Bob Brown: New challenges in using biological endpoints for epigenetic therapies in clinical trials Same genotype, different phenotype Same genome, different epigenome Same genotype, different phenotype High Grade Serous Ovarian Cancer: Similar genome, very different epigenome (TCGA) Many important genes are epigenetically silenced in malignant cells • Cell cycle: Rb, p16INK4a, p15INK4a, p14ARF • Signal transduction: RASSF1, APC • Apoptosis: DAPK, Caspase 8 • DNA repair: MLH1, MGMT, BRCA1 • Senescence: TERT, TERC • Invasion/metastasis: TIMP-3, E-cadherin APC = adenomatous polyposis coli DAPK = death-associated protein kinase MGMT = O-6-methylguanine-DNA methyltransferase 1. Jones & Baylin. Cell 2007;128:683–92 2. Teodoridis JM, et al. Drug Resistance Updates 2004;7:267–78 HDAC Acetyl Acetyl MBD protein Me Me Me DNA Histone Histone Acetyl HDAC MBD protein Me Me Me DNA Histone Histone HMT + Me Me Me MBD protein Me Me Me DNA Histone Histone HMT MBD protein Me Me Me DNA Me Me Me Epigenetic Therapies • DNA Methyltransferase (DNMT) Inhibitors – Azacytidine: approved in the EU for the treatment of patients with higher-risk MDS, CMML and AML – Decitabine: approved in the USA for the treatment of patients with all FAB classifications of MDS • Histone Deacetylase (HDAC) Inhibitors – Vorinostat: approval in US for treatment of advanced cutaneous T-cell lymphoma Challenges with current epigenetic therapies • Toxicity • Delivery • Short plasma half-life (although long pharmacodynamic half-life) • Lack of targeting • Do they work by epigenetic mechanism? • What are the chemotherapeutic epigenetic target? • Lack of predictive biomarkers • Do they target subpopulations of tumour stem cells? • How to design early clinical trials if only targeting subpopulation • Is the maximum biological dose the same as the maximum tolerated dose? Proof of Mechanism: Do they do what they say on the tin? Decitabine PK vs PD in PBCs 140 120 Decitabine AUC Peak plasma levels of decitabine correlate with demethylation in PBMCs 100 80 60 40 20 0 5 10 15 20 Methycytosine AAC CR-UK Phase I dose escalation trial of decitabine and carboplatin in patients with advanced solid tumours (Appleton et al 2007) Ratio 5-methylcytosine: cytosine in PBMC DNA (CRUK Phase I trial of Decitabine & carboplatin in advanced solid tumours) 4.5 4.0 Ratio Decitabine induces demethylation several days after treatment that reverses over time 45mg/m2 3.5 90mg/m2 3.0 135mg/m2 2.5 2.0 1.5 0 5 10 15 Days 20 25 Proof of Concept: Do they biologically do what they should? HbF Day 15 Day 12 Day 10 Day 8 K562 Day 1 aActin CP70 Proof of Concept: Gene expression (but not for all genes) Cycle 1 45 90 135 non-epithelial Proof of Concept: Apoptosis (normal or tumour?) CK18 (% of day 1) 200 150 100 50 0 2 4 6 8 10 12 14 16 18 20 22 Day Combination of DNMT and HDAC inhibitor enhances gene re-expression and chemosensitisation DAC DAC+ PXD101 Day 12 5 Day 16 Relati ve tumour vol ume Day 6 Day 9 4 3 2 1 0 Steele et al 2010 Control Cisplatin DAC DAC+Cisplatin PXD101 DAC+PXD101+Cisplatin 0 1 2 3 Time (Days) 4 5 6 Origin of Cancer – Role of Cancer Stem Cells (CSC)? Ovarian cancer cell lines & primary ascites contain Side Population cells PEO23: SP 6.90% PEO23 +verapamil: SP 0.00% Specimen_001_23.fcs 512 768 512 SP 256 256 0 0 256 424/44nm (L3)-A HOECHST BLUE-A 768 512 768 HOECHST RED-A Rizzo et al, 2011 SP 45neg live Ascites010509_CD45 FITC.fcs 1024 Specimen_001_23 Verapamil.fcs 1024 1024 1024 Patient Ascites SP 0.021% 768 512 SP 256 0 0 0 256 512 768 1024 HOECHST RED-A 0 256 512 768 1024 670nmLP (L3)-A Patient ascites SP: increases following treatment Cell lines derived from matched patient ascites Primary patient ascites Rizzo et al, 2011 IGROV1 SP cells have tumour stem cell like properties (a) Tumour Initiation (c) 2D colony formation Rizzo et al, 2011 (b) Spheroid growth (d) Repopulation Group Gene set Gene set name p value FDR Source ES Expressed Es exp2 ES2 0.39 0.7313 overexpressed in hES cells according to a meta-analysis Nanog targets nanog 0.128 0.32 ChIP array of Nanog in hES cells: activated genes Oct4 targets oct4 <10-6 <10-4 ChIP array of Oct4 in hES cells; activated genes Sox2 targets sox2 0.128 0.32 ChIP array of Sox2 in hES cells; activated genes NOS targets Nos <10-6 <10-4 overlap of above three sets Suz12 targets suz12 0.128 0.64 ChIP array of Suz12 in hES cells Eed targets eed 0.388 0.7313 ChIP array of Eed in hES cells H3K27 bound h3k27 0.254 0.645 ChIP array of trimethylated H3K27 in hES cells PRC2 targets prc2_targets1 0.128 0.64 overlap of three above sets PRC2 targets prc2_targets2 <10-6 <10-4 PRC2 repressed targets transcriptionally reactivated by DZNep PRC1 polycomb complex1 0.258 0.645 polycomb complex1 genes PRC2 polycomb complex2 <10-6 <10-4 polycomb complex2 genes NOS targets polycomb targets polycomb complex EZH2 and ABCB1 expression is increased in Side Population from patient ascites Patient Ascites sample number Ratio of expression of ABCB1 mRNA SP:non-SP Ratio of expression of EZH2 mRNA SP:non-SP 6 12.9 14.5* 7 3.1 4.2* 9 8.4 1.3 10 16.9 1.3 14 2.9 2.1* 16 3.7 5.9* 17 57.6 8.6* 18 10.3 1.6* 19 51.8 1.1 21 36.8 3.4* PRC2 is a protein complex that catalyses the protein methylation of lysines on histones (H3K27me3) IGROV1 PEO14 H3K27me3 Histone H3 h. 24 48 72 96 24 Control siRNA 48 72 96 EZH2 siRNA 24 48 72 96 24 Control siRNA 48 72 96 EZH2 siRNA Does targeting EZH2 reduce sustaining/ stem cells? (a) EZH2 knock-down using SiRNA (Rizzo et al., 2011) (b) EZH2 compounds (Chapman-Rothe, Shasaei, Rizzo, Cherblanc, Fuchter) H3K27me3 EZH2 Beta-actin EZH2 as a potential anti-cancer target ? • Many genes in cancer, including tumour suppressor genes, are epigentically silenced by mechanisms associated with H3K27me3 which can be independent of DNA methylation (Kondo et al Nat Genet, 2008; 40: 741-750) • H3K27me3 is somatically inherited during cell division (Margueron et al Nature, 2009. 461: 762-7) • Repressive chromatin marks in tumour stem cells may make genes vulnerable to CpG island DNA methylation (Ohm et al 2007, Nat Genet, 39; 237-242) • EZH2 is frequently over-expressed in a wide variety of tumour types and is driver of metastasis (Min et al Nature Medicine 2010, 16: 286-94) • EZH2 is essential for Glioblastoma cancer stem cell maintenance (Suvà et al, Cancer Res 2009 69:921) GOG 218 and ICON-7: results • both trials are positive, with highly significant improvements in progressionfree survival • overall survival analysis immature (too few events) • no new safety concerns (hypertension in >20%; bowel perforation in <2%) • and yet .... ↑ Burger et al. GOG study - presented at ASCO, Chicago, 2010. Perren T et al. (ICON-7) – presented at ESMO, Milan 2010. → GOG investigator analysis used CA125/RECIST-determined progression. If data censored for CA125, median PFS for Arm I and III increase to 12.0 m and 18.2 m, respectively. 23 CpG island methylation as a biomarker Stable in vivo and ex vivo Sensitive PCR based assays for single loci Array based methods for genome wide patterns Aberrant tumour methylation can be detected in tumour DNA in accessible body fluids DNA Methylation Prognostic Biomarkers in Wnt signalling pathway Red: SGCTG cohort & TCGA cohort Blue: SGCTG cohort only Orange: Absent in TCGA cohort Dai et al 2011 Systematic analysis of other pathways Table 3: Multivariate progression free survival analysis of loci significantly associated with Progression-free survival in univariate analysis Pathway/ family AKT/mTOR p53 BRCA1/2 Redox MMR HR Multivariate PFS analysis (n=111) genes HR 95% CI adjusted p value VEGFA AKT1 13.8 27.2 (0.9, 210.8) (2.2, 329.1) 0.06+ 0.009** VEGFB||DNAJC4 BAI1 BAX LRDD CCND1 HDAC4 HDAC11 PRDX2 TR2IT1 LIG1 MLH3 LRRC14||REC QL 16.2 33.8 1.7 21.2 4.6 4.7 7 2.8 28.4 1.8 218.6 (1.6, 162.1) (1.3, 866.5) (0.7, 4.3) (1.8, 250.5) (0.5, 37.9) (0.7, 32) (1, 47.8) (1.5, 5.5) (2.3, 352.4) (1.0, 3.5) (7.7, 6.2x103) 0.018* 0.033* 0.234 0.016* 0.161 0.110 0.048* 0.002** 0.009** 0.644 0.002*** 45.4 (0.4, 4.6x103) 0.105 Dai, Zeller, et al Epigenetics Unit Teams & support Imperial College: Institute of Cancer Research: Tumour DNA Methylation Profiling Epigenetics (stem cell) team • Constanze Zeller • Sian Rizzo • Elizabeth Evans • Alessandra Silva • Jenny Quinn • Jenny Quinn • Jens Teodoridis • Louisa Luk • Janet Graham • James Flanagan • Prof. Stan Kaye Chromatin targets • Nadine Chapman-Rothe • Gary Box, Sue Eccles • Ely Shamsaei • Ian Titley, Gowri Vijayaraghavan • Fanny Cherblanc • Craig Carden, Debbie Tandy • Matt Fuchter Bioinformatics • Wei Dai Tissue collection • Sadaf Ghaem-Maghami, Nona Rama, Amy Ford, Nicole Martin