Director Dr Gerald Batist, Professor Shirin Enger and Chief Operating Officer Miriam Santos Dutra of the McGill Centre for Translational Research in Cancer discuss the Centre’s essential role in advancing cancer research through the translation of laboratory discoveries into practical applications.
Founded in 1996, the McGill Centre for Translational Research in Cancer (MCTRC) unites a diverse team of clinical and fundamental scientists from across Quebec, focusing on advancing cancer research and improving patient outcomes. As part of both the Lady Davis Institute for Medical Research and the Segal Cancer Centre of the Jewish General Hospital (JGH), the MCTRC brings together over 60 multidisciplinary experts and leverages national and international collaborations to deliver patient-centred, cutting-edge research.
Over the years, the Centre has expanded to include numerous members dedicated to innovative cancer diagnostics, therapeutics, and translational research efforts, making significant strides in bridging the gap between laboratory discoveries and clinical applications.
The value of translational research
Fundamental research is essential for scientific inquiry, focusing on discovering new knowledge and principles behind various biological or physical phenomena. It often involves creating proof of concept to validate innovative ideas, paving the way for future cancer treatments and diagnostics. Translational research transforms these fundamental discoveries into real-world applications, rigorously validating discoveries in the clinical sphere to ensure accuracy and clinical relevance.
Dr Batist said: “We can visualise this process as an ascending spiral where clinical outcomes and real-world implementation also inform and refine fundamental research, shaping new hypotheses and guiding further scientific questions. The refined ideas are then translated back to the clinic, ensuring that scientific advancements evolve based on real-world impact and ultimately bridging the gap between the laboratory and the clinic for maximum community benefit.”
While translational research is a large domain, the MCTRC focuses downstream on the interface of these with the clinical testing and implementation while also providing feedback to discovery platforms for continuous refinement.
In addition to driving innovation, the MCTRC serves as a unique training ground for graduate and medical students. Immersed in this multidisciplinary environment, students gain invaluable experience working alongside scientists and clinicians from diverse specialities. This hands-on exposure to translational innovation cultivates the ability to collaborate across disciplines, preparing them to become the next generation of scientific and clinical innovators in oncology and beyond.
MCTRC: Our translational research infrastructure
For the past 30 years, the MCTRC has pioneered core platforms that drive the advancement of new experimental therapies. In the 1990s and early 2000s we implemented the George and Olga Minarik Research Pathology Facility, which advanced standards of practice in the handling of tissues in biopsy-driven clinical trials.
This ‘next generation’ biobanking provides tissue of metastatic tumours, where most new therapies are studied, as opposed to classical banking, where most specimens are derived from primary tumours removed at first diagnosis even before various lines of therapy likely selected important changes.
These studies helped establish standard operation procedures for tissue handling for major international studies of precision medicine based on metastatic tumour biopsies. Currently, the MCTRC hosts biobanks containing more than 5,000 tumour cases, including cancers of the breast, gynaecological area, skin, lung, gastrointestinal tract, head and neck, prostate and blood tumours.
The WINTHER study
One example of the fruit of this expertise is the WINTHER study, a biopsy-based trial run at the JGH, which exemplifies the power of collaborative, international research in advancing cancer treatment. With a focus on Precision Oncology, this initiative was run by the Worldwide Innovative Networking (WIN) Consortium, a collaboration of 39 centres across five continents, including the MCTRC. This trial performed genomic profiling of biopsies of metastatic tumours to identify targetable DNA aberrations and pioneered RNA-focused investigations.
The WINTHER study, together with several other trials, was run in the Maria Saputo Monticciolo Clinical Research Unit (CRU) located within the Segal Cancer Centre of the JGH, which ensures a seamless transition from discovery to clinical testing. It is part of a broader clinical research programme that is recognised as a site for early phase, ‘first-in-human’ studies of novel therapies and which has also pioneered investigator-led biopsy-based clinical trials, based on bio-samples obtained from patients in the contexts of specific treatments, either standard or experimental.
Our researchers analyse samples to better understand treatment effects in patients, including mechanisms of tumour resistance, immune activation, and potential side effects. Dr Santos, Chief Operating Officer of the MCTRC, highlighted: “This early validation and additional insight provide valuable information for companies looking to further develop and commercialise innovations emerging from the MCTRC.”
Through this comprehensive process, discoveries evolve into tangible benefits for cancer patients and people with lived experience of the disease, addressing important challenges and improving quality of life.
Liquid Biopsy Programme
More recently, the Centre has been shifting toward liquid biopsy as a safer and more convenient alternative to tissue for accessing tumours’ genetic information. Our Teamsters Liquid Biopsy Programme aims to establish a hospital based Liquid Biopsy platform that combines the use of state-of-the-art technologies, including digital droplet PCR (ddPCR) and tissue whole exome sequencing to develop unique personalised assays to detect tumour DNA present in the blood of cancer patients.
Drs Mark Basik, the MCTRC’s associate director and Adriana Aguilar MCTRC’s scientific advisor, are already applying this highly sensitive technology to detect minimal residual disease in the breast cancer setting, and are currently working on making it accessible to more patients.
The clinical utility of these circulating tumour DNA (ctDNA) assays will be validated in the FRIDA clinical trial that will launch in 2025 and will make use of ctDNA assays to identify breast cancer patients who may benefit from faecal microbial transplantation therapy. Dr Basik said: “Our goal is to explore the full potential of this technique to identify new cancer mutations that can act as biomarkers, especially for cancers that don’t have useful blood tests yet.”
Through a recent partnership with Illumina, the liquid biopsy platform will also offer in-house TrueSight Oncology 500 ctDNA tests to our cancer patients. Dr Aguilar said: “This technology is used for comprehensive genomic profiling in cancer, a test that detects mutations, copy number variations, fusions, and other genomic alterations in ctDNA, providing valuable insights for precision oncology without the need for invasive tissue biopsies.”
A study led by Dr April Rose will make use of the TSO500 liquid biopsy tests to identify advanced prostate cancer patients eligible for precision therapies. The molecular results will be discussed at the JGH’s molecular tumour board, the first of such tumour board in Quebec, to inform clinical decisions and support recommendations for potential clinical trials. Dr Batist said: “This approach has the potential to significantly impact our cancer patients by enabling non-invasive, real-time insights into their disease, helping guide more precise and personalised treatment strategies.”
The Warren Y Soper Clinical Proteomics Centre
The evolving data demonstrating that DNA mutations and RNA levels alone are often unreliable in predicting protein production and function led Professor Christoph Borchers of the MCTRC to found the Warren Y Soper Clinical Proteomics Centre. While genomic sequencing has revolutionised cancer research by identifying mutations associated with tumour development, it does not always predict how these mutations affect protein expression, modification, and activity.
Dr Borchers, Director of the Warren Y. Soper Clinical Proteomics Centre, said: “Many mutations are silent or do not significantly alter protein function, while others influence post-translational modifications, protein-protein interactions, and signalling pathways in ways that are not directly inferred from DNA or RNA sequencing alone. Proteins are the targets of newly developed medications – if you don’t know where you’re aiming, you won’t hit the bull’s eye.”
Inaugurated in 2024, the centre is sponsored through funds from the Canadian Foundation for Innovation, the Warren Y Soper Charitable Trust, and a partnership with Agilent Technologies and MRM Proteomics in Canada and Germany. This clinical proteomics laboratory will extend the proteomics capabilities of the MCTRC to involve clinical applications. These methods allow scientists to measure the exact amounts of proteins and metabolites in clinical samples like blood plasma and tumour tissues. They can study hundreds of different targets at the same time.
Technology development and applications are already being advanced in collaboration with national and international academic organisations, e.g. the University of Iceland, Iceland and the Leibniz Institute for Analytical Sciences in Germany, as well with industrial partners including Agilent, Bruker, and Biocrates (Innsbruck, Austria). We plan for these collaborations to turn into a public/private partnership led by the Warren Y. Soper Clinical Proteomics Centre at the Jewish General Hospital.
As the MCTRC intensifies its efforts in providing infrastructure to generate multi-omics data sets, including genomics, transcriptomics, proteomics, metabolomics and radiomics on patient-derived biospecimens and images, it creates pressure on the need for a comprehensive databank that integrates and harmonises these diverse large data sources.
Exactis
In addition to multi-omics data, we are collecting real-world data (RWD) that plays a critical role in shaping the future of precision medicine, clinical trials, and regulatory decision-making. The National Centre of Excellence in Precision Medicine, Exactis, exemplifies this through its Personalize My Treatment Registry, a comprehensive dataset capturing sequential patient data from 16 centers across Canada. This dataset, covering over 11,000 cancer trajectories, provides unique insights into patient responses to treatments in real-world clinical settings.
One of the key advantages of such real-world data collection is the ability to track patient trajectories over time. As Dr Batist highlights: “Understanding these trajectories is essential for analysing treatment patterns, drug administration practices, and patient outcomes across diverse healthcare environments.” Unlike traditional clinical trials that operate under controlled conditions, real-world data reflects actual patient experiences, offering a more nuanced understanding of how therapies perform outside of idealised settings. This data supports hypothesis confirmation and can create a simulated patient population for analysis.
The Radiation Biology and Medical Physics Laboratory
The Radiation Biology and Medical Physics Laboratory led by Dr Enger is a branch of the MCTRC that focuses on the development of next-generation data-driven biomarkers for precision medicine. These biomarkers integrate information from multiple data modalities, offering significant potential for advancing cancer care.
However, several challenges hinder the implementation of multimodal analysis, including reliance on paper-based clinical records, unstandardised medical information capture medical information, the manual analysis of pathology slides, and the absence of integrated databases that would enable seamless data aggregation. As a result, many large and valuable datasets remain under-explored, limiting progress in the field.
To address these barriers, this branch has led a pilot study at the JGH over the past two years, developing an automated workflow to collect, integrate, and anonymise pan-cancer data from various hospital departments to automatically feed the Personalize My Treatment Registry.
The result is a longitudinal multimodal database encompassing thousands of patients and integrating five critical datasets: medical imaging medical reports; pathology data derived from histopathology slides and molecular profiles; molecular data from genomic and proteomic analyses; and clinical data.
Dr Shirin Enger, Head of the lab, said: “By incorporating artificial intelligence, statistical modelling, and advanced image analysis tools, the lab is pioneering novel approaches to understanding cancer biology. These efforts aim to refine patient stratification for better treatment selection, identify subpopulations that may benefit from targeted therapies, optimise treatment planning and monitoring, as well as predict therapeutic responses.”
Furthermore, the lab is leveraging this multimodal database to develop predictive tools and preliminary biomarkers for different cancer types, characterising therapeutic effects, treatment outcomes, resistance to therapy, toxicity profiles, and inter/intra-lesion variability. For example, in a recent study, the group developed an automatic pipeline for temporal monitoring of radiotherapy-induced toxicities in head and neck cancer patients using textual clinical reports available in this database.
Multimodal data integration
In addition to advancing biomarker discovery, the team is tackling fundamental challenges associated with multimodal data integration. These include handling missing data across different modalities, effectively fusing diverse data types, and selecting and validating machine learning models to ensure reproducibility and generalisation.
One particularly innovative approach within the lab focuses on leveraging AI-driven digital histopathology analysis. Whole-slide digital pathology images provide a rapidly widely available and cost-effective alternative to large-scale genomic sequencing.
The team can extract genetic and epigenetic signatures by applying deep learning models to histopathology slides, classify tumour subtypes, and predict therapeutic responses. This approach is particularly valuable in overcoming challenges related to small patient cohorts and the high dimensionality of genomic data, as it allows researchers to derive critical insights into cancer biology from a commonly available clinical resource.
In other studies, real-world patient data has been used to design and develop novel radiotherapy treatment delivery systems as well as accurate, automated AI-based dose prediction models and treatment planning systems.
Powerful Partnerships: Key to advancing translational research at the MCTRC
Since its inception, the MCTRC has relied on strong partnerships to advance fundamental research into clinical benefits to patients. We are founding partners of several provincial, national and international networks of specialists and consortia.
Among the worldwide initiatives, the Centre is a member of the already mentioned WIN Consortium and the Cancer-Covid 19 Consortium (CCC19). WIN’s mission is to improve cancer patient survival and quality of life by fostering international partnerships among academic centres, life science and biotech organisations, technology companies, and non-profit entities. The consortium focuses on conducting worldwide clinical trials and research projects to translate novel cancer treatments from the laboratory to the clinic.
The CCC19 is a collaborative initiative comprising over 120 cancer centres and organisations across the globe with the objective of collecting and disseminating detailed, uniformly organised data on cancer patients diagnosed with COVID-19. This effort aims to inform clinical practice in real time by analysing observational data at scale.
The Centre is also part of Canadian-wide initiatives, including the Marathon of Hope Cancer Centres Network (MOHCCN), a pan-Canadian initiative inspired by Terry Fox’s vision to end cancer. Established by the Terry Fox Research Institute, the network unites leading cancer research and clinical centres across Canada to accelerate the adoption of precision medicine in cancer care.
Locally, the centre is a member of several initiatives, including Q-CROC and RRCancer. Q-CROC, the Quebec Clinical Research Organization in Cancer, is Quebec’s largest clinical oncology research network that includes all of the healthcare institutions conducting clinical oncology research in the province. The organisation brings together over 900 physicians, nurses, managers, and healthcare professionals.
RRCancer is a research network based in Quebec and established by the Fonds de la Recherche en Santé du Québec (FRQS) in 1998. Its goal is to bring together cancer researchers to foster collaboration and accelerate cancer research and treatment advancements. The infrastructure of the MCTRC forms the base of the Axis of Experimental Therapies.
In addition, the MCTRC maintains its tradition of long-lasting collaborations with private companies, including large pharmaceutical corporations, innovative start-ups, and medtech companies. These partnerships facilitate later steps of translating research into real-world applications, fostering innovation, and improving patient outcomes.
Our Annual Cancer Translational Research Horizon Meeting connects academia and industry to drive innovation in cancer research. It fosters new collaborations, accelerates translation into clinical applications, and expands career opportunities for trainees beyond academia.
By collaborating with established industry leaders and emerging companies, the MCTRC leverages a diverse range of expertise, resources, and technologies. These collaborations not only strengthen the research infrastructure but also contribute to the rapid development of new treatments, medical devices, and diagnostic tools, ensuring that the latest advancements in cancer care are accessible to patients more quickly and efficiently.
Future outlook
We are seeing more overlap between different medical fields, which is natural given the limited number of molecules and pathways involved. While stepping outside traditional areas of expertise can be challenging, a more integrated approach is important. Innovative research is key to improving patient care and outcomes in oncology and beyond.
MCTRC is leading this shift by breaking down silos and building multidisciplinary teams that bring together experts with diverse skills. By fostering collaboration and innovation, we help connect teams across disciplines, support their success by linking them with industry partners, and create opportunities to secure innovation funding. With this approach and the use of cutting-edge technologies, MCTRC is well-positioned to drive meaningful advancements in cancer research and develop more effective, personalized treatments for patients worldwide.
Please note, this article will also appear in the 21st edition of our quarterly publication.
Source link