Research Interests
Antiviral immunity, Auto-Immune Diseases, Cancer, Cell Signaling, Gene Regulation and Expression, Genetics, Infectious diseases, Inflammation, Innate immunity
Research Focus Teams
Cancer, COVID-19, Cardiovascular, Aging, Obesity, Crohn’s/Colitis
Departments
Contact
Email: ken.harder@ubc.ca
Office Phone: 604–827–4349
Publications
Lab Website
Dr. Harder received his PhD in genetics from the University of British Columbia (UBC) in 1996. He received Post-Doctoral training and was a Research Scientist at the Ludwig Institute for Cancer Research, Melbourne Tumour Biology Branch in Australia, from 1998 to 2006. In 2006 Dr. Harder returned to UBC as a Canada Research Chair in Host-pathogen/tumour biology. He is currently leader of the Department of Microbiology and Immunology’s Infection, Inflammation and Immunity (I3) research group within the Life Sciences Institute. Dr. Harder and his laboratory are interested in understanding how particular populations of immune cells develop and function in health and disease. His group has extensive expertise in diverse medical research disciplines including biochemistry, cellular immunology, hematology, autoimmunity as well as models of intestinal infection/inflammation and cancer.
Most recently, Dr. Harder’s group has developed and employed systems biology approaches to identify cytokine-mediated immunological perturbations associated with cancer. This is part of a larger goal to develop better tumour immunotherapy approaches based on our improved understanding of phagocyte biology and immunosuppressive pathways undermining tumour immunity. His research is heavily reliant on advancements in high-dimensional single cell technologies and bioinformatics, harnessing both mass cytometry (CyTOF) and new cellular indexing of transcription and epitopes scRNAseq (CITE-seq) platforms, to advance the systems level understanding of the immune system in health and disease.
The Harder lab has a long-standing interest in understanding the developmental programs guiding monocyte, macrophage, neutrophil and dendritic cell production from hematopoietic stem and progenitor cells. We are particularly interested in delineating the key phagocyte subsets, genes, and signalling pathways associated with chronic inflammatory diseases such as cancer, inflammatory bowel disease, heart disease, obesity, aging, and neurodegeneration, as recent studies by us and others have shown that phagocytes play key roles in each of these diseases.
Our studies have revealed the widespread impact that chronic inflammatory diseases have on these lineages of immune cells and our recent work has focused on understanding how key cytokines and signalling pathways control phagocyte fate and function. We contend that new treatments for these diseases will require a better understanding of phagocyte biology associated with disease, and will entail new strategies to target phagocytes with drugs, biologics, genetic modification strategies, or adoptive cell therapy approaches.
We employ a variety of cutting-edge techniques in our studies including systems biology single cell proteomic and transcriptomic technologies (CyTOF, scRNA-seq, CITE-seq) that rely on computational machine learning platforms to allow high dimensional, deep phenotyping of tens of thousands of individual cells in health and disease, simultaneously. Other technologies used in the lab include: genetically engineered mouse models, tissue culture/cellular immunology assays, flow cytometry, immunohistochemistry, molecular biology, CRISPR/Cas9 mutagenesis, siRNA technology, RNA-seq, new adoptive cell therapy approaches, and most recently lipid nanoparticle (LNP) mediated mRNA delivery in vivo.
High dimensional immunophenotyping of cancer. One of the primary focuses of the lab is to better understand the tumour-induced immunosuppressive pathways that limit anti-tumour immunity. However, studying the immune system in the context of cancer is a daunting task. The immune system is comprised of dozens of different cell types, each characterized by an even greater number of cell phenotypes associated with distinct developmental or activation states spread across multiple tissue compartments. To address this complexity, we are using, and further developing, high dimensional single cell analytical approaches to simultaneously interrogate the immune cell subsets, signalling pathway activation patterns, and gene expression programs, associated with cancer. Our studies have identified new cytokine-driven immunosuppressive pathways as well as cancer-induced phagocyte subsets that we hypothesize lead to suppression of adaptive immunity against cancer. We are using spontaneous mutagen and chronic inflammation-induced colon cancer models as well as syngeneic melanoma, colon, breast, and ovarian cell line and organoids systems for these studies.
* We are seeking exceptional, highly-motivated, MSc/PhD students for this project with an interest and demonstrated skills in computer science, programming and bioinformatics. Students with a combined major in computer science, genome sciences, and either biology or immunology are encouraged to apply. Please send a statement of interest, CV and academic transcripts to Dr. Harder.
Understanding and harnessing the function of patrolling monocytes (pMos) in disease. pMos are circulatory vessel migratory monocytes that play key roles in responding to tissue damage, heart disease and cancer. We are developing methods to grow pMos in the lab so that they can be harnessed for use following adoptive cell transfer to treat Alzheimer’s disease and cancer. We have identified a key protein tyrosine kinase regulating pMos numbers, and are studying the gene expression and signalling pathways that this kinase regulates in pMos, to better understand and control pMos development, function and lifespan.
See Roberts et al, 2020 (https://pubmed.ncbi.nlm.nih.gov/32151196/) for an example of our work in this area.
Phagocyte immune checkpoint regulation in the control of metabolic disease, aging, and cancer. Immune checkpoint blockade therapy has revolutionized the treatment of some cancers. These treatments arose from fundamental studies in mouse models of T cell signalling and the inhibitory receptor/ligand interactions governing T cell activation. However, inhibitory receptors are widely expressed across most immune cell lineages and are particularly abundant on monocytes, macrophages and dendritic cells. Our lab is studying how changes in the balance of stimulatory and inhibitory signalling impacts phagocyte function in cancer and metabolic diseases. For example, we know that the SIRPa/CD47 inhibitory receptor/ligand pair is an important regulator of macrophage phagocytosis of stressed, necrotic and tumour cells. We previously identified a key regulator of this receptor and are exploring how this pathway and others can be targeted to improve cancer immunotherapy and phagocyte targeting of stressed, senescent, or necrotic cells. We propose that future drugs targeting myeloid cell immune checkpoints will greatly improve our ability to treat chronic inflammatory diseases.
See Harder et al, 2001 (https://pubmed.ncbi.nlm.nih.gov/11672542/) for an example of our early work in this area.
Improving LNP technology to enhance phagocyte function in disease and to develop new cancer vaccines. LNP-mRNA vaccines from BioNtech/Pfizer and Moderna have been instrumental in reversing the course of the COVID-19 pandemic and have clearly shown the power of this technology against infectious disease. Together with our collaborators in the Nanomedicines Innovation Network (NMIN- https://www.nanomedicines.ca), we are working to develop new and improved LNP formulations encapsulating designer mRNAs to improve immune system function in cancer and other chronic inflammatory diseases. We are using high throughput microfluidic, single cell multiplexed transcriptomic and proteomic platforms, to screen libraries of distinct LNP formulations to identify those with enhanced immune subset tropisms and regulatory features. We envision that this project will lead to new nanomedicines and improved vaccine designs.