, Park BH, Kirn DH, Hwang TH. Oncolytic and immunotherapeutic vaccinia
induces antibody-mediated complement-dependent cancer cell lysis in humans. Sci Transl Med. 2013 May 15; 5(185):185ra63.
3.
Heo J, Reid T, Ruo L, Breitbach CJ, Rose S, Bloomston M, Cho M, Lim HY, Chung HC, Kim CW, Burke J, Lencioni R, Hickman T, Moon A, Lee YS, Kim MK,
Daneshmand M, Dubois K, Longpre L, Ngo M, Rooney C, Bell JC, Rhee BG, Patt R, Hwang TH, Kirn DH. Randomized dose-finding clinical trial of oncolytic
immunotherapeutic vaccinia JX-594 in liver cancer. Nat Med. 2013 Mar; 19(3):329-36.
4.
Le Boeuf F, Batenchuck C, Vähä-Koskela M, Breton S, Roy D, Lemay C, Cox J, Abdelbary H, Falls T, Waghray G, Atkins H, Stojdl D, Diallo JS, Kærn M, Bell JC. Modelbased rational design of an oncolytic virus with improved therapeutic potential. Nat Commun. 2013; 4:1974.
5.
Fletcher SP, Chin DJ, Ji Y, Iniguez AL, Taillon B, Swinney DC, Ravindran P, Cheng DT, Bitter H, Lopatin U, Ma H, Klumpp K, Menne S. Transcriptomic analysis of the
woodchuck model of chronic hepatitis B. Hepatology. 2012 Sep; 56(3):820-30.
6.
Russell SJ, Peng KW, Bell JC. Oncolytic virotherapy. Nat Biotechnol. 2012 Jul 10; 30(7):658-70.
7.
Freitas N, Salisse J, Cunha C, Toshkov I, Menne S, Gudima SO. Hepatitis delta virus infects the cells of hepadnavirus-induced hepatocellular carcinoma in
woodchucks. Hepatology. 2012 Jul; 56(1):76-85.
8.
Parato KA, Breitbach CJ, Le Boeuf F, Wang J, Storbeck C, Ilkow C, Diallo JS, Falls T, Burns J, Garcia V, Kanji F, Evgin L, Hu K, Paradis F, Knowles S, Hwang TH,
Vanderhyden BC, Auer R, Kirn DH, Bell JC. The oncolytic poxvirus JX-594 selectively replicates in and destroys cancer cells driven by genetic pathways commonly
activated in cancers. Mol Ther. 2012 Apr; 20(4):749-58.
9.
Jiang S, Yang Z, Li W, Li X, Wang Y, Zhang J, Xu C, Chen PJ, Hou J, McCrae MA, Chen X, Zhuang H, Lu F. Re-evaluation of the carcinogenic significance of hepatitis B
virus integration in hepatocarcinogenesis. PLoS One. 2012; 7(9):e40363.
10.
Breitbach CJ, Burke J, Jonker D, Stephenson J, Haas AR, Chow LQ, Nieva J, Hwang TH, Moon A, Patt R, Pelusio A, Le Boeuf F, Burns J, Evgin L, De Silva N, Cvancic
S, Robertson T, Je JE, Lee YS, Parato K, Diallo JS, Fenster A, Daneshmand M, Bell JC, Kirn DH. Intravenous delivery of a multi-mechanistic cancer-targeted oncolytic
poxvirus in humans. Nature. 2011 Aug 31; 477(7362):99-102.
11.
Tao Y, Ruan J, Yeh SH, Lu X, Wang Y, Zhai W, Cai J, Ling S, Gong Q, Chong Z, Qu Z, Li Q, Liu J, Yang J, Zheng C, Zeng C, Wang HY, Zhang J, Wang SH, Hao L, Dong
L, Li W, Sun M, Zou W, Yu C, Li C, Liu G, Jiang L, Xu J, Huang H, Li C, Mi S, Zhang B, Chen B, Zhao W, Hu S, Zhuang SM, Shen Y, Shi S, Brown C, White KP, Chen
DS, Chen PJ, Wu CI. Rapid growth of a hepatocellular carcinoma and the driving mutations revealed by cell-population genetic analysis of whole-exome data. Proc
Natl Acad Sci USA. 2011 Jul 19; 108(29):12042-7.
12.
Diallo JS, Roy D, Abdelbary H, De Silva N, Bell JC. Ex vivo infection of live tissue with oncolytic viruses. J Vis Exp. 2011 Jun 25; (52). doi;pii:2854.
13.
Heo J, Breitbach CJ, Moon A, Kim CW, Patt R, Kim MK, Lee YK, Oh SY, Woo HY, Parato K, Rintoul J, Falls T, Hickman T, Rhee BG, Bell JC, Kirn DH, Hwang TH.
Sequential therapy with JX-594, a targeted oncolytic poxvirus, followed by sorafenib in hepatocellular carcinoma: preclinical and clinical demonstration of
combination efficacy. Mol Ther. 2011 Jun; 19(6):1170-9.
47
Development of 2-[18F]fluoro-2-deoxy-D-galactose as a new molecular imaging probe for
hepatocellular carcinoma diagnosis
NSC-TFRI International Collaborative Research (2013-2016)
Investigator: François Benard, BC Cancer Agency
Scientific Summary: Liver cancer is a major cause of death among patients of east or southeast asian descent, as well
as other population groups, notably in central and west Africa. Diagnosis of liver cancer requires a combination of
several imaging techniques and biopsies. Despite this, diagnosis can remain inconclusive or difficult to establish in
patients at risk for liver cancer.
The purpose of this joint Taiwanese / Canadian research project is to evaluate novel imaging methods developed to
diagnose the most common form of liver cancer, hepatocellular carcinoma. We propose to use novel imaging probes
that have been reported to bind to liver cancers but not benign liver lesions that can be confused with liver cancer.
Three such imaging probes will be evaluated. 2-[ 18F]-fluoro-2-deoxy-D-gulcose, called [ 18F]FDG, is a radioactive sugar
that is widely used for cancer imaging with a device called positron emission tomography, or PET scans. We already
know that [ 18F]FDG cannot detect some liver cancers that are slow growing. 2-[ 18F]Fluoro-2-deoxy-Dgalactose([ 18F]FDGal), another radioactive sugar, has been recently reported to be highly effective at detecting liver
cancer. [ 18F]Fluorocholine ([ 18F]FCH), another molecule, is currently being evaluated in Taiwan and other jurisdictions
for this purpose. In 2010, a French researcher reported 80-90% detection rate by using [ 18F]FCH alone or in
combination with [ 18F]FDG. In 2011, a Danish researcher reported an even better result by using [ 18F]FDGal alone.
The Taiwan group will compare [ 18F]FCH and [ 18F]FDGal. The Canadian (Vancouver) group will compare [ 18F]FDGal and
[ 18F]FDG, which could be complementary to each other. Both groups will evaluate 50 patients each over a period of
three years. The results will be correlated with those of biopsies and clinical follow-up. Having two patient groups will
allow the researchers to compare two strategies, while minimizing the number of diagnostic tests that research
participants will have to undergo to evaluate the best diagnostic strategy. After the completion of these two trials, we
will compare the results with another on going multi-centre trial now already on schedule in Taiwan by 10 medical
centres using [ 18F]FCH vs. [ 18F]FDG.
This study will provide valuable data on whether these imaging agents can successfully differentiate malignant liver
lesions from benign ones. It will also provide information about whether these imaging agents can successfully assess
whether the cancer has spread outside the liver. It will provide data that will allow physicians to determine the optimal
imaging protocol to properly diagnose liver cancer.
48
Modeling and therapeutic targeting of the clinical and genetic diversity of glioblastoma
(2012-2017)
Investigators: Gregory Cairncross, Stephen Robbins, Samuel Weiss, University of Calgary; David Kaplan, UofT; Warren
Mason, Queen's University; Marco Marra, UBC
Scientific Summary: Glioblastoma (GBM) is a deadly brain cancer that has eluded major treatment advances. While all
agree that new therapies for GBM are needed, there is no consensus on how best to find them. With this project, we
employ a unique collection of cell lines established from GBM. These lines, referred to as brain tumour initiating cells
(BTICs), capture and retain the major genetic alterations that are present in the tumours from which they were
derived, in addition to maintaining many of the histological features of the parent tumour when grown in vivo.
This cell-based model system now provides our team at the universities of British Columbia, Calgary and Toronto with
the foundation for an innovative drug discovery and genome-sequencing program with real potential for rapid clinical
translation. Our experimental strategy begins with BTIC lines as a research tool for drug and target discovery and ends
with new therapeutics in early phase human testing in molecularly defined subpopulations of GBM, via a collaboration
with the NCIC Clinical Trials Group and its many participating Canadian centres. Our approach, which combines a
superior model system with high-throughput drug screening and genomics technologies, holds great promise.
Our singular objective is the discovery of new drug therapies for GBM within five years that will improve tumour
control and quality of life for patients with this disease. This project will also ensure that specialized laboratory models
of GBM are in hand to support future drug discovery.
Specific Aims:
 High-throughput screening of toolkit, NIH, and kinase inhibitor libraries against a panel of BTICs to enable rapid
identification of targeted drug therapies for GBM.
 Genome and transcriptome sequencing of BTICs, their parent tumours, and normal DNA to enable the discovery of
new drug targets, as well as correlate genotype with drug response.
 Continued establishment of BTIC lines from common and rare types of glioma.
 Pre-clinical testing of promising compounds in vivo.
 Clinical trials of promising compounds.
List of Key Publications:
Davis B, Shen Y, Poon CC, Luchman H, Stechishin OD1, Pontifex CS, Wu W, Kelly JJ, Blough MD; Terry Fox Research Institute
Glioblastoma Consortium. Comparative genomic and genetic analysis of glioblastoma-derived brain tumor-initiating cells and their
parent tumors. Neuro Oncol. 2015 Aug 5. pii: nov143. [Epub ahead of print]
Luchman HA, Stechishin OD, Nguyen SA, Lun XQ, Cairncross JG, Weiss S. Dual mTORC1/2 blockade inhibits glioblastoma brain tumor
initiating cells in vitro and in vivo and synergizes with temozolomide to increase orthotopic xenograft survival. Clin Cancer Res. 2014
Nov 15;20(22):5756-67.
Cusulin C, Chesnelong C, Bose P, Bilenky M, Kopciuk K, Chan JA, Cairncross JG, Jones SJ, Marra MA, Luchman HA, Weiss S7. Precursor
States of Brain Tumor Initiating Cell Lines Are Predictive of Survival in Xenografts and Associated with Glioblastoma Subtypes. Stem
Cell Reports. 2015 Jul 14;5(1):1-9. doi: 10.1016/j.stemcr.2015.05.010. Epub 2015 Jun 18.
49
Investigation of the pathogenesis of ASXL1 mutation in acute myeloid leukemia
NSC-TFRI International Collaborative Research (2013-2016)
Investigators: Keith Humphries, Aly Karsan, BC Cancer Agency; Hwei-Fang Tien, Yuan-Yeh Kuo, Wen-Chien Chou,
National Taiwan University College of Medicine
Scientific Summary: ASXL1 is the human homolog of Drosophila additional sex combs (Asx), which encodes a
chromatin-binding protein required for normal determination of segment identity in the developing embryo. ASXL1was
found to be mutated in AML and other myeloid malignancies. A team of researchers at the National Taiwan University
have analyzed the clinical implications of this mutation in a large cohort of their de novo AML. They found several
features of this mutation, including not correlating with a normal karyotype, frequent association with older age, male
sex, isolated trisomy 8, RUNX1 mutation, and expression of HLA-DR and CD34, but mutual exclusion with t(15;17),
complex cytogenetics, FLT3-ITD, NPM1 mutations, WT1 mutations, and expression of CD33 and CD15. Several studies
have shown that ASXL1 mutation is a poor prognostic factor. However, the pathophysiology underlying the mutation
remains largely unknown. In this collaborative project, the Taiwan and Canadian researchers propose to explore the
mechanisms of ASXL1 that would be of great value in understanding the processes of leukemogenesis and in searching
for novel therapy.
Specific Aims:
 To investigate the nature of human ASXL1 mutation in vivo. Is ASXL1 mutation a loss-of-function, gain-of-function,
or dominant-negative mutation in human AML?
 To understand how the ASXL1 mutation affect epigenetic regulation in vivo.
 To answer if ASXL1 mutation alone is sufficient for leukemogenesis.
 To explore the co-operation between mutations of ASXL1 and other genes such as RUNX1.
 To search for any novel therapy.
 Canadian investigators Drs. Humphries and Karsan will work closely with the Taiwan collaborators to analyze the
miRNA/mRNA expression profiles, characterize the ASXL1 mutant “knock-in” mouse model, validate the findings in
mutations from the discovery cohort, and provide expertise, training, and re-agents for retroviral/lentiviral gene
transfer (shRNA and mRNA) to identify collaborating genes with mutant ASXL1 that would accelerate
leukemogenesis or suppress leukemogenesis.
List of Key Publications:
1.
Chou WC, Huang HH, Hou HA, Chen CY, Tang JL, Yao M, Tsay W, Ko BS, Wu SJ, Huang SY, Hsu SC, Chen YC, Huang YN, Chang
YC, Lee FY, Liu MC, Liu CW, Tseng MH, Huang CF, Tien HF. (2010) Distinct clinical and biological features of de novo acute
myeloid leukemia with additional sex comb-like 1 (ASXL1) mutations. Blood 116: 4086-4094.
2.
Kuchenbauer F, Mah SM, Heuser M, McPherson A, Rüschmann J, Rouhi A, Berg T, Bullinger L, Argiropoulos B, Morin RD, Lai D,
Starczynowski DT, Karsan A, Eaves CJ, Watahiki A, Wang Y, Aparicio SA, Ganser A, Krauter J, Döhner H, Döhner K, Marra MA,
Camargo FD, Palmqvist L, Buske C, Humphries RK. (2011) Comprehensive analysis of mammalian miRNA* species and their role
in myeloid cells. Blood 118: 3350-8.
3.
Starczynowski DT, Morin R, McPherson A, Lam J, Chari R, Wegrzyn J, Kuchenbauer F, Hirst M, Tohyama K, Humphries RK,
Lam WL, Marra M, Karsan A. (2011) Genome-wide identification of human microRNAs located in leukemia-associated genomic
alterations. Blood 117: 595-607.
4.
Starczynowski DT, Kuchenbauer F, Wegrzyn J, Rouhi A, Petriv O, Hansen CL, Humphries RK, Karsan A.(2011) MicroRNA-146a
disrupts hematopoietic differentiation and survival. Exp Hematol 39: 167-178.
5.
Hou HA, Lin CC, Chou WC, Liu CY, Chen CY, Tang JL, Lai YJ, Tseng MH, Huang CF, Chiang YC, Lee FY, Kuo YY, Lee MC, Liu
MC, Liu CW, Lin LI, Yao M, Huang SY, Ko BS, Hsu SC, Wu SJ, Tsay W, Chen YC, Tien HF. (2013) Integration of cytogenetic and
molecular alterations in risk stratification of 318 patients with de novo non-M3 acute myeloid leukemia. Leukemia (Epub ahead of
print)
50
Pan-Canadian early lung cancer detection study (2008-2015)
Investigators: Stephen Lam, Ming Tsao; British Columbia Cancer Agency, University Hospital Network-Princess
Margaret Hospital (Co-Directors)
BCCA-VCH (Annette McWilliams, John Mayo, Richard Finley, John Yee, Ken Evans, Paola Nasute)
University of Calgary (Alain Tremblay, Paul Burrowes, Paul MacEachern)
University Hospital Network-Princess Margaret Hospital (Heidi Roberts, Geoff Liu, Frances Shepherd, Kam Soghrati,
Kazurhiro Yasufuku, John Thenganat, Charlie Chan, Natasha Leighl)
Juranvinski Cancer Centre (John Goffin, Serge Puksa, Lori Stewart, Allan McLellan, Bill Evans)
Ottawa Hospital Regional Cancer Centre (Garth Nicholas, Glen Goss, Jean M Seely, Kayvan Amjadi)
University of Laval (Simon Martel, Francis Laberge, Michel Gingras, Christian Couture)
Dalhousie University (Michael Johnson, Daria Manos)
Memorial University (Rick Bhatia)
Lung Cancer Risk Modeling (Martin Tammemagi, Don Sin, Geoff Liu)
Health Economics & QOL (Stuart Peacock, Bill Evans, Martin Tammemagi, Natasha Leigh, Sonya Cressman)
Blood Biomarkers (Geoffrey Liu, Don Sin)
COLD Network (Lung Function) (Wan Tan)
Quality Assurance (Nestor Muller (Radiology), Tom Sutedja (Bronchoscopy), Adi Gazdar (Pathology)
Scientific Advisory Committee (Christine Berg, John Field, James Jett)
Funding Partners: The Canadian Partnership Against Cancer, Lung Cancer Canada, Princess Margaret Cancer Centre
Foundation, BC Cancer Foundation
Scientific Summary: Sophisticated but relatively expensive technologies such as low dose spiral computed tomography
(CT) and autofluorescence Bronchoscopy (AFB) exist for detection of early lung cancer. The inclusion of low cost risk
modeling and biomarkers to select population cohorts with the highest risk of lung cancer development may provide a
cost effective application of relatively expensive, yet effective, detection methods. Our objective is to develop a new
multi-modal early detection strategy that integrates risk modeling, spirometry, AFB and blood biomarkers with CT for
early detection of lung cancer.
The study has a 6.4% cancer detection rate. Seventy-six percent of the cancers were detected from abnormalities
observed at the baseline scan and 24% were incidence cancers. CT scan data from the study was used as a
development data set to determine the malignancy of lung nodules on first screen CT. The study has produced a highly
predictive tool based on patient and nodule characteristics to accurately estimate the probability that lung nodules
detected on baseline screening CT are malignant. The results were published in the N Engl J Med 2013;369:910-919.
The study aims to complete a third round of screening on enrolled participants to contribute additional information
regarding the frequency and duration of LDCT screening.
Pending Manuscripts:
1.
2.
3.
4.
Low prevalence of high grade lesions detected with autofluorescence bronchoscopy in the setting of lung cancer screening in
the pan-Canadian Lung cancer screening study. Alain Tremblay, et.al. Abstract published and presented at 2015 Chest
Conference. Pending journal submission.
Computer vision Tool and Technician as first reader of lung cancer screening CT. Alexander Ritchie, et.al. Pending journal
submission.
Pan-Canadian Early Detection of Lung Cancer Study Design and Selected Findings (baseline paper). Martin Tammemagi.
Draft manuscript Oct 2015.
Predication of lung cancer in abnormal computed tomography screens in the national lung screening trial and pan-Canadian
early detection of lung cancer study. Martin Tammemagi. Comment: this study will extend our existing NEJM nodule
51
6.
7.
8.
9.
prediction model and will be applicable in post-baseline screening situations. This model is expected to receive a great deal
of attention and application, as our initial model did.
Factors associated with quality of life in former and current smokers. Sociodemographic, smoking, morbidity, pulmonary
function and symptom factors and quality of life in former and current smokers. Martin Tammemagi. Planned submission
2016
Screen-detected cardiovascular disease – Impact on mortality and lung cancer screening. Martin Tammemagi.
Pulmonary symptoms and lung cancer risk in smokers at high risk for lung cancer. Martin Tammemagi. Planned submission
2016.
List of Key Publications and Abstracts:
1.
2.
3.
4.
5.
6.
7.
8.
9.
10.
11.
12.
13.
14.
15.
16.
Tremblay, Alain. Low prevalence of high grade lesions detected with autofluorescence bronchoscopy in the setting of lung
cancer screening in the pan-Canadian Lung cancer screening study. Abstract published and presented at Oct 2015 Chest
Conference.
Ritchie, Alex. Computer vision tool and technician as first reader of lung cancer screening CT. IASLC Mini 36.05 Denver CO,
Sept 2015
Ritchie, Alex. Automated measurement of malignancy risk of lung nodule detected by screening computed tomography.
IASLC Mini 36.04 Denver CO, Sept 2015
Lam,Stephen. Clinical Trials: who are the proper cohorts and how do you recruit subjects? IASLC Mini 14.02 Denver CO,
Sept 2015
Cressman, Sonya. Economic Evidence for the use of risk selection and risk-stratification for lung cancer screening programs.
IASLC Oral 09.07 Denver CO, Sept 2015
Tammemagi M, Lam S, McWilliams A, Sin D. Incremental value of pulmonary function and sputum DNA image cytometry in
lung cancer risk prediction. Cancer Prev Res (Phila). 2011 Apr;4(4):552-61. Epub 2011 Mar 15.
Leung JM, Mayo J, Tan W, Tammemagi M, et.al. Plasma pro-surfactant protein B and lung function decline in smokers. Eur
Respir J 2015;45:1037-1045 doi:10.1183/09031936.00184214
Cressman S, Lam S, Tammemagi MC, Evan WK, Leighl NB, et.al. Resource utilization and costs during the initial years of
lung cancer screening with computed tomography in Canada. J Thorac Oncol 2014 Oct:9(10):1449-58. Doi:
10.1097/JTO.0000000000000283
Tammemagi MC, Lam S. State-of-the-Art Review: Current Issues in Low-dose Computed Tomography Screening for Lung
Cancer. BMJ British Medical Journal. 2014 (Clinical research ed). ePublished in advance of print 27 May 2014;348. DOI:
10.1136/bmj.g2253. PMID: 24865600
Lam S, McWilliams A, Mayo J, Tammemagi M. Computed tomography screening for lung cancer: what is a positive screen?
Ann Intern Med. 2013 Feb 19;158(4):289-90. Doi:10.7326/0003-4819-158-4-201302190-00011.
Tammemagi M, Mayo J, Lam S. Cancer in Pulmonary Nodules Detected on First Screening CT - Reply to Correspondence to
the Editor. The New England journal of medicine 2013;369:2061-2.
Probability of Cancer in Pulmonary Nodules Detected on First Screening CT. Annette McWilliams, Martin C. Tammemagi,
John Mayo, et.al. New England Journal of Medicine 2013;369:910-919 September 2013.
Pro-Surfactant Protein B as a Biomarker for Lung Cancer Prediction. Don D. Sin, Martin Tammemagi, Stephen Lam, Matt J.
Barnett, Xiaobo Duan, Anthony Tam, Heidi Auman, Ziding Feng, Gary Goodman, Samir Hanash, Ayumu Taguchi for the
Pan-Canadian Early Lung Cancer Study group. J Clin Oncol 2013; 31: 4536-4543
15th World Conference of Lung Cancer, Sidney, Australia. Resource Utilization and cost screening high risk individuals for
lung cancer in Canada. (Oral Presentation) October 2013. Sonya Cressman
15th World Conference of Lung Cacner, Sidney, Australia. Lung Density versus emphysema as predictor of malignancy risk of
pulmonary nodules detected on first screening CT. (Oral Presentation) October 2013. Keishi Ohtani
15th World Conference of Lung Cancer, Sidney, Australia. Factors Associated with smoking cessation in Participants of the
Pan Canadian Early lung cancer study. (Oral Presentation) October 2013. Geoffrey Liu.
52
Translational research in lung cancer: From molecular markers/targets to therapeutic
applications
NSC-TFRI International Collaborative Research (2013-2016)
Investigators: Stephen Lam, Wan Lam, BC Cancer Agency; Pan-Chyr Yang, Chong-Jen Yu, National Taiwan University
Medical College and Hospital
Scientific Summary: Lung cancer is the leading cause of cancer mortality worldwide as well as in Taiwan. Delayed
diagnosis, early metastasis, poor treatment outcome and rapid emergence of drug resistance are the present obstacles
for the management of lung cancer patients. Dr. Martin Tammemagi has developed a lung cancer risk prediction model
in never smokers using the PLCO dataset (from the NCI sponsored Prostate, Lung, Colorectal, and Ovarian Cancer
Screening Trial) (PMID: 21606442). Drs. Tammemagi and Stephen Lam demonstrated that pulmonary function and
sputum DNA image cytometry added value to their lung cancer risk prediction model (PMID: 21411501). Dr. Lam’s
team validated this prediction model in a pan-Canadian lung cancer biomarker screening trial, which was supported by
TFRI (PMID: 24004118). The utility of this prediction tool can be tested in never and ever smoker populations in
Taiwan. Furthermore, the incremental value of the inclusion of specific genetic biomarkers can be investigated. We
hypothesize that the pan-Canadian risk model is applicable for predicting lung cancer risk in the Taiwan population.
The goal is to validate our PLCO model (validated in a TFRI sponsored pan-Canadian trial) in a Chinese population and
determine if genetic markers add incremental value to risk assessment in never and ever Chinese and Caucasian
population.
Specific Aims:
 Evaluate the utility of the Tammemagi PLCO model risk model in the Taiwanese population.
 Optimize such a model for early detection of lung cancer in Taiwan.
 Test the incremental value of genetic markers to predict lung cancer risk versus the Tammemagi PLCO model that
includes family history and lung function (FEV1%).
Impact: Early detection is critical to the reduction of lung cancer mortality. The development of a risk model optimized
for the Taiwan population will have the potential for improving clinical practice. Furthermore, such a risk model can be
adapted for other populations in Asia and worldwide.
53
A pan-Canadian platform for the development of biomarker-driven subtype specific
management of ovarian carcinoma (2010-2018)
Investigators: Anne-Marie Mes-Masson, Diane Provencher, Kurosh Rahimi, Francis Rodier, John Stagg, CHUM Research
Centre; David Huntsman, Aline Talhoukr, Anna Tinker, Dianne Miller, Blake Gilks, Brad Nelson, Peter Watson, BC Cancer
Agency, VGH; Martin Koebel, Helen Steed; Alberta Health Services; Hal Hirte, Hamilton Health Sciences Centre; Ted
Brown, Joan Murphy, Barry Rosen, Helen Mackay, Marcus Bernadini, Patricia Shaw, Blaise Clarke, Amit Oza, UHN;
Trevor Shepherd, London Health Sciences Centre; Janet Dancey, Barbara Vanderhyden, Johanne Weberpals, Micheal
Fungkee Fung, Ottawa Regional Cancer Center; Walter Gotlieb, Jewish General Hospital, Montreal; Mark Natchigal,
University of Manitoba; Patricia Tonin, McGill University; Alain Piché, University of Sherbrooke; Isabelle Bairati; Dimcho
Bachvarov, Marie Plante, Bernard Tetu, CHUQ, Université Laval; Robin Uqhart, Dalhousie University; Eva Grunfeld,
OICR
Scientific Summary: Ovarian cancer is the second most common gynecological cancer and the leading cause of death
from gynecological malignancies. Early detection of ovarian cancer is rare and little is known about the natural history
of disease progression. More recently, the notion that ovarian cancer is a single disease has given way to a more
sophisticated concept of subsets of ovarian disease that may be associated with very different molecular events. While
platinum/taxane-based treatment is currently the gold standard in first-line therapy, failure of this treatment in a
significant portion of patients remains a serious problem. Identifying non-responders, and offering these individual
alternative first-line treatments remains one of the most important aspects in the initial clinical management of the
ovarian cancer patient, and is the focus of future clinical trials.
In order to address these issues, the research team proposes the following specific aims:
 A validated classification system for ovarian cancer that stratifies cases into groups with different natural histories
and chemotherapy response rates, and a trained pathology community ready to effectively perform this
classification with QA program to maintain excellence.
 A cohort of over 2,000 ovarian cancer sub-typed cases (COEUR) to be interrogated for biomarkers predictive of
treatment response, and analysis would be extended to address response in sub-type specific ovarian disease.
 The biomarkers with the highest predictive value will be carried forward for correlative studies in sub-type specific
ovarian cancer trials.
 A pan-Canadian team of clinical researchers working within a collaborative framework to reduce ovarian cancer
mortality.
List of Key Publications:
1.
2.
3.
Lee S. et al. Calibration and optimization of p53, WT1, and Napsin A immunohistochemistry ancillary tests for histotyping of
ovarian carcinoma: Canadian Immunohistochemistry Quality Control (CIQC) experience. Int J Gunecol. Pathol. (2015).
Le Page, C. et al.: Report on a quality control exercise performed on specimens from Canadian biobanks participating in the
COEUR specimen repository. Biopreservation and Biobanking (2013) 11(2), 83:93.
4.
Le Page, C. et al: Predictive & Prognostic Protein Biomarkers in Epithelial Ovarian Cancer –Recommendations for Future Studies:
Cancer (2010) 2, 913-954.
5.
Köbel M. et al.: The biological and clinical value of p53 expression in pelvic high-grade serous carcinomas. J Pathol. (2010)
222(2):191-8.
54
Selective Therapies Program Collaboration: Therapeutic targets validation in ovarian cancer
(2012-2015)
Investigators: Anne-Marie Mes-Masson (Institut du cancer de Montréal/CRCHUM, Medicine Dept., Université de
Montréal, Montreal - PI), Robert Rottapel (Ontario Institute for Cancer Research, Dept. of Medical Biophysics, University
of Toronto Toronto - PI), Diane Provencher (Dept. Ob/Gyn, University of Montreal, Montreal - PI) and Laudine Communal
(Institut du cancer de Montréal/CRCHUM, Montreal - Project Manager)
Collaborators : Mauricio Medrano (Ontario Institute for Cancer Research, Toronto), Fabrice Sircoulomb (Ontario Institute
for Cancer Research, Toronto).
Scientific Summary: Ovarian cancer is the most lethal of gynecological cancer and effective therapies are still lacking as
the majority of patients develop resistance to first-line chemotherapy. In order to improve cancer outcomes, the
Selective Therapy Program (STP) was launched by the Terry Fox Research Institute (TFRI)/Ontario Institute for Cancer
Research (OICR). This program has identified new promising therapeutic targets using an integrative genomic, proteomic
and functional approach1. Here we characterized and validated the relevance of selected candidates as therapeutic
targets or biomarkers in High Grade Serous Epithelial Ovarian Cancer (HGS-EOC).
A systematic candidate characterization includes a first step of expression evaluation by western blot in EOC cell lines2.
Candidate expression is then evaluated by immunofluorescence (IF) on a tissue-microarray (TMA) consisting of 101 cases
of HGS-EOC. Multi-labeling IF conditions were defined to allow the discrimination of epithelial and stromal cells as well
as nuclei and cytoplasmic compartments3. Candidate expression levels are accurately quantified in relevant
compartments with a powerful image analysis procedure (VisiomorphTM) and are correlated with patient clinical
parameters in order to determine their relevance and to prioritize them for further studies.
Promising results were obtained for three candidates so far. High expression of CD151 and APBB3 protein were
correlated with poor patient prognosis in our HGS-EOC TMA and in the pan-Canadian TFRI COEUR cohort comprised of
983 HGS tumours. Further analyses showed that CD151 depletion impaired survival, proliferation and tumor growth in
murine models of a subset of HGS-EOC cell lines. In addition, low JAMA protein expression was correlated with poor
prognosis in the 101 cases HGS-EOC TMA. Further analyses are ongoing to confirm the potential of CD151, APBB3 and
JAMA as biomarkers and/or therapeutic targets.
All the candidates showed a good expression in HGS-EOC suggesting that they can be further studied as therapeutic
targets. In addition, some of the candidates seem promising as prognostic markers. Systematic review of all candidates
will reveal those best suited to be further studied as therapeutic targets or prognostic markers.
List of Key Publications:
1. Marcotte R, Brown KR, et al.: Essential gene profiles in breast, pancreatic, and ovarian cancer cells. Cancer discovery 2012, 2(2):172189.
2. Fleury H*, Communal L*, et al.: Novel high-grade serous epithelial ovarian cancer cell lines that reflect the molecular diversity of both
the sporadic and hereditary disease. Genes & Cancer 2015. Accepted. *These co-authors have contributed equally.
3. Labouba I, Le Page C, Communal L, et al.: Potential Cross-Talk between Alternative and Classical NF-κB Pathways in Prostate Cancer
Tissues as Measured by a Multi-Staining Immunofluorescence Co-Localization Assay. PLoS One 2015, 10(7):e0131024.
1. Medrano M, Communal L, et al: Interrogation of functional cell surface markers identifies CD151 dependency in high-grade serous
ovarian cancer. In preparation.
55
Efficacy of optically guided surgery in the management of early-stage oral cancer: The
Canadian optically guided approach for oral lesions surgical (COOLS) trial (2010 -2016)
Investigators: BCCA/BCCRC/UBC: Catherine Poh, Scott Durham, Calum MacAulay; BCCA/BCCRC/SFU: Miriam Rosin,
Stuart Peacock, Kitty Corbett; University of Calgary: Joseph Dort; University of Alberta: Hadi Seikaly, University of
Manitoba: Paul Kerr; Sunnybrook Health Sciences Centre: Kevin Higgins; London Health Sciences Centre: John Yoo;
Dalhousie University: Robert Hart.
Scientific Summary: The COOLS trial is a multicentre Phase III randomized control trial that is evaluating the clinical
efficacy of an optical tool to reduce local recurrence of oral cancers and severe dysplasia. The tool identifies alteration
of tissue autofluorescence (FV) around oral lesions and uses such change to delineate surgical margins. The study will
recruit a total of 400 patients with oral severe dysplasia or higher. Patients will be randomized into either FV-guided
(experimental arm) or white light (current standard of care) surgery.
The trial has four goals: 1) To collect clinical evidence of the comparative effectiveness of the two treatments. 2) To
collect molecular and phenotypic evidence in margins to test if FV produces a shift in surgical field, sparing normal
tissue while catching high-risk occult tissue. 3) To collect relative cost-effective evidence of the two treatments in both
the cost per avoided recurrence and the cost per quality-adjusted life years (QALYs) gained. 4) To develop a knowledge
translation (KT) strategy that will foster the dissemination of FV-guided surgery across Canada and globally.
As of September 30, 2015, we have seven sites actively engaging in steady patient follow-up: Our final accrual
number stands at 427 patients (246 cancerous and 181 high-grade lesions). All of them are being actively followed up
with 84% of the projected visits completed at the end of this reporting period. To date, there have been 29 cases of
local recurrence, the primary endpoint during the follow-ups.
On June 8th 2015, we held the 5th COOLS annual meeting in Winnipeg, with all the site co-ordinators and the
participating site surgeons in attendance. We committed to continue collaborating with the PanCanNOCC network with
the consensus that this historical teamwork provides a valuable infrastructure for knowledge sharing, improves patient
care and clinical results, and pushes forward Canadian oral cancer disease prevention and treatment.
Continuation of sample pipeline and molecular analysis for margin samples: We have developed a pipeline for sample
flow from acquisition to documentation, histological review, and sample selection for analysis, processing and delivery
to laboratories for molecular and phenotypic analysis. Preliminary quantitative tissue pathology (QTP) and loss of
heterozygosity (LOH) analysis shows positive correlations of these endpoints with margin evaluation using histology
and FV status. Early data suggest that we may be able to spare low-risk tissue at surgical margins.
Health Economics Team: The Health Economics Team (Goal 3) has been developing its analysis plan using interim data
from the COOLS Trial. Unit costs for time and equipment have been applied to surgical capture forms and subpopulation analyses have been applied to investigate differences in clinical populations. These methods and some
interim findings have been submitted and presented at national and international conferences, with feedback from
health economics experts. A qualitative “face-to-face interview” component has been appended to this goal, and has
received provisional approval from the University of British Columbia Research Ethics Board – interviews began in fall
2013. Interim data analysis will continue as the trial increases its recruitment and follow-up.
Knowledge Translation: Knowledge translation (KT) in the COOLS trial entails health service data collection to prepare
for dissemination and scale-up of fluorescence visualization (FV)-guided surgery beyond the trial, if warranted by study
results. KT discovery and application throughout the trial is informed by Social Marketing and Diffusion of Innovations
change theories. The discovery phase, which has been completed, involved data collection from study site surgeons,
pathologists, clinic staff, and patients that explored factors and processes of FV-guided surgery important for clinical
practice change. Future application-phase activities will include developing and testing of an acceptable, appropriate
56
KT scale-up strategy. Both phases use a between-case, compare and contrast approach concurrent with qualitative
data collection to identify emerging themes and inform subsequent steps.
List of Key Publications:
1.
Poh, C.F., Anderson, D.W., Durham, J.S., Chen, J., Berea, K.W., MacAulay, C.E., Rosin, M.P. A novel optically-guided surgical
approach improves local recurrence of early-stage oral cancer. (Accepted to JAMA – Otolaryngology Head and Neck Surgery;
September 18, 2015)
2.
Lee AM, Cahill L, Liu K, MacAulay C, Poh C, Lane P. Wide-field in vivo oral OCT imaging. Biomedical optics express.
2015;6(7):2664-74.
3.
Towle R, Tsui IF, Zhu Y, MacLellan S, Poh CF, Garnis C. Recurring DNA copy number gain at chromosome 9p13 plays a role in
the activation of multiple candidate oncogenes in progressing oral premalignant lesions. Cancer Med. 2014;3(5):1170-84.
4.
Towle R, Gorenchtein M, Dickman C, Zhu Y, Poh C, Garnis C. Dysregulation of microRNAs Across Oral Squamous Cell
Carcinoma Fields in Non-smokers. Journal of Interdisciplinary Medicine and Dental Science. 2014;2(4):9.
5.
Laronde DM, Williams PM, Hislop TG, Poh C, Ng S, Zhang L, Rosin MP. Decision making on detection and triage of oral mucosa
lesions in community dental practices: screening decisions and referral. Community dentistry and oral epidemiology.
2014;42(4):375-84.
6.
Laronde DM, Williams PM, Hislop TG, Poh C, Ng S, Bajdik C, Zhang L, MacAulay C, Rosin MP. Influence of fluorescence on
screening decisions for oral mucosal lesions in community dental practices. J Oral Pathol Med. 2014;43(1):7-13.
7.
Towle R, Truong D, Hogg K, Robinson WP, Poh CF, Garnis C. Global analysis of DNA methylation changes during progression of
oral cancer. Oral Oncol. 2013;49(11):1033-42.
8.
El Hallani S, Poh CF, Macaulay CE, Follen M, Guillaud M, Lane P. Ex vivo confocal imaging with contrast agents for the detection
of oral potentially malignant lesions. Oral Oncol. 2013;49(6):582-90.
9.
Contaldo M, Poh CF, Guillaud M, Lucchese A, Rullo R, Lam S, Serpico R, MacAulay CE, Lane PM. Oral mucosa optical biopsy by
a novel handheld fluorescent confocal microscope specifically developed: technologic improvements and future prospects. Oral
Surg Oral Med Oral Pathol Oral Radiol. 2013;116(6):752-8.
10.
Zhang LW, Poh CF, Williams M, Laronde DM, Berean K, Gardner PJ, Jiang HJ, Wu L, Lee JJ, Rosin MP. Loss of Heterozygosity
(LOH) Profiles-Validated Risk Predictors for Progression to Oral Cancer. Cancer Prev Res. 2012;5(9):1081-9.
11.
Poh CF, Zhu Y, Chen E, Berean KW, Wu L, Zhang L, Rosin MP. Unique FISH Patterns Associated with Cancer Progression of
Oral Dysplasia. J Dent Res. 2012;91(1):52-7.
12.
Maclellan SA, Lawson J, Baik J, Guillaud M, Poh CF, Garnis C. Differential expression of miRNAs in the serum of patients with
high-risk oral lesions. Cancer Med. 2012;1(2):268-74.
13.
MacAulay C, Poh CF, Guillaud M, Williams PM, Laronde DM, Zhang LW, Rosin MP. High throughput image cytometry for
detection of suspicious lesions in the oral cavity. Journal of Biomedical Optics. 2012;17(8).
14.
Gorenchtein M, Poh CF, Saini R, Garnis C. MicroRNAs in an oral cancer context - from basic biology to clinical utility. Journal of
dental research. 2012;91(5):440-6.
15.
Poh CF, MacAulay CE, Laronde DM, Williams PM, Zhang LW, Rosin MP. Squamous cell carcinoma and precursor lesions:
diagnosis and screening in a technical era. Periodontology 2000. 2011;57:73-88.
16.
Poh CF, Durham JS, Brasher PM, Anderson DW, Berean KW, MacAulay CE, Lee JJ, Rosin MP. Canadian Optically-guided
approach for Oral Lesions Surgical (COOLS) trial: study protocol for a randomized controlled trial. BMC Cancer. 2011;11:462.
17.
Lane P, Poh CF, Durham JS, Zhang LW, Lam SF, Rosin M, MacAulay C. Fluorescence-guided surgical resection of oral cancer
reduces recurrence. Proc Spie. 2011;7883.
18.
Tsui IFL, Poh CF, Garnis C, Rosin MP, Zhang LW, Lam WL. Multiple pathways in the FGF signaling network are frequently
deregulated by gene amplification in oral dysplasias. International Journal of Cancer. 2009;125(9):2219-28.
19.
Tsui IFL, Garnis C, Poh CF. A Dynamic Oral Cancer Field Unraveling the Underlying Biology and Its Clinical Implication.
American Journal of Surgical Pathology. 2009;33(11):1732-8.
20.
Poh CF, MacAulay CE, Zhang LW, Rosin MP. Tracing the "At-Risk" Oral Mucosa Field with Autofluorescence: Steps Toward
Clinical Impact. Cancer Prev Res. 2009;2(5):401-4.
57
The Canadian Prostate Cancer Biomarker Network (CPCBN) (2010-2016)
Investigators: Fred Saad, Anne-Marie Mes-Masson, Mathieu Latour, Pierre Karakiewicz, Jean-Baptiste Lattouf, LouisMathieu Stevens, Mathieu Latour, Dominique Trudel, John Stagg, CHUM; Marie-Paule Jammal, Cité de la Santé de Laval;
Jean-Benoît Paradis, Complexe Hospitalier de la Sagamie; Armen Aprikian, Simone Chevalier, Simon Tanguay, Jacques
Lapointe, Fadi Brimo, McGill University Health Center; Louis Lacombe, Alain Bergeron, Yves Fradet, Hélène Larue, CHUQ;
Neil Fleshner, Rob Bristow, Theodorus van der Kwast, Antonio Finelli, Shabbir Alibhai, Natasha Leigh, UHN; Laurence
Klotz, Margaret Fitch, Sunnybrook Hospital; Darell Drachenberg, Manitoba Prostate Center; Martin Gleave, Ladan Fazli,
Alan So, Colin Collins, VPC; Simon Sutcliffe, BCCA.
Scientific Summary: Prostate cancer is the most commonly diagnosed cancer with an estimated 23,600 new cases in
2013 and it is the third leading cause of cancer-related death in Canadian men. The introduction in the 1990s of prostate
specific antigen (PSA) as a screening tool greatly facilitated the diagnosis of prostate cancer and in particular favoured
the detection of early stage, and, in some, cases low-grade (Gleason 6 or less) tumours. In patients with low- grade
tumours, it is presently difficult to differentiate between low- and high-risk disease, which contributes to the
overtreatment of men for whom interventional therapy is neither required, nor appropriate, to ensure a lifespan
uncompromised by cancer or its therapeutic consequences. Therefore, there is an urgent need for new prognostic tools
that will allow the distinction between low-grade tumours requiring definitive therapy and those that are best suited for
observation. When patients are under active surveillance (AS), practitioners routinely measure PSA levels and monitor
signs of disease progression through regular biopsies and digital rectal exams. This delays curative treatment in low-risk
patients until there are indications that the disease is progressing, at which time active treatment is initiated. Moreover,
there is also a need to identify biomarkers that will add to the currently used clinical and pathological parameters to
identify patients at high-risk of cancer recurrence and/or progression that may benefit from adjuvant or neo-adjuvant
therapies. This would have the potential of directing high-risk patients to multi-modal therapy and/or trials with novel
therapies in order to limit their disease. Accurate and individualized risk stratification may have profound individual
(lower recurrence rates, better quality of life) and societal (lower cost, better use of health resources) implications.
To accomplish its goals, the CPCBN assembled a large tissue microarray (TMA) series of 1,508 radical prostatectomy
specimens associated with extensive clinico-pathological data. This important resource is presently being shared
among Canadian researchers to validate biomarkers related to prostate cancer patient prognosis. In addition, the
CPCBN also created a TMA series of 125 biopsy specimens from intermediate-risk patients treated by radiotherapy in
combination or not with hormonotherapy. Due to the nature of biopsy specimens, this TMA will be used only for binary
markers in immunohistochemistry. In collaboration with GenomeDx, the CPCBN moved towards an RNA/DNA approach
for its cohort of patients treated by radiotherapy. Microarray expression data GenomeDx signature category of risk and
copy number alterations from over 200 patient specimens will be available for researchers. The CPCBN will also move
towards a RNA/DNA approach for the active surveillance cohort of 250 patients. To access the CPCBN TMA series and
extracted material or profiling data, researchers must fill an application form for their proposal to be evaluated by the
study committee.
The CPCBN is focused on the identification of biomarkers that predict risk in order to inform clinical management
decisions. Despite the fact that there are clear advantages from a health/quality of life/ health economic viewpoint to
AS, its uptake within the Canadian context has not been studied. Indeed, the extent to which it is practiced, the barriers
to its implementation, and health professional/societal views on its acceptance in the Canadian context are poorly
documented. Using database interrogation and chart review approaches in four different provinces (Quebec, Ontario,
Manitoba, British Columbia) the CPCBN monitored AS in men that underwent a biopsy in 2010 to provide evidence for
the extent of active surveillance uptake in Canada. In depth analysis will be performed to understand the root of any
significant differences that might exist between provinces/centres.
In parallel, using a focus-group approach, patients and health care providers of the same four provinces were
interrogated to identify perceived barriers and facilitators to AS. The rich data collected during the focus-group approach
will be used to inform questionnaires that can provide a quantitative measure of the importance of different barriers
and facilitators to AS from the patient and practitioner point of view.
58
Ultimately, the CPCBN aims to reduce the impact of prostate cancer by incorporating key molecular information about
expression, prognosis, response and outcome into algorithms defining optimized, individualized therapy.
The program is also defining how best to transfer this new knowledge within the Canadian health care setting. In
particular, this approach has the potential to stratify patients with low-risk disease, as determined by current criteria,
into a larger group for whom no further therapy is required to achieve survival unimpeded by prostate cancer (active
surveillance) from a small group whose disease, despite being apparently low-risk, will progress and result in premature
death if left untreated.
Specifics Aims:
Biomarker Core







Assembly of tissue micro-array- (TMA) based validation platforms:
1,500 radical prostatectomy specimens
 125 biopsy specimens from intermediate risk patients treated by radiotherapy
DNA/RNA extraction with profiling/copy number variation
o >200 biopsy specimens from intermediate risk patients treated by radiotherapy
 250 biopsy specimens from low-risk patients followed by active surveillance – profiling to be
confirmed
Validation of biomarkers:
Specific to low-risk disease that will not progress (biopsy based) to safely follow them by active surveillance and
avoid therapeutic complication
Specific to patients with a high-risk of progression/recurrence to combine their initial treatment with adjuvant
therapies.
Establishment of a nomogram to facilitate prostate cancer patient management.
Knowledge to Action Core
 Snapshot of active surveillance uptake in Canada for the year 2010.
 Identification through focus groups of the barriers in the offer, acceptance and adherence to active surveillance
in Canada.
List of Key Publications:
1.
2.
3.
4.
5.
6.
Leclerc BG, Charlebois R, Chouinard G, Allard B, Pommey S, Saad F, Stagg J. CD73 expression is an independent prognostic factor
in prostate cancer. Clin Cancer Res. 2015 Aug 7.
Trudel D, Zafarana G, Sykes J, Have CL, Bristow RG, van der Kwast T. 4FISH-IF, a four-color dual-gene FISH combined with p63
immunofluorescence to evaluate NKX3.1 and MYC status in prostate cancer. J Histochem Cytochem. 2013 Jul;61(7):500-9.
Dal Pra A1, Lalonde E, Sykes J, Warde F, Ishkanian A, Meng A, Maloff C, Srigley J, Joshua AM, Petrovics G, van der Kwast T, Evans
A, Milosevic M, Saad F, Collins C, Squire J, Lam W, Bismar TA, Boutros PC, Bristow RG. TMPRSS2-ERG status is not prognostic
following prostate cancer radiotherapy: implications for fusion status and DSB repair. Clin Cancer Res. 2013 Sep 15;19(18):5202-9
Fleshner NE, Kapusta L, Donnelly B, Tanguay S, Chin J, Hersey K, Farley A, Jansz K, Siemens DR, Trpkov K, Lacombe L, Gleave
M, Tu D, Parulekar WR.Progression from high-grade prostatic intraepithelial neoplasia to cancer: a randomized trial of combination
vitamin-E, soy, and selenium. J Clin Oncol: Jun 10;29(17):2386-90 (2011).
Klotz L, Zhang L, Lam A, Nam R, Mamedov A, Loblaw A. Clinical results of long-term follow-up of a large, active surveillance cohort
with localized prostate cancer. J Clin Oncol: 28(1):126-31 (2010).
Lessard L, Karakiewicz PI, Bellon-Gagnon P, Alam-Fahmy M, Ismail HA, Mes-Masson AM, Saad F. Nuclear localization of nuclear
factor-kappaB p65 in primary prostate tumours is highly predictive of pelvic lymph node metastases. Clin Cancer Res: 12(19):57415 (2006).
59
TFRI_portfolio_2015.indd 6
15-10-28 2:24 PM
TFRI_portfolio_2015.indd 7
15-10-28 2:24 PM
glossary
TFRI_portfolio_2015.indd 8
BCCA
BC Cancer Agency
BCCRC
BC Cancer Research Centre
CDRD
Centre for Drug Research and Development
CHUM
Centre hospitalier de l’Université de Montréal
CHUQ
Centre hospitalier de l’Université de Quebéc
CRCHUM
Centre de recherche du Centre hospitalier de l’Université de Montréal
MSGSC
Michael Smith Genome Sciences Centre
OCI
Ontario Cancer Institute
OHRI
Ottawa Hospital Research Institute
OICR
Ontario Institute of Cancer Research
PMCC
Princess Margaret Cancer Centre
SFU
Simon Fraser University
TBCC Tom Baker Cancer Centre, Calgary
UBC
University of British Columbia
UdeM
Université de Montréal
UHN
University Health Network
UofM
University of Manitoba
UofT
University of Toronto
VGH Vancouver General Hospital
VPC
Vancouver Prostate Centre
15-10-28 2:24 PM
The Terry Fox Research Institute
from coast to coast
BC
AB
SK
MB
ON
NL
QC
PEI
NB
NS
TFRI is an Institute without walls linking the capabilities
of 73 leading cancer care and cancer research institutes
and universities organized through six regional “nodes”.
NATIONAL PARTNERS
PRAIRIES
QUEBEC
Canadian Institutes of Health Research
Canadian Tumour Repository Network (CTRnet)
Genome Canada
Lung Cancer Canada
The Terry Fox Foundation
C ancerCare Manitoba
Children’s Hospital Research Institute of Manitoba
Saskatchewan Cancer Agency
Research Manitoba
University of Manitoba
University of Saskatchewan
Centre hospitalier de L’Université de Montréal
Centre hospitalier Universitaire du Québec
CHU Sainte-Justine Fondation
Institut Universitaire de Cardiologie et de
Pneumologie de Québec
Jewish General Hospital
Institut de recherche en Immunologie et
cancérologie
Fonds de recherche Québec – Santé
Fondation centre de cancérologie Charles-Bruneau
L’Institut de Recherches Cliniques de Montréal
McGill University Goodman Cancer Centre
McGill University Health Centre
McGill University
Quebec Breast Cancer Foundation
The Cole Foundation
Université de Montréal
Université Laval
Université Sherbrooke
BRITISH COLUMBIA
BC Cancer Agency
BC Cancer Foundation
The Centre for Drug Research and Development
Genome British Columbia
St. Paul’s Hospital (Providence Health)
Simon Fraser University
Team Finn Foundation
University of British Columbia
Vancouver Coastal Health Research Institute
ALBERTA
Alberta Cancer Foundation
Alberta Health Services
Alberta Innovates – Health Solutions
Cross Cancer Institute
Genome Alberta
Tom Baker Cancer Centre
University of Alberta
University of Calgary
ONTARIO
BioCanRx
Brock University
Children’s Hospital of Eastern Ontario
Hospital for Sick Children
Juravinski Cancer Centre
London Health Sciences Centre
McMaster University
Mount Sinai Hospital
Ontario Cancer Institute
Ontario Institute for Cancer Research Ottawa Hospital Research Institute
Queen’s University
Sunnybrook Research Institute
Thunder Bay Research Institute
University Health Network (Princess Margaret Cancer Centre)
University of Guelph
University of Ottawa
University of Toronto
ATLANTIC
Atlantic Cancer Research Institute (Moncton)
Capital District Health Authority
Dalhousie University (Halifax)
Isaac Walton Killam Health Centre
New Brunswick Health Research Foundation
Memorial University of Newfoundland (St John’s)
New Brunswick Cancer Network
QEII Health Sciences Centre (Halifax)
The University of New Brunswick
The University of Prince Edward Island
675 West 10th Ave / Vancouver, BC / Canada / V5Z 1L3T
604.675.8222 / [email protected] / www.tfri.ca
TFRI_portfolio_2015.indd 9
15-10-28 2:24 PM