Earlybird ticket allocation is almost exhausted - secure your spot now!
Earlybird ticket allocation is almost exhausted - secure your spot now!
Signed in as:
filler@godaddy.com
Talk Title: TBA
Talk Title: The molecular basis of target recognition and cleavage by CRISPR-Cas13
Talk Title: TBA
Talk Title: Next Generation CRISPR tools to model and interrogate cancer
Abstract Title: Advancing the genetic engineering toolbox by combining AsCas12a knock-in mice with ultra-compact screening
Authors: Wei Jin1,2,3,4,#, Yexuan Deng1,2,3,4,5,#, John E. La Marca1,2,3,4,#, Emily J. Lelliott1,2,3,4, Sarah T. Diepstraten3,4, Christina König1,2,3, Lin Tai1, Valentina Snetkova6, Kristel M. Dorighi6, Luke Hoberecht7, Millicent G. Hedditch3, Lauren Whelan3, Geraldine Healey1, Dan Fayle1, Kieran Lau1, Margaret A. Potts1,2,3,4, Moore Z. Chen8, Angus P. R. Johnston8, Yang Liao1,2, Wei Shi1,2, Andrew J. Kueh1,2,3,4, Benjamin Haley6,9,*, Jean-Philippe Fortin7,*, Marco J. Herold1,2,3,4,*,^
Affiliations: 1Olivia Newton-John Cancer Research Institute, Heidelberg, Melbourne, Australia 2School of Cancer Medicine, La Trobe University, Bundoora, Melbourne, Australia 3The Walter and Eliza Hall Institute of Medical Research, Parkville, Melbourne, Australia 4Department of Medical Biology, University of Melbourne, Parkville, Melbourne, Australia 5The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China 6Department of Molecular Biology, Genentech, Inc., South San Francisco, California, USA 7Computational Sciences, Genentech, Inc., South San Francisco, California, USA 8Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Melbourne, Australia 9current address: Université de Montréal, Centre de recherche de l'Hôpital Maisonneuve Rosemont #share first authorship *share senior authorship ^corresponding author
Full Abstract: CRISPR-Cas9 technology has revolutionized genetic research, particularly in the discovery of novel tumour drivers and resistance factors through CRISPR screens. Cas12a (Cpf1) distinguishes itself from Cas9 by short crRNAs and a unique PAM site . Additionally, intrinsic RNase activity simplify multiplexed gene targeting via the processing of individual crRNAs from a pre-crRNA-encoding RNA. Here, we present a mouse model that constitutively expresses enhanced Acidaminococcus sp.Cas12a (enAsCas12a) linked to an mCherry fluorescent reporter. We validated robust expression of Cas12a in our mouse model across multiple tissues, with a particular focus on haematopoietic organs, a key facet of our research. We demonstrate efficient single and multiplexed gene-editing in vitro, using primary and transformed cells from enAsCas12a mice. We further demonstrate successful in vivo gene-editing, using normal and cancer-prone enAsCas12a stem cells to reconstitute the haematopoietic system of wild-type mice. We next generated compact, genome-wide Cas12a knockout libraries targeting each gene with four crRNAs encoded across one (Scherzo) or two (Menuetto) vectors, and demonstrated the utility of these libraries across multiple screens: in vitro enrichment screening in lymphoma cells, in vitro drop-out screening in immortalised MDFs, and in vivo enrichment screening in the haematopoietic lineage of mice to identify lymphoma-driving events. The consistency and robustness of the data extracted from each of our screens underscore the high effectiveness and broad potential of these new tools. Finally, we demonstrate CRISPR multiplexing via simultaneous gene knockout (via Cas12a) and activation (via dCas-SAM) using primary T cells and mouse dermal fibroblasts (MDFs). This highlights the compatibility of our enAsCas12a mouse model with other CRISPR technologies. Collectively, our enAsCas12a mouse model and accompanying crRNA expression libraries enhance genome engineering capabilities and complement current CRISPR technologies.
Abstract Title: Using a spatial CRISPR screen to identify regulators of the immune microenvironment in non-small cell lung cancer
Authors: Marina Leiwe1,2, Claire Marceaux1,2, Daniel Batey1, Velimir Gayevskiy1, Tracy Leong3, Marie-Liesse Asselin-Labat1,2
Affiliations: 1Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville Victoria 3052, Australia 2Department of Medical Biology, The University of Melbourne, Parkville, Victoria 3010, Australia 3Olivia Newton John Cancer Research Institute and Austin Health, Heidelberg, Victoria, 3084
Full Abstract: Although immunotherapy has shown major success in the clinic, most lung cancer patients fail to respond long-term to the current treatment options. Immune cell composition and spatial organisation of the tumour microenvironment (TME) determine patient response to immunotherapy. However, how cancer cells modulate cellular arrangements within the TME remains poorly understood. Here, we combine a spatial in vivo CRISPR screen, called Perturb-map, with spatial omic technologies to decipher the mechanisms tumour cells employ to reshape the immune microenvironment in non-small cell lung cancer.
Perturb-map adds spatial resolution to pooled CRISPR screens, hence allowing the identification of extracellular gene functions within the native tissue environment.
As this approach is limited in the number of genes that can be studied in parallel, we curated a candidate gene library based on two RNA-seq datasets of lung adenocarcinoma patient biopsies: 1) comparing tumours sensitive or resistant to anti-PD-L1 therapy and 2) comparing tumours with high or low immune infiltration. Correlating these two transcriptomic datasets identified genes associated with both, response to immune checkpoint blockade and immune infiltration, suggesting these gene products may be involved in immunotherapy resistance via modulation of the tumour immune microenvironment.
We have completed a pilot screen depleting 12 candidate genes in murine lung carcinoma cells before injecting pooled knock-out cells into immunocompetent mice to grow orthotopic tumours. We identified two genes, which when depleted, decreased the number of tumour lesions as well as the percentage of total tumour burden compared to control lesions. Our ongoing work uses multiplex imaging techniques to investigate how the individual gene depletions influence composition of the immune microenvironment.
Genes identified in our study may suggest novel drug targets aiming to recruit immune cells to the tumour site and may improve patient outcome in combination with conventional immunotherapies like immune checkpoint blockade.
Abstract Title: Recursive CRISPR screening identifies a functional link between P300 and SF3B3 in transcriptional elongation control
Authors: Dane Vassiliadis 1,2, Jesse J. Balic1,2, Andrea Gillespie1,2, Olivia Braniff1,2, William Rothnie1,2, Oliver Sinclair1, Kelsy Prest1,2, Andrew Das1,2, Ching-Seng Ang3, and Mark A. Dawson1,2,4,^.Dane Vassiliadis1,2,*,^, Jesse J. Balic 1,2, Andrea Gillespie 1,2, Olivia Braniff 1,2, William Rothnie 1,2, Oliver Sinclair 1, Kelsy Prest 1,2, Andrew Das 1,2, Ching-Seng Ang 3, and Mark A. Dawson 1,2,4.
Affiliations: 1 Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia; 2 Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia; 3 Bio21 Mass Spectrometry and Proteomics Facility, The University of Melbourne, Parkville, Victoria 3052, Australia; 4 The University of Melbourne Centre for Cancer Research, The University of Melbourne, Melbourne, Victoria, Australia; 2 Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia; 3 Bio21 Mass Spectrometry and Proteomics Facility, The University of Melbourne, Parkville, Victoria 3052, Australia; 4 The University of Melbourne Centre for Cancer Research, The University of Melbourne, Melbourne, Victoria, Australia.
Full Abstract: The expression of eukaryotic protein-coding genes is the product of transcriptional (e.g. initiation, elongation and termination) and co-transcriptional processes (e.g. splicing, modification and export). How diverse input signals are relayed to, and interpreted by transcription factors and associated co-factors to govern RNA Pol II activity remains unclear. Here we developed a gene expression reporter system traceable in real-time by flow cytometry and combined this with CRISPR/Cas9 screens to identify epistatic relationships underpinning transcriptional co-activator inhibition in human cells. We find that loss of the U2 spliceosome factor SF3B3 broadly desensitises cells to transcriptional co-activator inhibition, with short, exon-dense genes displaying the most prominent desensitisation. We show that splicing remains functional despite SF3B3 loss and that the desensitisation effect is mediated by the physical presence, but not the catalytic activity of the acetyltransferase and transcriptional co-activator P300. Mechanistically, SF3B3 depletion drives the redistribution of chromatinised P300 to newly licensed cis regulatory elements enriched for JUN/AP1; and perturbs the pause release and elongation dynamics of RNA Pol II. Moreover, the association of key transcriptional complexes, elongation factors and cyclin dependent kinases with RNA Pol II is reduced following SF3B3 depletion. Finally, using a CRISPR base editor screening approach, we performed tiled mutagenesis of P300 and discovered key residues in the histone acetyltransferase domain that either promote or prevent desensitisation to transcriptional coactivator inhibition. Together these data suggest a splicing-independent functional link between SF3B3, P300 and RNA Pol II required for the coordination and licensing of transcriptional elongation.
Abstract Title: Utilising CRISPR/Cas9 screening to uncover novel immunotherapies for targeting bone metastases
Authors: Thomas B. Chadwick1,2, Joan So1,2, Laura Vojtech1,2, Amina Ismail1,2, Lap Hing Chi1,2, Kaylene J. Simpson1,3,4, Marco J. Herold5,6, Belinda S. Parker1,2
Affiliations: 1 Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC 2 Cancer Evolution and Metastasis Program, Peter MacCallum Cancer Centre, Melbourne, VIC 3 Victorian Centre for Functional Genomics, Peter MacCallum Cancer Centre, Melbourne, VIC 4 Department of Biochemistry and Pharmacology, University of Melbourne, Parkville, VIC 5 Blood Cancer and Immunotherapy Lab, Genome Engineering and Cancer Modelling Program, Olivia Newton-John Cancer Research Institute, Heidelberg, VIC 6 School of Cancer Medicine, La Trobe University, Bundoora, VIC
Full Abstract: Despite improved detection and treatment of early-stage breast cancer, metastatic disease remains a clinical challenge as conventional and immune-based therapies offer little long-term survival benefit. About 75% of breast cancer metastases occur in bone, which can be treatment resistant and associated with severe morbidity and high mortality rates. The biological changes promoting outgrowth of disseminated cancer cells in bone are not fully understood, but one mechanism is microenvironment-induced immune suppression that renders bone metastases unresponsive to anti-tumour immunity. Our laboratory has previously identified a critical pathway associated with bone metastatic outgrowth and reduced cellular immunogenicity: suppression of tumour-intrinsic type I interferon (IFN) signalling, including hundreds of interferon-regulated genes crucial for innate and adaptive immune response and anti-tumour immunity. Thus, targeted approaches to restore IFN signalling in metastatic cells could have important therapeutic implications. This project used genome-wide CRISPR/Cas9 screening in bone metastatic lines in vitro to identify novel targets to restore IFN signalling. We transduced bone metastatic cells isolated from the syngeneic mouse models of breast cancer with an interferon-stimulated response element (ISRE)-GFP/Luc2 reporter system, which revealed significant reductions in ISRE activity both at baseline and with TLR-agonist stimulation compared to primary tumour lines. After Cas9 transduction, we performed a CRISPR/Cas9 knockout screen, and isolated GFP positive cells (indicating enhanced ISRE activity) via flow cytometry. This screen identified >100 genes that, when suppressed, restore type I IFN signalling, including confirmed targets relevant to the cGAS/STING signalling pathway. These key hits are being validated for their ability to alter IFN signalling and cellular immunogenicity in vitro and will be further validated for their effectiveness alongside traditional therapies in vivo. Identified through ISRE reporter and CRISPR/Cas9 screening, these gene targets offer novel therapeutic strategies for combatting late-stage breast cancer, enhancing IFN signalling in bone metastasis and increasing cancer vulnerability to immune-based therapeutics.
Abstract Title: Genome-wide CRISPR knockout screen to identify host factors in avian influenza virus infection
Authors: Debolina Majumdar1, Balaji Manicassamy2, Emily Hann1, Daniel Layton1, Arjun Challagulla1
Affiliations: 1 CSIRO Health and Biosecurity, Australian Centre for Disease Preparedness, Geelong, VIC 3220, Australia. 2 Department of Microbiology and Immunology, University of Iowa, Iowa City, lowa, USA
Full Abstract: Avian influenza (AI) poses a significant challenge to the global poultry industry, with current biosecurity measures and vaccination programs proving insufficient. Recent advances in gene editing technologies offer transformative potential by enabling complete resistance to infections within a single generation. However, successful implementation of gene editing relies on the identification of key host targets and molecular pathways that confer resistance to the diseases. Genome-wide CRISPR knockout (GeCKO) is a powerful tool that enables unprecedented opportunity to dissect the specific biological roles of genes in a massive-parallel approach under the influence of virus infection. Although numerous studies have identified host factors critical for influenza virus infection in human cells, comparable research in chicken cells remains unexplored, highlighting a significant gap in our understanding of host-pathogen interactions in avian species. In this study, we are conducting a GeCKO screen in chicken fibroblast DF1 cells to identify host factors required for AI infection. This will involve the development of a chicken GeCKO cell library, in which cells are targeted by gRNAs on a genome-wide scale. Subsequently, we will conduct an H7N7 AI infection survival screen on GeCKO library cells, performing multiple rounds of AI infection with virus-induced cell death serving as the readout. Following next-generation sequencing (NGS) and bioinformatics analyses, we will validate the highly enriched genes from the survival screen through a series of functional analyses. The outcomes of this GeCKO screening may provide new insights into the AI-host interface, enhance our understanding of host-directed strategies, and facilitate the discovery of novel gene-editing targets to mitigate AI in chickens.
Abstract Title: Promoting cell growth to prevent the spread of head and neck cancer
Authors: Yuchen Bai, Charbel Darido
Affiliations: Peter MacCallum Cancer Center
Full Abstract: Head and neck cancer (HNC) presents significant challenges in treatment due to its high metastatic propensity, leading to poor survival rates. 50% of patients develop metastasis within 9-month diagnosis, nearly all within 5-year treatment, reducing overall survival below 10%. Current therapies focus on rapidly dividing cancer cells but face limitations as metastasis-prone cells often adopt slower growth patterns, evading treatment. This resistance fuels cancer recurrence and complicates effective intervention, highlighting an urgent need for innovative therapies. A paradigm shift is essential to overcome these challenges and effectively target metastatic progression in HNC.
To comprehensively explore metastatic HNC, single-cell RNA sequencing data from patients' primary and nodally invasive tumors was analyzed. Trajectory analysis, based on cycling and EMT scores, elucidated molecular changes along metastatic progression. SCIENIC reconstructed gene regulatory networks during the dynamic transcriptional shifts, identifying key regulators responsible for the switch between proliferation and metastasis. A combinatorial genetic screen was applied in vivo to confirm the the genetic architecture that guides cellular decisions in metastatic HNC.
We confirmed the mutually exclusive expression of proliferation and metastasis cascades in the primary tumors and lymph nodes of HNC. Cells fell continuously along a gradient of proliferation to metastasis progression, revealing distinct waves of gene regulation. We incorporate a combinatorial perturbation sequencing into our trajectory analysis to confirm the dependency of the proliferative shut-down on progression along the EMT continuum. Combined genetic alterations can efficiently reverse the metastasis into proliferation, implying the close interaction between proliferative and metastatic cascades co-regulating cellular state of metastatic HNC.
Our study combines single-cell trajectory analysis with high-throughput genetic screening, which constitutes a powerful approach for identifying upstream signals of pathways that regulate cellular phenotypes. Indeed, these experiments uncover a combination of transcription factors that drive cells towards metastasis, explaining how cells switch between the discrete cellular states.
Abstract Title: Using genome-wide CRISPR-Cas9 knockout screens to sensitise metastatic breast cancer natural killer (NK) cell-mediated killing
Authors: Neil L1,2,, Liyanage C1,2, Brown R1,2, Srivaths A1,2, Vervoort S3, Yeo B4, Conor Kearney1,2,, Pal B1,2
Affiliations: 1 School of Cancer Medicine, La Trobe University, Bundoora, Victoria 3086, Australia 2Olivia Newton-John Cancer Research Institute, Heidelberg, Victoria 3084, Australia 3 The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia 4Austin Health, Heidelberg, Victoria 3084, Australia
Full Abstract: Distant metastasis is a major cause of breast cancer related mortality. Approximately one-third of patients diagnosed with early Triple-negative breast cancer (TNBC) subtype will develop distant metastases, who often face highly toxic and largely ineffective treatments. The median overall survival for metastatic TNBC patients is 11-13 months. Natural killer (NK) cells are recognised as the first line of defence against metastasis in solid tumours, including breast cancer. Harnessing NK cells to target metastatic TNBC presents a promising yet underexplored treatment avenue. We aim to develop NK cell-based immunotherapies targeting metastatic breast cancer and improve patient outcomes.
Our preliminary data on different breast cancer cell lines indicates that NK-cell mediated killing of breast cancer cells is independent of MHC-class I expression, opposing what is typically expected in the current literature. Therefore, we completed genome-wide CRISPR screens in a highly metastatic TNBC cell line. In this screen, we identified gene candidates whose activation or suppression could potentially sensitise cancer cells to NK cell-mediated killing. Here, we have prioritised two genes for inhibition: one gene known to promote cancer cell growth and immune evasion, and a target gene that is essential for mitosis, of which inhibition could induce immunogenic cell death via NK and T cells. We have validated these targets in vitro confirming increased NK cell-mediated killing of cancer cells and future studies will involve in vivo experiments to confirm their role in reducing metastatic burden.
Abstract Title: CRISPR in pest species Tribolium Castaneum
Authors: Daisy WILSON KOCHER, Dr. Charles Robin
Affiliations: The University of Melbourne
Full Abstract: The red flour beetle, Tribolium castaneum, is an emerging model organism. This species is also a major pest of stored grains that impacts farmer’s profits and food security globally. Agriculturalists have been using the fumigant phosphine to control T. castaneum infestations for decades. Unfortunately, resistance to phosphine has been recorded in many locations across sites world-wide. Compounding the problem, there is no alternative pesticide on the market that is as food-safe as phosphine. This project aims to use CRISPR-Cas9 technology to elucidate the identity / function of genes involved in phosphine resistance.
Gene knock-ins and gene knock-outs have both been performed in T. castaneum embryos using CRISPR-Cas9. In this study a family of Cytochrome-P450s (the CYP346B gene family) are targeted for gene knock-out because there is evidence that each of these genes are overexpressed in phosphine resistant T. castaneum These genes are adjacent to each other on chromosome 5 and arose from gene duplication, meaning that they can be targeted in tandem with CRISPR. Our design allows us to tease out the impacts of each gene on phosphine resistance phenotype. We have also performed a gene knock-out on Cytochrome-b5-fatty-acid-desaturase. This gene has been associated with phosphine resistance through variant linkage analysis. It is unclear whether a beetle homozygous for a loss-of-function alleles is viable, and our experiment will determine the answer. We also aim knock-in a wild-type copy of this gene to a phosphine resistant strain of T. castaneum in order to perform complementation tests. CRISPR is a tool that has and will continue to provide valuable insights into T. castaneum biology.
Abstract Title: Identifying Optimal Tumour-specific Promoters for CRISPR/Cas9 Engineering of Armoured CAR T Cells with Enhanced Safety and Efficacy
Authors: Kah Min Yap1,2, Amanda X. Y. Chen1,2, Imran G. House1,2, Phillip K. Darcy1,2 and Paul A. Beavis1,2
Affiliations: 1. Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia, 2. Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia
Full Abstract: Background: The tremendous success of chimeric antigen receptor-T (CAR-T) cell therapy in haematological malignancies has not been recapitulated in solid tumours, owing to tumour-induced immunosuppression, tumour heterogeneity and inefficient tumour trafficking. One promising solution includes “armouring” CAR-T cells with therapeutic transgenes. Indeed, we demonstrated that CAR-T cells engineered to express dendritic cell growth factor Flt3L could effectively engage host anti-tumour immunity crucial for overcoming antigen-negative relapse. However, synthetic promoters have demonstrated insufficiencies in driving tumour-restricted cytokine expression, which had caused systemic toxicities and trial termination. The advent of CRISPR/Cas9 gene-editing tool has enabled the precise engineering of CAR-T cells for safety and efficacy enhancements. We previously showed that CRISPR/Cas9-mediated knock-out (KO) of immunosuppressive gene A2AR enhanced CAR-T cell function. Now, we aim to exploit a CRISPR/Cas9-mediated knock-in (KI) strategy to leverage endogenous gene regulatory elements to restrict transgene expression to tumour for enhanced safety and efficacy.
Methods: Genome-wide RNA sequencing was performed on CAR-T cells isolated from tumours and spleens of mice. 27 genes upregulated in intratumoural relative to splenic CAR-T cells were identified as potential KI sites. As KI disrupts target gene expression, the impact of each gene KO on CAR-T cell function/phenotype was first assessed. 7 genes without adverse impact following KO had GFP knocked in.
Results: NR4A2 and RGS16 emerged as tumour-specific promoters upon KI. While NR4A2 was highly tumour-restricted and could deliver highly toxic cytokines (e.g., IL-12) without inducing toxicities in mice, RGS16 had high intratumoural expression and could mediate the efficacy of less potent cytokines (e.g., IL-2).
Conclusions: Endogenous tumour-specific promoters enabled the generation of IL-12- and IL-2-expressing CAR-T cells with enhanced safety and efficacy in syngeneic and xenogeneic mouse models that was concomitant with improved CAR-T cell polyfunctionality and activation of host anti-tumour immunity. Notably, this CRISPR-KI strategy was applicable using patient-derived CAR-T cells, demonstrating its clinical translatability.
Abstract Title: Expanding the molecular biology toolkit for paediatric cancer: generation of a comprehensive library of CRISPR/Cas cell lines
Authors: Alice Salib1, Teresa Sadras2,3, Fatimah Jalud2,3, Tasnia Ibnat2,3, Paul G Ekert1,2,4,5,6
Affiliations: 1. Translational Tumour Biology, Children’s Cancer Institute, Australia. 2. Peter MacCallum Cancer Centre, Australia 3. The Sir Peter MacCallum Department of Oncology, University of Melbourne, Australia 4. School of Clinical Medicine, University of New South Wales, Australia 5. University of New South Wales Centre for Childhood Cancer Research, University of New South Wales, Australia 6. Cancer Immunology Program, Peter MacCallum Cancer Centre, Australia
Full Abstract: Personalised medicine initiatives like the Zero Childhood Cancer Program have highlighted the genomic heterogeneity of paediatric cancers, and the value of molecular characterisation to deliver more effective treatments. As novel variants are identified, there is a growing need to characterise their functions and therapeutic targetability. To address this, we are developing a comprehensive library of CRISPR/Cas-engineered isogenic paediatric cancer cell lines, enabling manipulation of gene expression through knockout (CRISPR/Cas9), knockdown (CRISPR/Cas13), or overexpression (CRISPRa) strategies. A nuclease-dead Cas13d variant (dCas13d) serves as a control. Leveraging these tools, we aim to functionally characterise candidate driver genes and novel variants in paediatric malignancies.
To date, we have generated 22 CRISPR/Cas cell lines, encompassing a wide range of paediatric cancers including neuroblastoma, medulloblastoma, rhabdomyosarcoma, osteosarcoma, and Ewing's sarcoma. Across the CRISPR/Cas13d lines generated, we observe variable doxycycline (dox)-induced Cas13d expression. Further, our findings suggest guide expression levels may limit gene silencing efficiency. Clonogenic assays revealed a dose-dependent reduction in colony-forming capacity upon Cas13d induction, independent of guide expression. Our results indicate that careful control of both guide and Cas13d expression is needed and strategies to mitigate Cas13d toxicity while enhancing guide efficacy are in progress.
In parallel, we are using CRISPR/Cas9 cell lines we have generated in combination with a unique pipeline we have developed to generate PDGFRA structural variants by targeting intronic regions adjacent to the deleted exons. This approach enables us to recreate these PDGFRA variants endogenously to further characterise their function.
In summary, the cell lines developed offer a valuable resource for investigating the molecular mechanisms driving paediatric cancers. Furthermore, they serve as a toolkit for identifying and exploring potential therapeutic vulnerabilities in paediatric cancer to select targeted and more effective treatments for patients.
Abstract Title: Reactivation of δ-globin via CRISPR in treating β-haemoglobinopathies
Authors: Wing Fuk Chan, James J. J. The, Bradon Rumler, Stephanie Anderson, Huimin Cai, Zane S. Kaplan, Andrew C. Perkins
Affiliations: 1Australian Centre for Blood Diseases, Monash University
Full Abstract: β-haemoglobinopathies are inherited blood disorders that affect haemoglobin in erythrocytes, with hundreds of thousands of affected new-borns each year. While sickle cell disease results from a single-nucleotide substitution, β-thalassaemia is caused by hundreds of different mutations, making the direct repair of mutations challenging. As such, a universal therapeutic approach is widely sought.
Much of the effort has been put into reactivating the foetal γ-globin by various means in the adult cells. As an alternative, δ-globin is expressed in adult red blood cells and shares a much higher sequence homology to β-globin. As such, δ-globin, and the resultant haemoglobin A2 (HbA2), is biochemically more similar and fully functional as the conventional adult haemoglobin HbA. However, δ-globin is expressed at a low level and HbA2 only constitutes 2-3% of the total adult haemoglobin. The low expression is believed to be due to mutations at the promoter that disrupt transcription factor binding. Nevertheless, overexpression of δ-globin transgene is able to rescue the sickling phenotype of red cell in a mouse model. As such, we propose reactivating the δ-globin by editing the promoter as effective alternative therapeutics of SCD and β-thalassaemia.
In this project we employed CRISPR gene editing techniques to install an KLF1 motif at the δ-globin promoter in the immortalised HUDEP-2 cell line. Through RNP transfection along with an HDR repair template, we were able to drive the expression of δ-globin to ~160 fold in the HUDEP-2 cells compared to the control. The upregulated δ-globin transcript level is around 60% of that of β-globin. Upon differentiation, we observed the HbA2 level is upregulated to 20% by HPLC. Using NGS, we demonstrated the editing is highly efficient with over 50% HDR events. Lastly, we showed that this upregulation of δ-globin is highly dependent on the insertion site at the promoter.
Abstract Title: Genome-wide CRISPR Screen Identify Menin as a Mediator of Encorafenib Plus Cetuximab Resistance in BRAF V600E Mutant Colorectal Cancer
Authors: Akash Srivaths (1), Tirta Mario Djajawi (1), Kristen Needham (1), Chloe Gerak (2), Liam Neil (1), Sarahi Mendoza Rivera (2), Bhupinder Pal (1), Stephin Vervoort (2), John Mariadason (1), Conor Kearney (1)
Affiliations: 1. Olivia Newton-John Cancer Research Institute, 2. Walter and Eliza Hall Institute of Medical Research
Full Abstract: Colorectal cancer is the second most commonly diagnosed cancer and one of the leading causes of cancer-related deaths in Australia. Mutations in the BRAF gene are frequently detected in several cancers including colorectal cancer. Up to one in five patients with metastatic colorectal cancer have a BRAF mutation, with the V600E substitution being the most prevalent. These patients generally have a poor prognosis. A combination of encorafenib plus cetuximab has significantly improved the overall survival of such patients, compared to standard chemotherapy regimens in clinical trials, establishing it as the new standard of care for colorectal cancer with BRAF V600E mutation. While this treatment strategy is promising, it does not eradicate the tumor completely as tumor-intrinsic resistance poses a challenge. We performed whole-genome CRISPR screens to identify such resistance-causing genes in response to encorafenib plus cetuximab. We identified that MEN1 (Menin) confers resistance to encorafenib plus cetuximab treatment in colorectal cancer with BRAF V600E mutation. Indeed, MEN1 knockout, or treatment with revumenib, a small-molecule inhibitor of the Menin-MLL interaction, augmented the cytostatic effect of encorafenib plus cetuximab in BRAF V600E mutant colorectal cancer cells, whereas re-expression of MEN1 into MEN1-null cells reversed this phenotype. We further demonstrated that the protective role of menin is dependent on its interaction with MLL1, a histone methyltransferase. Taken together, we identify Menin as a key factor in driving encorafenib plus cetuximab resistance, providing rational for targeting of Menin in combination with encorafenib plus cetuximab for improved treatment of colorectal cancers with BRAF V600E mutation.
Abstract Title: High depth broad scale CRISPR screening to identify fundamental synthetic lethal targets to P300/CBP inhibition
Authors: Oliver Sinclair, Jesse Balic, Tim Somervaille, Mark Dawson
Affiliations: Peter MacCallum Cancer Centre, Cancer Research UK Manchester Institute
Full Abstract: Multiple myeloma (MM) and acute myeloid leukaemia (AML) are distinct haematopoietic malignancies which together provide a large unmet clinical need. While both are discrete they progress stepwise through premalignant states, and share dependency on aberrant transcriptional programmes that block differentiation. This dependency renders both diseases susceptible to inhibition of core transcriptional machinery, including the transcriptional co-activators p300 and CBP. These highly homologous acetyltransferases occupy (super)enhancer and promoter cis-regulatory elements to critically govern cell-type and disease-specific gene regulatory networks. In MM, p300/CBP activity sustains elevated IRF4 levels, driving proliferation and survival. In AML, p300/CBP supports MYC expression, promoting rapid growth.
This work investigates CCS1477, a novel p300/CBP bromodomain inhibitor in clinical evaluation. While early trials report responses in 67% of MM patients, relapse and resistance remain significant barriers. To address these challenges, we performed unbiased CRISPR screening across AML and MM cell lines to identify synthetic lethal targets and resistance mechanisms.
CRISPR screens were conducted in five cell lines across two doses and timepoints, with the hope of identifying fundamental sensitising hits to p300/CBP inhibition. Hits were ranked by average RRA scores, revealing the metazoan INO80 chromatin remodelling complex as a key sensitising target. Validation in both disease states and in screen-naïve cell lines confirmed these hits sensitize cells to CCS1477 and to alternative p300/CBP inhibitors targeting histone acetyltransferase activity.
Our findings establish the INO80 complex as a critical regulator of p300/CBP-driven transcription and a disease, cell line and inhibitor agnostic synthetic lethal target to enhance efficacy. These hits would be overlooked in standard individual screening approaches. Therefore, this study presents a robust CRISPR screening approach to prioritise synthetic lethal targets which outperforms standard single cell line approaches.
Abstract Title: Identifying New Roles For Ubiquitination Machinery In Regulating Dendritic Cell Function
Authors: Huw Morgan1, Haiyin Liu1, Laura E Edgington-Mitchell1, Justine D Mintern1
Affiliations: 1Department of Biochemistry and Pharmacology, The Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Australia
Full Abstract: Ubiquitination is a post-translational modification that regulates protein degradation and activity, such as in regulating inflammatory signalling or protein surface expression. A cascade of enzymes carries out ubiquitination of proteins. One key player is the E3 ubiquitin (Ub) ligases, which are involved in the addition of ubiquitin. This project aims to unveil novel roles for these enzymes in immunity, focusing on dendritic cells (DCs). DCs are antigen-presenting cells specialised in regulating T cells during the adaptive immune response, maintaining the balance between health and disease. Of the over 600 known and putative E3 Ub ligases, only a handful have a described role in DC function. This project aims to identify other E3 Ub ligases that participate in the regulation of DCs.
To identify E3 Ub ligases that participate in DC function, we employed an arrayed CRISPR/Cas9 screen of all known and putative E3 Ub ligases and scaffold proteins in the mutuDC line. The expression of surface markers involved in DC function were analysed by spectral flow cytometry to assess the impact of gene knockout on DCs. This screen identified Cul3 as a new regulator of DC function. The screen results were confirmed in bone marrow-derived dendritic cells (BMDCs) differentiated from gene-edited Cas9 transgenic haematopoietic progenitors. Knockout of Cul3 upregulated the expression of co-stimulatory molecules CD80 and CD86 in unstimulated BMDCs and upregulated the expression of the inhibitory molecule PD-L2 following TLR3 and TLR9 stimulation, relative to wild-type BMDCs.
Using a functional genomics approach, we have identified new roles for ubiquitination machinery in regulating the function of DCs. We identified a new role for Cul3, a gene with no known role in DC function. Further exploration of these pathways will gain insight into the complex regulatory network of DCs and highlight new ways to manipulate DC function in health and disease.
Abstract Title: Tumour-site directed expression of adenosine receptor subtype 1 (A1R) enhances CAR T cell function and improves efficacy against solid tumours
Authors: Kevin Sek (1,2), Amanda Chen (1,2), Thomas Cole (1,2), Philip K. Darcy (1,2,6), Paul A. Beavis (1,2,6)
Affiliations: (1) Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, 3000, Victoria, Australia, (2) Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, 3010, Australia, (3) Department of Pathology, University of Melbourne, Parkville.
Full Abstract: The efficacy of Chimeric Antigen Receptor (CAR) T cells against solid tumours is limited by immunosuppressive factors including adenosine, which suppresses CAR T cells through activation of the A2A receptor (A2AR). Deletion of the A2AR with CRISPR/Cas9 significantly enhances the in vivo efficacy of both mouse and human CAR T cells. Alternatively, CAR T cells were engineered to express A1R, a receptor that signals inversely to A2AR. Using murine and human CAR T cells, constitutive A1R overexpression was demonstrated to significantly enhanced CAR T cell effector function but at the expense of CAR T cell persistence. Through a novel CRISPR/Cas9 “knock-in” approach CAR T cells were engineered to express A1R upon CAR T cell activation, which led to enhanced anti-tumour efficacy concomitant with improved long-term persistence and was dependent on the transcription factor IRF8. These data provide a novel approach for enhancing CAR T cell efficacy in solid tumours and provides proof of principle for site-directed expression of factors that promote effector T cell differentiation.
Abstract Title: Improving neuroblastoma response to BH3-mimetic drugs with CRISPRi
Authors: Ziyan Liu, Shuai Huang, Grant Dewson
Affiliations: WEHI
Full Abstract: High-risk neuroblastoma patients face a poor prognosis and limited response to chemotherapy, with 50% of children relapse following aggressive chemotherapy regimens, and median overall survival after relapse is 11 months. Hence there is critical need to identify more effective therapeutic targets with limited toxicities. Targeted BH3 mimetic drugs such as VENCLEXTA®/Venetoclax/ABT-199 are changing the treatment landscape for certain blood cancers, but their use in neuroblastoma has been hindered by resistance. To address this challenge, this study leverages CRISPRi screening technology to identify targets that can sensitize neuroblastoma to venetoclax treatment and overcome resistance.
Investigating these targets and utilizing the ubiquitin system could uncover synergistic pathways, enhancing venetoclax efficacy in neuroblastoma. By elucidating the underlying molecular mechanisms, this project aims to advance targeted therapeutic strategies and improve outcomes for neuroblastoma patients.
Abstract Title: Utilising CRISPR technologies and high-throughput screening to interrogate the biology of secreted factors: a CXCL9 and CXCL10 case study.
Authors: Emily B. Derrick, Phillip K. Darcy, Paul A. Beavis
Affiliations: (1) Peter MacCallum Cancer Centre, (2) The Sir Peter MacCallum Department of Oncology, University of Melbourne
Full Abstract: Immune checkpoint blockade (ICB) has revolutionised the treatment of numerous cancer types, including melanoma and non-small cell lung carcinoma. ICB targets immune-inhibitory molecules on the surface of T cells, unleashing their anti-tumour potential. Despite ICB’s success, a high frequency of patients fail to respond to this therapy. A key limiting factor to ICB responses is the number of T cells that infiltrate the tumour microenvironment. T cell infiltration in the context of ICB has been shown to be dependent on chemoattractant molecules CXCL9 and CXCL10. We have previously demonstrated that these chemokines are predominantly produced by intratumoral macrophages. Therefore, we aimed to identify strategies to enhance CXCL9/10 production in macrophages and subsequently improve T cell infiltration in solid tumours. To screen for secreted factors, we utilised CRISPR/Cas9-mediated homology directed repair (HDR) to generate a cell line that expressed GFP and BFP as a bona fide readout of CXCL9/10 production. Using this cell line, we performed a whole-genome CRISPR/Cas9 screen to identify genetic regulators of chemokine production. We successfully identified numerous regulators, including PTPN2 as a negative regulator of CXCL9/10 in macrophages. Using the same cell line, we performed a flow cytometry-based drug screen to identify novel compounds that induce CXCL9/10 expression. From a novel library containing 20000 compounds, several drug candidates improved chemokine expression and are currently being validated. By utilising CRISPR-HDR to generate a reporter cell line, we were able to establish robust platforms to interrogate the biology of secreted factors, through both whole-genome CRISPR/Cas9 screening and flow cytometry-based drug screening. This work has identified new therapeutic strategies that improve CXCL9/10 expression, which can be utilised in combination with ICB to improve clinical outcomes for patients.
Abstract Title: Screening for novel regulators of GATA3 expression
Authors: Serena R. Kane1,2, Milevskiy MJG1,2, Fu N1,2, Lindeman G1,2,3,4, and Visvader JE1,2
Affiliations: 1 ACRF Cancer Biology and Stem Cells Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia. 2 Department of Medical Biology, The University of Melbourne, Parkville, VIC 3010, Australia. 3 Department of Medicine, The University of Melbourne, Parkville, VIC 3010, Australia. 4 Parkville Familial Cancer Centre and Department of Medical Oncology, The Royal Melbourne Hospital and Peter MacCallum Cancer Centre, Parkville, VIC 3052, Australia. 5 Duke-NUS Medical School, Singapore 169857.
Full Abstract: The GATA3 gene is vital for mammary gland development and plays important roles in breast cancer. Breast cancers with high GATA3 expression are associated with better clinical outcomes and improved response to endocrine therapy compared to GATA3 low tumours. Increasing GATA3 expression may be clinically favourable in breast cancer. I aimed to identify novel regulators of GATA3 through whole genome CRISPR screening using GATA3 reporter breast cancer cell lines. Three reporter cell lines were established by inserting a fluorescent reporter at the end of the natural GATA3 gene. I performed whole genome CRISPR knock-out screens with these reporter cell lines, sorting the top and bottom 10% of cells based on reporter fluorescence. Multiple candidates were investigated, with many knockouts not altering GATA3 protein expression. A putative positive regulator of GATA3 expression was identified and validated in multiple cell lines. I then investigated other members of the complex that this candidate acts through. Knocking out other genes from this complex also decreased GATA3 expression. These results suggest that we have identified a novel mechanism that positively regulates GATA3 expression.
Abstract Title: Genome-wide CRISPR-Cas9 Screens on Tumour Cells under Vδ2+ γδ T Cell Immune Pressure
Authors: Andrew Li (1), Conor J Kearney (1), Lisa A Mielke (1), Stephin J Vervoort (2), Andreas Behren (1,3), Kok Fei Chan (1)
Affiliations: (1) Olivia Newton-John Cancer Research Institute, and School of Cancer Medicine, La Trobe University, Heidelberg, Victoria, Australia (2) The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia (3) Department of Medicine, University of Melbourne, Parkville, Victoria, Australia
Full Abstract: Gamma-Delta (γδ) T cells are a non-conventional subset of T cells which are able to recognize tumour cells independent of human leukocyte antigen (HLA) molecules. The predominant Vδ2+ subset residing in the peripheral blood has been found to be activated through recognition of a complex formed by BTN2A1/3A1 molecules, which upregulate in response to endogenous metabolic dysfunction. Previous studies have identified an anti-tumour role for Vδ2+ γδ T cells, alongside antigen presenting and immunomodulatory capabilities. Considerable interest has therefore been generated for their application in tumour immunotherapy treatments such as adoptive cell transfer, as the recognition of conserved moieties allows for allogenic transfer with minimal risk of graft-versus-host disease (GvHD). However, while treatments involving γδ T cells have shown prior success against haematological cancers, durable patient responses have yet to be achieved against solid malignancies including melanoma. To better elucidate the potential of Vδ2+ γδ T cells in adoptive cell therapy, we have employed the use of advance whole-genome CRISPR-Cas9 screens to target individual gene knockouts in human melanoma cells across >19000 genes. These screens investigate the mechanisms that allow melanoma cell survival despite immune pressure from Vδ2+ γδ T cells. Here we identify gene candidates that may contribute to tumour immune evasion from Vδ2+ γδ T cell-mediated killing which require further investigation.
Abstract Title: Modelling BRPF1-associated neurodevelopmental disorders
Authors: Sophie TB Morgan, Hannah K Vanyai and Marnie E Blewitt
Affiliations: Walter Eliza Hall Institute of Medical Research, The University of Melbourne
Full Abstract: Bromodomain and PHD finger containing protein 1 (BRPF1) is a chromatin reader which complexes with histone acetyltransferases KAT6A and KAT6B. The BRPF1 complex plays essential roles in neurodevelopment, with heterozygous BRPF1 loss leading to neurodevelopmental disorders (NDDs), primarily the BRPF1 haploinsufficiency disorder Intellectual Developmental Disorder with Dysmorphic Facies and Ptosis (IDDDFP). Indeed, heterozygous loss of KAT6A and KAT6B also leads to NDD’s. These disorders are characterised by intellectual disability and global developmental delay.
The molecular mechanism of BRPF1, KAT6A and KAT6B has not been thoroughly investigated owing to issues in reliable detection methods and low endogenous expression. Using CRISPR-Cas9 editing via ribonucleoprotein nucleofection, we have successfully HiBiT tagged BRPF1, KAT6A and KAT6B, as well as BRPF family members BRPF2/BRD1 and BRPF3 in human neuroblastoma SH-SY5Y cells. Furthermore, we have introduced IDDDFP patient pathogenic variants into HiBiT-BRPF1 to directly investigate the molecular consequences of different variants.
For the first time, we have identified the genomic binding targets of BRPF1 through Cut&Run using the HiBiT antibody, and will extend this to the other targets. Additionally, using the HiBiT toolkit, we have detected BRPF1 and complex members via immunofluorescence microscopy, on-blot luciferase assay and luminescence plate-based methods. Furthermore, experiments carried out in BRPF1 patient pathogenic variant lines support that IDDDFP is a BRPF1 haploinsufficiency disorder, with patient variants resulting in a loss of function.
Abstract Title: Developing CRISPR-Cas13 antiviral therapeutics for respiratory pathogens of pandemic potential
Authors: Moe EI Mohamad1,2,3, Wei Zhao1, Rob de Rose1,4, Stanislav Kan1, Wenxin Hu2,3, Joshua Casan2,3, Honglin Chen2,3, Paula Cevaal1, Ajantha Rhodes1, Danielle Fong1, Nadia Saraya1, Michael Roche1, Damian Purcell1, Joseph A Trapani2,3, Matthew McKay5, Mohamed Fareh2,3, Sharon R Lewin1,7
Affiliations: 1 The Peter Doherty Institute for Infection and Immunity, The University of Melbourne and Royal Melbourne Hospital, Melbourne, Australia 2 Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Australia 3 Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Australia 4 Department of Chemical Engineering, The University of Melbourne, Melbourne, Australia 5 Department of Electrical and Electronic Engineering, The University of Melbourne, Melbourne, Australia 6 Murdoch Children’s Research Institute, Royal Children's Hospital, Melbourne, Australia 7 Department of Infectious Diseases, Alfred Hospital and Monash University, Melbourne, Australia
Full Abstract: There is an urgent need for effective antiviral therapies, especially against respiratory viruses of pandemic potential. We previously showed that CRISPR-Cas13 inhibits SARS-CoV-2 replication in vitro. The study aimed to test CRISPR-Cas13 delivery as mRNA in lung-targeting lipid nanoparticles (LNP) intravenously and intranasally. We used a novel LNP formulation, LNP-X, to optimize CRISPR Cas13b delivery to Vero-GFP cells. We co-encapsulated mRNA encoding for Cas13b together with a guide RNA (crRNA) targeting the coding sequence in the gene for GFP, and achieved a 90% reduction in GFP expression with 250 ng mRNA (n=3). We then delivered mRNA encoding for Cas13b with a crRNA targeting the nucleocapsid gene in SARS-CoV2 using LNP-X. Following 24 hours of mRNA delivery to Vero cells, the cells were infected with either the ancestral SARS-CoV2 or Omicron BA.1. After 24 hours, we observed a 3.8 and 2.4-log reduction in infectious virus in supernatant respectively, compared to an identical LNP which carried a non-targeting crRNA (negative control). We also tested LNP-X with and without modifications in Calu-3 cells grown in air-liquid interface (ALI) cultures. We encapsulated the LNPs with a nanoluciferase tagged-CRISPR Cas13b mRNA (Cas13b-NanoLuc) and transfected the cells with 1000 ng mRNA. One LNP induced a 3-fold increase in Nanoluc expression compared to the other LNPs. For in vivo delivery, we encapsulated Cas13b-NanoLuc mRNA in an LNP that contains the cationic lipid DOTAP, known to target the lungs following intravenous administration. After administering 15 µg Cas13b-Nanoluc intravenously, lung Nanoluc expression was 9-fold higher than in the spleen and liver. Cas13b can be delivered as mRNA in vitro in cell lines and organoid, and in vivo using novel LNPs. In vitro, Cas13b mRNA and crRNA targeting nucleocapsid can potently inhibit SARS CoV2 replication. This approach could be a promising platform for therapeutics for respiratory pathogens of pandemic potential.
Abstract Title: Harnessing Cas13 to target Multiple Myeloma
Authors: Joshua King, Joe Trapani, Ricky Johnstone, Mohamed Fareh
Affiliations: Peter MacCallum Cancer Centre, The University of Melbourne
Full Abstract: Cas13 is a Cas nuclease with RNase activity. It utilises a guide RNA (gRNA) to complimentarily base-pair, and subsequently cleave and degrade target RNA. Numerous groups have co-opted Cas13 for highly selective degradation of RNA molecules in a variety of cell lines. There has also been significant interest into harnessing Cas13 as a personalised therapeutic. By contrast, a number of groups have reported that certain Cas13 orthologues, upon cleaving target RNA, proceed to indiscriminately degrade bystander RNA, thereby adversely affecting host cell viability. It has been postulated that this collateral activity may be harnessed for selective cell elimination. We, and others, have therefore proposed that Cas13 may represent a promising Oncology therapeutic. We aim to exploit Cas13 to i) silence undruggable oncogenic RNA molecules and ii) induce of transcriptome destruction in selective cancer cells.
Multiple Myeloma (MM) is the most common haematological cancer in Australia. Approximately 15% of MM patients exhibit a translocation of chromosome 4 and 14 (t(4;14)) that leads to the overexpression of the epigenetic regulator NSD2. This enzyme drives global epigenetic rewiring and dysregulation of a plethora of oncogenic pathways implicated in MM pathogenesis. There are currently no approved therapeutics targeting NSD2 available to MM t(4;14) patients.
This project aims to characterise and exploit RfxCas13d activity for i) programmed on-target cleavage of RNA, and ii) collateral activity in human, multiple myeloma cell lines (HMCLs). We first seek to elucidate the determinants of RfxCas13d activity, and how it impacts HMCLs. We then aim to program RfxCas13d to target IgH-NSD2 fusion transcripts to drive selective death of t(4;14) HMCLs. Finally, we aim to develop lipid nanoparticles to deliver RfxCas13d and personalised gRNAs to patient-derived MM cells as a proof of principle therapeutic.
Talk Title: CRISPR engineering of armored CAR T cells enables tumor restricted payload delivery with enhanced efficacy and safety
Abstract Title: TAK1 protects tumour cells from combined CTL-derived TNF and IFN-y
Authors: Tirta M. Djajawi, Anne Huber, Akash Srivaths, Oliver Ozaydin, Sarahi Mendoza Rivera, Stephin J. Vervoort and Conor J. Kearney
Affiliations: Olivia Newton-John Cancer Research Institute, Melbourne, VIC
Full Abstract: Cancer immunotherapies, particularly immune checkpoint blockade, have demonstrated remarkable efficacy in some cancer types including melanoma. However, regardless of the clinical success of checkpoint inhibitor therapies, many patients do not respond or develop resistance, leading to relapse. Whereas some mechanisms of tumour immune escape are understood, the full spectrum of immune evasion routes are not well understood. Given the issue of high non-response rates and patient relapse, there is an urgent need to molecularly investigate tumour immune evasion tactics in order to develop improved immunotherapy approaches.
Here, we conducted a kinome-wide CRISPR-Cas9 screen in melanoma cells to identify factors that limit CD8+ T cell-mediated anti-tumour immunity. We identified that TAK1 suppresses combined CTL-derived IFNy- and TNF-induced cell death, thus protecting from CD8+ T cell-mediated killing. Indeed, TAK1 knockout or pharmacological targeting of TAK1 with the clinical inhibitor Takinib enhances CD8+ T cell-mediated cancer cell killing in a TNF and IFNy-dependent manner, while re-introduction of TAK1 in TAK1-deficient cells reverses this effect. Furthermore, through genome-wide CRISPR screening in a TAK1 deficient background, we identified STAT1, RIPK1 and Caspase-8 as key effectors of the combined TNF and IFNy-induced cell death. Taken together, we identify TAK1 as a negative regulator of anti-tumour immunity, unveiling TAK1 targeting as a novel immunotherapeutic approach or as an adjunct to existing immunotherapies.
Abstract Title: Generating CRISPR/CAS9 armoured TCR-T cells for the treatment of solid tumours
Authors: Phoebe Dunbar (1, 2), Amanda Chen (1, 2), Kah Min Yap (1, 2), Phil Darcy (1, 2), Paul Beavis (1, 2)
Affiliations: 1. Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, 3000, Victoria, Australia. 2. Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, 3010, Australia
Full Abstract: CAR T cell therapy has exhibited remarkable clinical success in the treatment of haematological malignancies, however, their efficacy in solid tumours is limited by antigen heterogeneity and immunosuppression imposed by the tumour microenvironment. To overcome these barriers, ‘armoured’ CAR T cells which secrete pro-inflammatory cytokines have been developed. However, toxicities related to the unrestricted expression of the armouring transgene has limited the application of these CAR T cells in the clinic. Our lab has developed a novel CRISPR/Cas9-mediated homology directed repair (HDR) strategy whereby we aimed to engage endogenous gene regulatory mechanisms drive transgene expression in a tumour-localised manner (in review, Nature). Genome-wide RNA-sequencing was used to identify genes in CAR T cells with tumour-specific expression. The promotor NR4A2 was identified as a key candidate due to its inhibitory role in T cells. This novel CRISPR HDR strategy was employed to knock in proinflammatory cytokine, IL-12 into NR4A2 promoter which supports the delivery of cytokines directly to the tumour site, leading to enhanced anti-tumour efficacy and long-term survival of mice in both syngeneic and xenogeneic models.
Although we have developed a novel technology that tightly restricts expression of pro-inflammatory cytokines to T cell activation, the application of this technology in a CAR T setting is limited by on-target toxicity in healthy cells where tumour antigen is expressed. I therefore hypothesise that the application of tumour neoantigen specific TCR-T cells would provide an even greater safety profile for this technology. I have generated T cells expressing an engineered TCR targeting the tumour neoantigen KRAS G12D and subsequently verified that the NR4A2 promotor effectively turns on within this neoantigen specific TCR-T cell setting. I have further engineered TCR-T cells to express an NR4A2/IL-12 knock in, which elicits improved anti-tumour efficacy and an improved safety profile in xenogeneic mouse models.
Abstract Title: Robust Genomewide Application of Deactivated CRISPR-Cas9 Systems for Expression Regulation
Authors: Emily Anderson1, Clarence Mills1, Joanna Gawden-Bone2, Jera Law1, Abhijit Patil2, Matthew Brockman3, Brian Ziemba1, Andrew Riching1, Kevin Hemphill1, Simon Scrace2, Zaklina Strezoska1, and Josien Levenga1
Affiliations: 1. Revvity, Lafayette, Colorado, United States 2. Revvity, Cambridge, United Kingdom 3. Revvity, Mulgrave, Australia
Full Abstract: CRISPR-Cas9 has been engineered for modulating transcription and epigenetics; CRISPR activation (CRISPRa) and CRISPR interference (CRISPRi) systems act via a nuclease-deactivated Cas9 (dCas9) fused to modifiers and targeted to promoters by gene-specific synthetic guide RNAs (gRNAs). We have optimized CRISPR-based modulation for use in primary and difficult cells (requiring chemically synthesized gRNAs transiently delivered with dCas9 mRNA or ribonucleoprotein (RNP)). Synthetic gRNA delivery was also tested for streamlined high-throughput arrayed screening using stably-expressing Cas9 cell lines. Finally, high-titer All-in-one dCas9-effector lentiviral systems were produced allowing up to full genome pooled screens.
We demonstrate a simple, robust CRISPRa approach compatible with dCas9 fused to the VP64-p65-Rta tripartite activator (dCas9-VPR). Endogenous activation can be tuned/enhanced using multiple chemically modified synthetic gRNAs. Using the All-in-one dCas9-VPR lentiviral system, we have conducted a whole-genome CRISPRa screen, which faithfully recapitulates data from prior drug-resistance screening using the complex, three-vector Synergistic Activation Mediator (SAM).
The Dharmacon CRISPRi system is more efficient than first-generation CRISPRi. This novel dCas9 fusion uses domains from transcriptional repressors Sal-like protein 1 (SALL1)/Sin3 histone deacetylase corepressor complex component (SDS3). An unbiased protein interaction experiment demonstrates dCas9-SALL1-SDS3 interacts with key members of the histone deacetylase and Swi-independent 3 complexes (endogenous functional effectors of SALL1 and SDS3). dCas9-SALL1-SDS3 with chemically synthesized gRNAs can be used orthogonally to siRNA to verify downstream phenotypes. Finally, we performed the first genome-wide pooled screen with single lentiviral vector (All-in-one) dCas9-SALL1-SDS3 CRISPRi and observed enhanced essential gene dropout compared to first-generation single-vector dCas9-KRAB CRISPRi.
These CRISPR modulation systems were optimized for short-term, non-viral delivery or for pooled screening with high-titer lentiviral libraries. These formats enable simple, robust, and rapid gain- or loss-of-function experiments. Complex end-point assays are now accessible, and scale up to high throughput arrayed or pooled screening platforms is possible for multiple compounds, hypotheses, or cell types.
Talk Title: TBA
Abstract Title: Cas9-NG: a new vision for autosomal dominant Retinitis Pigmentosa precision therapeutics
Authors: Ashleigh B Geiger (1,2), Fatwa Adikusuma (1,2), Laurence OW Wilson (3), Louise J Robertson (1,2), John PM Wood (1), Robert J Casson (1), Paul Q Thomas (1,2).
Affiliations: 1. Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, Australia. 2. South Australian Health and Medical Research Institute Gene Editing Program, Adelaide, Australia. 3. CSIRO Digital Genome Engineering, New South Wales, Australia.
Full Abstract: Autosomal dominant Retinitis Pigmentosa (adRP) is an important cause of progressive, irreversible blindness. At least 12% of all non-syndromic adRP is due to the Pro23His mutation in Rhodopsin (RHO), a photoreceptor-specific GPCR that is essential for retinal function and health. The Pro23His mutation operates via dominant-negative and toxic gain-of-function mechanisms, rendering gene replacement strategies non-viable. Conversely, as RHO is partially haplosufficient, selective ablation of the mutant allele using CRISPR/Cas9 technology represents a tantalising prospect for treatment. However, guide RNA (gRNA) design is heavily restricted due to the unavailability of canonical SpCas9 5’-NGG-3’ PAM sequences at this locus. In this study, we leveraged alternative CRISPR/Cas9 platforms to target the RHO Pro23His mutation in vitro and in vivo. To initially assess disease-allele targeting activity, we generated a genetic cell model using Prime Editing. Candidate gRNAs were tested, and deep sequencing identified several which displayed highly efficient and selective targeting of the mutant allele. Encouraged, we tested our most promising candidates in a humanised preclinical RHO Pro23His mouse model, delivering AAV-CRISPR via intravitreal injection. 70 days post-injection, we assessed AAV-CRISPR delivery success via immunofluorescence, identifying robust evidence of Cas9 transduction in target cells of treated retinae. Excitingly, phenotype testing via dark-adapted electroretinogram revealed a treatment group with significantly improved photoreceptor activity compared with untreated controls, suggesting sustained retinal neuroprotection. To our surprise, this treatment utilised the engineered CRISPR variant Cas9-NG, which is currently absent from the CRISPR therapy literature due to perceived inefficiency and non-specificity. Importantly, these results are accompanied by molecular-level evidence of Cas9-NG activity against the human RHO Pro23His allele in treated murine retinae. Together, these data provide preclinical evidence that potentiates our allele-specific targeting approach as a candidate treatment for RHO Pro23His adRP, and expands the CRISPR therapeutic toolbox via the novel application of Cas9-NG.
Talk Title: TBA
Olivia Newton-John Cancer Research Institute
Level 5, 145 Studley Road, Heidelberg, Victoria 3084, Australia
Copyright © 2023 Olivia Newton-John Cancer Research Institute -
All Rights Reserved.