Torsten Nielsen

Professor

Research Interests

Biomarker development
Breast Cancer
Cancer Diagnosis and Detection
Cancer of the Musculoskeletal System
Clinical trials
Epigenomics
Experimental Therapeutics
Immuno-oncology
Tissue-based diagnostic technologies
Translational research
Genetically engineered mouse models

Relevant Thesis-Based Degree Programs

Affiliations to Research Centres, Institutes & Clusters

Research Options

I am available and interested in collaborations (e.g. clusters, grants).
I am interested in and conduct interdisciplinary research.
 
 

Research Methodology

Tissue Microarrays
NanoString
Cancer Treatment & Outcome data
Epigenomics
Spatial transcriptomics

Recruitment

Doctoral students
Postdoctoral Fellows
2024
2025

Fusion oncogene biology and experimental therapeutics Cancer biomarker development: immuno-oncology, in situ multiplex diagnostics, NanoString profiling

  • proven record of research productivity – a graduate degree and/or research papers in PubMed-indexed journals
  • real cancer research training & experience – not just undergraduate coursework
  • you can bring a new skill, method or approach to my laboratory, and better yet can independently think of a way that it could be applied to build on my published research
  • you are approaching me with a good idea related to the fields I am working in, based on clinical need or technological innovation
  • truly excellent English language skills – written and spoken
  • track record of winning major awards for academics and/or research
I am open to hosting Visiting International Research Students (non-degree, up to 12 months).
I am interested in supervising students to conduct interdisciplinary research.

Complete these steps before you reach out to a faculty member!

Check requirements
  • Familiarize yourself with program requirements. You want to learn as much as possible from the information available to you before you reach out to a faculty member. Be sure to visit the graduate degree program listing and program-specific websites.
  • Check whether the program requires you to seek commitment from a supervisor prior to submitting an application. For some programs this is an essential step while others match successful applicants with faculty members within the first year of study. This is either indicated in the program profile under "Admission Information & Requirements" - "Prepare Application" - "Supervision" or on the program website.
Focus your search
  • Identify specific faculty members who are conducting research in your specific area of interest.
  • Establish that your research interests align with the faculty member’s research interests.
    • Read up on the faculty members in the program and the research being conducted in the department.
    • Familiarize yourself with their work, read their recent publications and past theses/dissertations that they supervised. Be certain that their research is indeed what you are hoping to study.
Make a good impression
  • Compose an error-free and grammatically correct email addressed to your specifically targeted faculty member, and remember to use their correct titles.
    • Do not send non-specific, mass emails to everyone in the department hoping for a match.
    • Address the faculty members by name. Your contact should be genuine rather than generic.
  • Include a brief outline of your academic background, why you are interested in working with the faculty member, and what experience you could bring to the department. The supervision enquiry form guides you with targeted questions. Ensure to craft compelling answers to these questions.
  • Highlight your achievements and why you are a top student. Faculty members receive dozens of requests from prospective students and you may have less than 30 seconds to pique someone’s interest.
  • Demonstrate that you are familiar with their research:
    • Convey the specific ways you are a good fit for the program.
    • Convey the specific ways the program/lab/faculty member is a good fit for the research you are interested in/already conducting.
  • Be enthusiastic, but don’t overdo it.
Attend an information session

G+PS regularly provides virtual sessions that focus on admission requirements and procedures and tips how to improve your application.

 

ADVICE AND INSIGHTS FROM UBC FACULTY ON REACHING OUT TO SUPERVISORS

These videos contain some general advice from faculty across UBC on finding and reaching out to a potential thesis supervisor.

Great Supervisor Week Mentions

Each year graduate students are encouraged to give kudos to their supervisors through social media and our website as part of #GreatSupervisorWeek. Below are students who mentioned this supervisor since the initiative was started in 2017.

 

I am pleased to have Dr. Nielsen as a supervisor for my PhD studies at UBC. For the past three years, I got to know an incredibly nice and supportive mentor that always guides me towards my goals. My vision to discover new and better ways to improve breast cancer patients' lives brought me 10,000km across the world to work under the supervision of Dr. Nielsen as an expert leader in the field. He provided me with the ideal learning atmosphere of skills, knowledge, resources, leadership and supportive interpersonal relationship to help achieve my goals. He diligently encourages me to maximize my potentials and draw up plans of actions to succeed all along the path of my career.

Karama Asleh (2018)

 

Graduate Student Supervision

Doctoral Student Supervision

Dissertations completed in 2010 or later are listed below. Please note that there is a 6-12 month delay to add the latest dissertations.

Clinical utility of biomarkers for basal-like breast cancer (2022)

The genomic subtyping of breast cancers into luminal A, luminal B, human epidermal growth factor receptor-2 (Her2)-Enriched and basal-like has remarkably advanced breast cancer diagnosis, treatment, and outcome. However, comparatively few advancements have been made in identifying biomarkers that can tailor treatments for the aggressive basal-like subtype. This group is clinically approximated as triple negative breast cancer (TNBC), characterized by immunohistochemical negativity for estrogen receptor, progesterone receptor, and Her2. However, this definition identifies a biologically heterogenous group, highlighting the complexity of guiding therapeutic choices for TNBC including those with basal-like molecular biology.My research goals have included identifying improved diagnostic biomarkers that can guide better therapeutic options for these aggressive cancers in a clinically-applicable manner using immunohistochemistry, RNA expression, and proteomic profiling. First, applying an optimized immunohistochemical panel, defined by nestin positivity or INPP4B negativity, on 239 specimens from the phase III SBG0102 clinical trial, I was able to identify those with a basal-like gene expression subtype and to predict which metastatic breast cancer patients benefit from gemcitabine chemotherapy. Then, using a 770-gene RNA panel targeting multiple biological mechanisms and additional 30-custom genes related to capecitabine metabolism on 111 TNBC specimens from the phase III FinXX adjuvant capecitabine trial, I found that genes and metagenes related to immune response (cytotoxic cells, PDL2), endothelial, mast cells and 38 individual genes are the most significantly associated with capecitabine benefit. Finally, to characterize the heterogeneity of basal-like and triple negative breast cancers beyond current genomic classifications, I used a method called SP3-Clinical Tissue Proteomics, compatible with routine clinical specimens, for comprehensive protein profiling which revealed distinct subgroups within basal-like and triple negative breast cancers. Specifically, within 88 TNBC samples, four proteomic clusters were evident, which had features of “basal-immune hot”, “basal-immune cold”, “mesenchymal”, and “luminal”, each with distinct clinical outcomes. This work sets a foundation for developing clinically-applicable tests that can tackle the diversity of basal-like and triple negative breast cancers using standard formalin-fixed pathology clinical trial materials. The findings presented may guide the selection of breast cancer patients for clinical trial evaluation of existing chemotherapy or emerging therapies.

View record

The immune microenvironment of sarcomas: a comprehensive evaluation of infiltrating immune cells and checkpoint biomarkers in musculoskeletal tumors (2021)

Sarcomas are aggressive cancers of the connective tissues, such as bone, muscle, cartilage, and fat. Despite their diverse origins, sarcomas are predominantly treated by surgery and radiation, as conventional chemotherapy has limited benefit for most subtypes. When sarcomas recur or metastasize, there are few options for systemic therapy, and prognosis is very poor. Despite advancements in our understanding of the molecular drivers of sarcomas, almost no new treatments have proven benefit for metastatic sarcomas. Immunotherapy has shown value for other cancers, such as melanoma and lung cancer; however, sarcomas lag behind the common cancers in our understanding of their immune microenvironment and potential for treatment with immunotherapeutics. Early trials using single-agent immune checkpoint inhibitors in sarcomas delivered mixed results, but these studies somewhat indiscriminately lumped together different sarcoma subtypes that might have critical immunological differences. My study employs tissue microarrays incorporating 1360 sarcoma specimens (spanning 23 subtypes) to characterize immune infiltrates and expression of targetable immune biomarkers, using immunohistochemistry. Genomically-complex sarcoma types – driven by mutations and/or copy-number alterations – are found to have much higher levels of lymphocytic and phagocytic immune infiltrates than translocation-associated sarcomas. Across nearly all subtypes, tumor-associated macrophages outnumber tumor-infiltrating lymphocytes, predominately M2 (anti-inflammatory) macrophages. Expression of the target of first-generation immune checkpoint PD-(L)1 is uniformly low, but expression of LAG-3 and TIM-3 – emerging immune checkpoints – is significantly more common. Expression of anti-phagocytic immune checkpoint CD47 is yet more predominant, displaying all-or-nothing expression with 100% positivity seen in over half of positive cases. To further characterize the lymphocytic response, T-cell receptor (TCR) sequencing was performed on specimens from 25 sarcoma patients on a clinical trial of tremelimumab (anti-CTLA-4) with durvalumab (anti-PD-L1). We found that the TCR repertoire is richer and more diverse among the genomically-complex sarcomas relative to the translocation-associated sarcomas, and following immune checkpoint blockade, we observed an overall increase in the clonality of the peripheral TCR repertoire. My study demonstrates a tangible positive relationship between genomic complexity and immunogenicity, and highlights novel immune checkpoints of relevance to sarcomas. As such, this work provides the essential translational background to direct the use of immunotherapy in sarcoma management.

View record

Immune biomarkers in breast cancer pathology specimens: characterization and clinical implications (2019)

Immunotherapy is dramatically changing the landscape of cancer treatment and is becoming incorporated into the standard of care for some tumor types. Until recently, breast cancer has not been generally considered particularly immunogenic. However, breast cancer is a heterogeneous disease and increasing evidence suggests that patients with basal-like breast cancer, an aggressive subtype lacking targeted therapy options, may be amenable to immunotherapy. My research goals have included the investigation and clinical characterization of two emerging targetable immune checkpoint biomarkers: lymphocyte-activation gene 3 (LAG-3) and the T-cell Immunoglobulin and Mucin domain-containing molecule 3 (TIM-3), by applying immunohistochemistry to a well-annotated tissue microarray cohort of 3,992 breast cancers. As an additional research goal, I evaluated a novel in situ multiplex biomarker assessment method (Nanostring-based digital spatial profiling-DSP) for its compatibility with breast cancer tissue microarrays, to generate immune profiles from patient surgical specimens. I report that the expression of LAG-3 or TIM-3 on intra-epithelial tumor-infiltrating lymphocytes (iTILs) was observed in a minority of cases (11%) in the whole cohort, but was significantly enriched in basal-like breast cancers (33% and 28% of basal-like breast cancers being infiltrated with LAG-3+ and TIM-3+iTILs, respectively). Furthermore, I found that LAG-3+iTILs and TIM-3+iTILs were present in breast cancers co-infiltrated with established immunotherapy targets (program cell death-1/PD-1 and its ligand, PD-L1). In multivariate analyses, LAG-3+iTILs or TIM-3+iTILs were independent favorable prognostic factors in breast cancer patients. In the last part of the thesis, I profiled the tumor immune microenvironment of two basal-like-enriched breast cancer cohorts, quantifying the expression of 31 immuno-oncology biomarkers using DSP. I then validated the digital counts for CD8 and PD1 by comparing with immunohistochemistry, and CD45 digital counts by comparing with hematoxylin & eosin-stained stromal TILs counts. Lastly, I identified a 4-biomarker signature indicative of a pre-existing immunity in breast cancer patients. The body of work presented here may help guide the selection of breast cancer patients for clinical trial evaluation of emerging immunotherapy agents. Furthermore, I show that digital spatial profiling technology can efficiently and quantitatively profile immune expression on breast cancer patient specimens using only a tiny fraction from precious tumor samples.

View record

The FUS-DDIT3 interactome in myxoid liposarcoma (2019)

Myxoid liposarcoma is a malignant fatty tumour that develops in the deep soft tissue. While local control rates are good, current chemotherapy options remain ineffective against metastatic disease. Myxoid liposarcoma is characterized by a balanced translocation involving FUS (12q13) and DDIT3 (16p11). The resulting FUS-DDIT3 oncoprotein is proposed to function as an aberrant transcription factor but its exact mechanism of action has remained unclear, rendering it difficult to formulate rational strategies for targeted therapy. A current gap in knowledge behind the oncogenic functions of FUS-DDIT3 is the lack of comprehensive data on its interactome, which could identify the oncoprotein's key partners. This study utilized immunoprecipitation-mass spectrometry to identify the FUS-DDIT3 interactome in whole cell lysates of myxoid liposarcoma cells, and results showed an enrichment of RNA processing proteins. RNA-seq analysis was performed on myxoid liposarcoma cells after FUS-DDIT3 knockdown to look for changes in the alternative splicing profile, but no evidence of such changes was seen. Further TMT-labeled immunoprecipitation-mass spectrometry analyses in nuclear lysates of myxoid liposarcoma cells showed that members of several chromatin regulatory complexes were present in the FUS-DDIT3 interactome. These complexes included the NuRD, SWI/SNF, PRC1, PRC2 and MLL1 COMPASS-like complexes. Co-immunoprecipitation experiments validated the association of FUS-DDIT3 with BRG1/SMARCA4, BAF155/SMARCC1, BAF57/SMARCE1, HDAC2, KDM1A, and MTA1. Knockdown of FUS-DDIT3 also reduced H3K27ac levels at the promoter of a gene target, PTX3. Other sarcoma fusion oncoproteins have been reported to interact with chromatin regulators and affect histone modifications or chromatin remodeling as one of their oncogenic mechanisms. Data from this study suggests that FUS-DDIT3 may utilize a similar epigenetic mechanism of action, providing potential candidates for targeted therapy as epigenetic aberrations are potentially reversible by existing and emerging epigenetic drugs.

View record

Characterization of Epithelioid Sarcoma Using Massively Parallel DNA and RNA Sequencing and In Vitro Models (2017)

Epithelioid sarcoma is a soft tissue tumor with an unusual predilection for the distal extremities in young adults. Despite wide-margin resections the 10-year survival is in the range of 50%. The biology of epithelioid sarcoma remains incompletely understood, but one key feature is the loss of SMARCB1. We use whole genome sequencing of five cases of epithelioid sarcoma matched to normal germline DNA, looking for mutations other than SMARCB1. These index cases are supplemented with three additional tumors and three cell lines that undergo whole transcriptome sequencing and are analyzed for somatic point mutations, copy number changes, translocations, and expression patterns. Unlike the situation in other SMARCB1 inactivated tumors, we find a complex genome with a relatively high mutational burden. However, aberrations of SMARCB1 remain the only consistent mutation. Some cases do not show biallelic DNA-level inactivation of this gene which leads us to examine other possible second-hit silencing mechanisms. With the significance attributed to SMARCB1 loss in the genomic landscape of epithelioid sarcoma, we explore two approaches, namely EZH2 and SMARCA4 inhibitions, to specifically exploit this abnormality and suggest novel therapeutic options.

View record

Targeted activity of histone deacetylase inhibition in synovial sarcoma (2017)

Synovial sarcoma is an aggressive soft tissue cancer affecting primarily adolescents and young adults and characterized by the known chimeric fusion oncoprotein SS18-SSX resulting from a t(X;18) translocation. Oncoprotein-mediated interactions with chromatin-modification proteins are known to influence transformation; however targeted therapies are not currently available. Patients remain at high-risk for local recurrence and metastasis and see a 10-year survival rate of approximately 50%. Accordingly, it was hypothesized that therapeutic compounds able to disrupt the SS18-SSX driving complex would be clinically effective against synovial sarcoma. The proximity ligation assay was employed to study the drug-mediated disruption of the key SS18-SSX-mediated protein interactions that drive sarcomagenesis, and was developed for use to demonstrate targeted drug efficacy. Results identified HDAC inhibitors as potent in disrupting the oncogenic protein complex. The combination of HDAC and proteasome inhibitors was further found to be synergistic against synovial sarcoma. This pre-clinical study was used as support for a novel clinical trial proposal for translocation-associated soft tissue sarcomas. The effects of HDAC inhibition on genome-wide transcription patterns was further assessed by RNA-seq analysis. Uniquely to synovial sarcoma, it was revealed that HDAC inhibition elicits loss of the SS18-SSX oncoprotein and provokes significant pro-apoptotic BIK expression correlating to CDKN2A reactivation. From this, the contributing role of CDKN2A reactivation and reactive oxygen species accumulation in HDAC-inhibitor mediated apoptosis in synovial sarcoma was demonstrated.

View record

Clinical performance of diagnostic, prognostic and predictive immunohistochemical biomarkers for hormone receptor-negative breast cancer (2015)

Gene expression profiling of breast cancer delineates a particularly aggressive subtype referred to as “basal-like”, which comprises ~15% of all cases, afflicts younger women and is refractory to endocrine and anti-HER2 therapies. Immunohistochemical surrogate definitions for basal-like breast cancer, such as the ER/PR/HER2 triple negative phenotype and models incorporating positive expression of cytokeratin 5 (CK5/6) and/or epidermal growth factor receptor are more amenable to implementation in a clinical setting. Despite this and the fact that basal-like breast carcinomas are being increasingly recognized as a distinct clinical entity, there is no diagnostic method used and reported in routine practice. Without a reproducible test to identify this aggressive subtype in the clinic there will be no ability to establish clearly defined intake criteria for subtype-specific clinical trials, translating to no progress in the management of this form of the disease and little change in breast cancer survival rates for the foreseeable future.A first evaluation of performance of the triple negative definition and various surrogate immunopanels for basal-like breast cancer in clinical laboratories is described in the initial chapters of this dissertation. Considerable staining variability of individual biomarkers included in immunopanels typically led to only moderate concordance with a gene expression gold standard for identification of basal-like breast carcinomas. Lack of standardization was the underlying reason for all of the observed variability, supporting the notion that further standardization efforts through continual participation in external quality assurance programs are needed before routine diagnosis of basal-like breast carcinomas could be made in a clinical setting.In light of this, we sought to identify more easy-to-interpret and robust biomarkers for this disproportionately deadly type of breast cancer. A parallel comparison of 46 proposed immunohistochemical biomarkers of basal-like breast cancer was performed against a gene expression profile gold standard. Results from that survey determined that loss of expression of INPP4B and positive expression of nestin had the strongest associations with this aggressive subtype. Paving the way for further studies, this comprehensive immunohistochemical biomarker survey is a necessary step to determine an optimized surrogate immunopanel that best defines basal-like breast cancer in a practical and clinically-accessible way.

View record

Biological classification of clinical breast cancer using tissue microarrays (2008)

Gene expression profiles have identified five major molecular breast cancer subtypes (Luminal A, Luminal B, Basal-like, HER2+/estrogen receptor− , and Normal Breast-like) that show significant differences in survival. The cost and complexity of gene expression technology has impeded its clinical implementation. By comparison, immunohistochemistry is an economical technique applicable to the standard formalin-fixed, paraffin-embedded material commonly used in hospital labs, and has the advantage of simultaneously interpretation with histomorphology. In this thesis, I hypothesize that a surrogate panel of immunohistochemical biomarkers can be developed to discriminate the breast cancer biological subtypes. The main study cohort consists of over 4000 primary invasive breast tumors, assembled into tissue microarrays. These patients were referred to the British Columbia Cancer Agency between 1986-1992 and have staging, pathology, treatment and follow-up information. In summary, our results demonstrate that (1) the rabbit monoclonal antibody, SP1, is an improved standard for immunohistochemiscal estrogen receptor assessment in breast cancer; (2) the transcription factor, GATA-3, is almost exclusively expressed among estrogen receptor positive tumors but does not seem to predict for tamoxifen response among estrogen receptor positive patients; (3) the proliferation marker, Ki-67, together with HER2 can segregate Luminal A from Luminal B subtypes, which carry distinct risks for breast cancer relapse and death; and (4) the inclusion of the basal markers EGFR and ck5/6 to “triple negative” breast cancers provides a more specific definition of basal-like breast cancer that better predicts patient survival. These results consistently demonstrate that an immunopanel of six biomarkers (estrogen receptor, progesterone receptor, HER2, Ki-67, epidermal growth factor receptor and cytokeratin 5/6) can be readily applied to standard pathology specimens to subtype breast cancer samples based on their underlying molecular biology. These findings have been considered sufficient to justify application of this panel onto NCIC (MA5, MA12) and CALGB (9341 and 9741) clinical trials specimens. This followup work which is underway and will determine if the six marker immunopanel can guide decisions about which patients need aggressive systemic drug treatment, and thereby ensure patients get the most effective, individualized adjuvant systemic therapy for their breast tumor.

View record

Synovial sarcoma : translating gene expression into patient care (2008)

Synovial sarcoma is a soft tissue tumor defined by the presence of t(X;18)(p11.2;q11.2), fusing the SYT (SS18) gene on chromosome 18 and one of three SSX genes on chromosome X. T(X;18) results in production of a fusion protein (SYT-SSX) that is thought to underlie synovial sarcoma pathogenesis through aberrant targeting of both activating (trithorax, SWI/SNF) and repressing (Polycomb) transcription factors when expressed in a stem or progenitor-like cellular background.Clinically, synovial sarcomas present considerable diagnostic and therapeutic challenges. Whereas the classical biphasic histology is distinctive, the more common monophasic histology can be difficult to differentiate from other spindle cell tumors. In these situations, detection of t(X;18) is the gold standard for diagnosis, but it is a specialized and time-consuming process. Immunohistochemistry can be helpful, but no marker that is both highly sensitive and specific is available. Here I describe a fluorescence in situ hybridization based method employing an SYT break-apart probe set that can expedite detection of t(X;18). I also report that TLE1, which was identified in gene expression studies as a good discriminator of synovial sarcoma from other mesenchymal tumors, is a highly sensitive and specific immunohistochemical marker for synovial sarcoma. Both of these novel diagnostic techniques are applicable to small tissue samples such as core needle biopsies and are now being used clinically.The diagnosis of synovial sarcoma carries a poor prognosis and the 10-year overall survival rate is approximately 50%, most of whom are young adults. The addition of chemotherapy to surgical resection (the mainstay of treatment) does not appear to improve overall survival. Thus, there is a strong need for development of a clinically effective systemic therapy to improve patient outcome. I describe preclinical studies that demonstrate the Hsp90 inhibitor 17-allylamino-17-demethoxygeldanamycin (17-AAG) inhibits proliferation of synovial sarcoma by inducing apoptosis and that this is associated with degradation of multiple receptor tyrosine kinases and disruption of the SYT-SSX-β-catenin interaction. I also identify a subset of synovial sarcoma cells, typified by expression of CD133, which exhibit stem-like properties and are relatively resistant to doxorubicin but susceptible to 17-AAG at clinically relevant concentrations.

View record

 
 

If this is your researcher profile you can log in to the Faculty & Staff portal to update your details and provide recruitment preferences.

 
 

Sign up for an information session to connect with students, advisors and faculty from across UBC and gain application advice and insight.