Centre for Cancer Research - Research Publications

Permanent URI for this collection

Search Results

Now showing 1 - 5 of 5
  • Item
    No Preview Available
    Inhibition of METTL3 Results in a Cell-Intrinsic Interferon Response That Enhances Antitumor Immunity
    Guirguis, AA ; Ofir-Rosenfeld, Y ; Knezevic, K ; Blackaby, W ; Hardick, D ; Chan, Y-C ; Motazedian, A ; Gillespie, A ; Vassiliadis, D ; Lam, EYN ; Tran, K ; Andrews, B ; Harbour, ME ; Vasiliauskaite, L ; Saunders, CJ ; Tsagkogeorga, G ; Azevedo, A ; Obacz, J ; Pilka, ES ; Carkill, M ; Macpherson, L ; Wainwright, EN ; Liddicoat, B ; Blyth, BJ ; Albertella, MR ; Rausch, O ; Dawson, MA (AMER ASSOC CANCER RESEARCH, 2023-10-05)
    UNLABELLED: Therapies that enhance antitumor immunity have altered the natural history of many cancers. Consequently, leveraging nonoverlapping mechanisms to increase immunogenicity of cancer cells remains a priority. Using a novel enzymatic inhibitor of the RNA methyl-transferase METTL3, we demonstrate a global decrease in N6-methyladenosine (m6A) results in double-stranded RNA (dsRNA) formation and a profound cell-intrinsic interferon response. Through unbiased CRISPR screens, we establish dsRNA-sensing and interferon signaling are primary mediators that potentiate T-cell killing of cancer cells following METTL3 inhibition. We show in a range of immunocompetent mouse models that although METTL3 inhibition is equally efficacious to anti-PD-1 therapy, the combination has far greater preclinical activity. Using SPLINTR barcoding, we demonstrate that anti-PD-1 therapy and METTL3 inhibition target distinct malignant clones, and the combination of these therapies overcomes clones insensitive to the single agents. These data provide the mole-cular and preclinical rationale for employing METTL3 inhibitors to promote antitumor immunity in the clinic. SIGNIFICANCE: This work demonstrates that METTL3 inhibition stimulates a cell-intrinsic interferon response through dsRNA formation. This immunomodulatory mechanism is distinct from current immunotherapeutic agents and provides the molecular rationale for combination with anti-PD-1 immune-checkpoint blockade to augment antitumor immunity. This article is featured in Selected Articles from This Issue, p. 2109.
  • Item
    No Preview Available
    CRISPR-ChIP reveals selective regulation of H3K79me2 by Menin in MLL leukemia
    Gilan, O ; Talarmain, L ; Bell, CC ; Neville, D ; Knezevic, K ; Ferguson, DT ; Boudes, M ; Chan, Y-C ; Davidovich, C ; Lam, EYN ; Dawson, MA (NATURE PORTFOLIO, 2023-10)
    Chromatin regulation involves the selective recruitment of chromatin factors to facilitate DNA repair, replication and transcription. Here we demonstrate the utility of coupling unbiased functional genomics with chromatin immunoprecipitation (CRISPR-ChIP) to identify the factors associated with active chromatin modifications in mammalian cells. Specifically, an integrated reporter containing a cis-regulatory element of interest and a single guide RNA provide a chromatinized template for a direct readout for regulators of histone modifications associated with actively transcribed genes such as H3K4me3 and H3K79me2. With CRISPR-ChIP, we identify all the nonredundant COMPASS complex members required for H3K4me3 and demonstrate that RNA polymerase II is dispensable for the maintenance of H3K4me3. As H3K79me2 has a putative oncogenic function in leukemia cells driven by MLL translocations, using CRISPR-ChIP we reveal a functional partitioning of H3K79 methylation into two distinct regulatory units: an oncogenic DOT1L complex directed by the MLL fusion protein in a Menin-dependent manner and a separate endogenous DOT1L complex, where catalytic activity is directed by MLLT10. Overall, CRISPR-ChIP provides a powerful tool for the unbiased interrogation of the mechanisms underpinning chromatin regulation.
  • Item
    No Preview Available
    Inhibition of the CtBP complex and FBXO11 enhances MHC class II expression and anti-cancer immune responses
    Chan, KL ; Gomez, J ; Cardinez, C ; Kumari, N ; Sparbier, CE ; Lam, EYN ; Yeung, MM ; Garciaz, S ; Kuzich, JA ; Ong, DM ; Brown, FC ; Chan, Y-C ; Vassiliadis, D ; Wainwright, EN ; Motazedian, A ; Gillespie, A ; Fennell, KA ; Lai, J ; House, IG ; Macpherson, L ; Ang, C-S ; Dawson, S-J ; Beavis, PA ; Wei, AH ; Burr, ML ; Dawson, MA (CELL PRESS, 2022-10-10)
    There is increasing recognition of the prognostic significance of tumor cell major histocompatibility complex (MHC) class II expression in anti-cancer immunity. Relapse of acute myeloid leukemia (AML) following allogeneic stem cell transplantation (alloSCT) has recently been linked to MHC class II silencing in leukemic blasts; however, the regulation of MHC class II expression remains incompletely understood. Utilizing unbiased CRISPR-Cas9 screens, we identify that the C-terminal binding protein (CtBP) complex transcriptionally represses MHC class II pathway genes, while the E3 ubiquitin ligase complex component FBXO11 mediates degradation of CIITA, the principal transcription factor regulating MHC class II expression. Targeting these repressive mechanisms selectively induces MHC class II upregulation across a range of AML cell lines. Functionally, MHC class II+ leukemic blasts stimulate antigen-dependent CD4+ T cell activation and potent anti-tumor immune responses, providing fundamental insights into the graft-versus-leukemia effect. These findings establish the rationale for therapeutic strategies aimed at restoring tumor-specific MHC class II expression to salvage AML relapse post-alloSCT and also potentially to enhance immunotherapy outcomes in non-myeloid malignancies.
  • Item
    Thumbnail Image
    Targeting Menin disrupts the KMT2A/B and polycomb balance to paradoxically activate bivalent genes
    Sparbier, CE ; Gillespie, A ; Gomez, J ; Kumari, N ; Motazedian, A ; Chan, KL ; Bell, CC ; Gilan, O ; Chan, Y-C ; Popp, S ; Gough, DJ ; Eckersley-Maslin, MA ; Dawson, S-J ; Lehner, PJ ; Sutherland, KD ; Ernst, P ; McGeehan, GM ; Lam, EYN ; Burr, ML ; Dawson, MA (NATURE PORTFOLIO, 2023-02)
    Precise control of activating H3K4me3 and repressive H3K27me3 histone modifications at bivalent promoters is essential for normal development and frequently corrupted in cancer. By coupling a cell surface readout of bivalent MHC class I gene expression with whole-genome CRISPR-Cas9 screens, we identify specific roles for MTF2-PRC2.1, PCGF1-PRC1.1 and Menin-KMT2A/B complexes in maintaining bivalency. Genetic loss or pharmacological inhibition of Menin unexpectedly phenocopies the effects of polycomb disruption, resulting in derepression of bivalent genes in both cancer cells and pluripotent stem cells. While Menin and KMT2A/B contribute to H3K4me3 at active genes, a separate Menin-independent function of KMT2A/B maintains H3K4me3 and opposes polycomb-mediated repression at bivalent genes. Release of KMT2A from active genes following Menin targeting alters the balance of polycomb and KMT2A at bivalent genes, facilitating gene activation. This functional partitioning of Menin-KMT2A/B complex components reveals therapeutic opportunities that can be leveraged through inhibition of Menin.
  • Item
    Thumbnail Image
    Pharmacologic Reduction of Mitochondrial Iron Triggers a Noncanonical BAX/BAK Dependent Cell Death
    Garciaz, S ; Guirguis, AA ; Muller, S ; Brown, FC ; Chan, Y-C ; Motazediani, A ; Rowe, CL ; Kuzich, JA ; Chan, KL ; Tran, K ; Smith, L ; MacPherson, L ; Liddicoat, B ; Lam, EYN ; Caneque, T ; Burr, ML ; Litalien, V ; Pomilio, G ; Poplineau, M ; Duprez, E ; Dawson, S-J ; Ramm, G ; Cox, AG ; Brown, KK ; Huang, DCS ; Wei, AH ; McArthur, K ; Rodriguez, R ; Dawson, MA (AMER ASSOC CANCER RESEARCH, 2022-03)
    UNLABELLED: Cancer cell metabolism is increasingly recognized as providing an exciting therapeutic opportunity. However, a drug that directly couples targeting of a metabolic dependency with the induction of cell death in cancer cells has largely remained elusive. Here we report that the drug-like small-molecule ironomycin reduces the mitochondrial iron load, resulting in the potent disruption of mitochondrial metabolism. Ironomycin promotes the recruitment and activation of BAX/BAK, but the resulting mitochondrial outer membrane permeabilization (MOMP) does not lead to potent activation of the apoptotic caspases, nor is the ensuing cell death prevented by inhibiting the previously established pathways of programmed cell death. Consistent with the fact that ironomycin and BH3 mimetics induce MOMP through independent nonredundant pathways, we find that ironomycin exhibits marked in vitro and in vivo synergy with venetoclax and overcomes venetoclax resistance in primary patient samples. SIGNIFICANCE: Ironomycin couples targeting of cellular metabolism with cell death by reducing mitochondrial iron, resulting in the alteration of mitochondrial metabolism and the activation of BAX/BAK. Ironomycin induces MOMP through a different mechanism to BH3 mimetics, and consequently combination therapy has marked synergy in cancers such as acute myeloid leukemia. This article is highlighted in the In This Issue feature, p. 587.