71: Targeted cancer genetics

71: Targeted cancer genetics

EACR-23 Oral Presentations, Monday 7 July 2014 / European Journal of Cancer 50, Suppl. 5 (2014) S12–S20 generally to investigate the connection betwee...

75KB Sizes 1 Downloads 111 Views

EACR-23 Oral Presentations, Monday 7 July 2014 / European Journal of Cancer 50, Suppl. 5 (2014) S12–S20 generally to investigate the connection between RNF20 and inflammationdriven cancer. Our studies revealed a role for RNF20 in regulating an inflammatory response mediated through NF-úB. Specifically, RNF20 and H2Bub1 were found to reduce NF-úB-dependent transcription in noncancerous cells. We identified a possible mechanism by which RNF20 promoted the binding of homodimers of the p50 NF-úB subunit to target gene promoters, thus repressing target gene expression. Conversely, depletion of RNF20 and H2Bub1 led to recruitment of p65 and induction of target gene expression. Consistent with these in vitro findings, RNF20+/− mice were prone to more severe acute and chronic inflammation after dextran sodium sulfate (DSS) treatment, and succumbed more rapidly to inflammation-associated colorectal tumors. Importantly, human colorectal tumors often exhibit reduced levels of H2Bub1. Taken together, these results demonstrate that RNF20 may be an important regulator of inflammation, and an inhibitor of inflammationinduced cancer. Conclusion: Our findings suggest that RNF20 and H2Bub1 are negative regulators of inflammation and inflammation-induced cancer. Furthermore, constitutively low H2Bub1 levels may predispose humans to inflammatory disease and cancer. No conflict of interest. 67 Unexpected cancer-related properties of BRCA1 Y. Hu1 , S. Dimitrov1 , H. Wang1 , D. Livingston1 . 1 Dana Farber Cancer Institute, Emil Frei Professor of Genetics and Medicine Harvard Medical School, Boston Massachusetts, USA The BRCA1 tumor suppressor gene encodes multiple, alternatively spliced products, one of which, p220, is its prime cancer-suppressing protein. p220 is known to suppress breast and ovarian cancer, and germ line BRCA1 mutation-carrying males are largely protected against developing BRCA1-driven disease. p220 is a large, multifunctional, chromatin-binding E3 ubiquitin ligase that binds numerous partner proteins and participates in a multitude of processes that support the maintenance of genome integrity. Some of these activities arise in response to active genome damage and support proper DNA damage repair and checkpoint activation. Others engage in regulating centrosome duplication and normal mitotic events like spindle pole development and outcomes like proper chromosome segregation. Yet others are directed at proper heterochromatin development and the repression of satellite RNA synthesis. A long-standing paradigm is that BRCA1 cancer results from a loss of BRCA1 function in the cells of the breast and ovarian/fallopian tube epithelium, a notion that is underscored by the fact that virtually all inherited BRCA1 cancers are known to have undergone LOH at BRCA1 with maintenance of the mutant allele. That said, we have recently accumulated evidence that, much like what happens when p220 abundance is greatly reduced, too much p220, which can arise either in G1 or in S/G2 from a defect in its mRNA turnover, is also associated with chronic genome damage. Similarly, we have identified another biochemical process that normally results in the poly ADP ribosylation of a specific domain of p220 in cells and that, when defective, leads to an exaggerated p220 response to the development of double strand genomic breaks. This p220-dependent hyperrecombinational DNA damage repair response is, unexpectedly, associated with the development of gross genome disorder, much like what happens when p220 expression is lost in cells. Since the development of chronic genome disorder is a major contributor to the development of BRCA1 cancer, these results trigger speculation that BRCA1 can elicit a tumorigenic response in two ways − (a) by its loss of function and (b) by the retention, deregulation, and hyperactivity of one or more of its key DNA damage response functions. If shown to be true, such a bimodal model would significantly alter the paradigm that explains the genesis of BRCA1 cancer. No conflict of interest.

Monday 7 July 2014

14:00−15:45

Symposium

Novel Cancer Therapies 68 Novel therapeutic approaches to lung cancer No abstract received. No conflict of interest information specified. 69 The TGF-beta pathway as a therapeutic target ´ University Hospital, Barcelona, Spain J. Seoane1 . 1 Vall D’Hebron During the past decades, the TGF-beta pathway has recently emerged as a putative therapeutic target against cancer. However, TGF-beta has a complex role in cancer. During tumour progression TGF-beta becomes an oncogenic

S17

factor inducing proliferation, angiogenesis, invasion, and metastasis, as well as suppressing the anti-tumoral immune response. The precise understanding of the role of TGF-beta in oncogenesis is required in order to design optimal therapeutic approaches and select the patient population that may benefit from an anti-TGF-beta therapy. We will review the rationale for evaluating TGF-beta signalling inhibitors as cancer therapeutics, and the progress made in the preclinical and clinical testing of anti-TGF-beta compounds in the context of glioblastoma. No conflict of interest. 70 Proffered Paper: Inhibition of RNA Polymerase I transcription by CX-5461 as a completely new approach to treat highly refractory haematological malignancies R. Hannan1 , N. Hein1 , S.E. O’Brien2 , D. Drygin3 , C. Cullinane1 , G. Matthews1 , R.W. Johnstone4 , R.B. Pearson4 , G. McArthur4 , S. Harrison4 . 1 Peter MacCallum Cancer Centre, Research, Melbourne Victoria, Australia, 2 Senhwa Biosciences Inc, Research and Development, San Diego CA, Australia, 3 Pimera Inc, Research and Development, San Diego CA, USA, 4 Peter MacCallum Cancer Centre, Resarch, Melbourne Victoria, Australia Background: Recent findings by our group have been instrumental in the development of the novel selective inhibitor of RNA Polymerase I (Pol I), CX-5461 (Drygin et al., Cancer Research, 2011; Bywater et al. Cancer Cell, 2012). This work has led to the fundamental discovery that ribosomal gene transcription by Pol I is not simply a ‘house keeping’ process in cancer cells but is highly regulated to maintain their viability (Bywater et al., Nature Reviews Cancer 2013). Strikingly, inhibition of Pol I transcription shows a profound selectivity for malignant over normal cells in preclinical studies. Material and Methods: To explore the therapeutic potential of Pol I transcription inhibition via CX-5461 in hematological malignancies refractory to standard chemotherapy, we employed mouse models of highly aggressive MLL-driven Acute Myeloid Leukemia (AML) (MLL/AF9 + Nras and MLL/ENL + Nras) and V*ú-Myc-driven Multiple Myeloma (MM). Results: CX-5461 administration significantly increased the overall survival time of AML-bearing mice (MLL/ENL Nras median17 days for vehicle vs 36 days for drug, P < 0.0001; MLL/AF9 Nras median 15 days for vehicle vs 23 days for drug, P 0.0009) without severe adverse effects. The extended survival of MLL-driven AML bearing mice was associated with, induction of cell death and an aberrant G2/M cell cycle progression. In contrast, treatment of Nras AML-bearing mice with Cytarabine/Doxorubicin (5:3) resulted in a significantly lower survival advantage as compared to animals treated with CX-5461 therapy. Similarly, in MM-bearing mice, chronic CX-5461 administration robustly reduced the secretion of serum monoclonal Ig, and significantly prolonged the overall survival time (V*ú-Myc median 102 days for vehicle vs 210 days for drug, ongoing) demonstrating that CX-5461 exhibits potent anti-tumour activity in MM model. Conclusions: These data demonstrate that hyperactivated Pol I transcription can be successfully targeted through small molecule inhibitors to treat models of human AML and MM that are refractory to standard therapy. Based on these and previously published results (Bywater et al., Cancer Cell, 2012) and a favorable toxicology profile, we have recently initiated a first-in-human phase I clinical trial of this first-in-class drug, CX-5461 in patients with advanced hematological malignancies. No conflict of interest. 71 Targeted cancer genetics 2 C. Von Kalle1 , S. Frohling ¨ , H. Glimm1 , S.M. Pfister3 , T. Zenz2 . 1 National Center for Tumor Diseases (NCT) and German Cancer Research Center, Translational Oncology DKFZ-Heidelberg, Heidelberg, Germany, 2 National Center for Tumor Diseases (NCT) German Cancer Research Center and Heidelberg University Hospital, Translational Oncology DKFZ-Heidelberg, Heidelberg, Germany, 3 German Cancer Research Center and Heidelberg University Hospital, Pediatric Neurooncology DKFZ-Heidelberg, Heidelberg, Germany

Many recurrent genetic aberrations have been detected through cancer genome sequencing, however, their associated functional dependencies remain elusive in many cases. Moreover, tumors that are currently classified according to pathology often display heterogeneous biologic characteristics and clinical course. We have implemented a multi-pronged approach to systematically investigate genotype-phenotype correlations in a broad spectrum of human cancers through comprehensive genomic profiling, functional annotation of genetic variants, and ex vivo drug screening. This multidimensional strategy will enable stratification of patients according to specific, context-dependent vulnerabilities that can be exploited for therapeutic benefit. We have determined the functional consequences of multiple genetic abnormalities occurring in patients with acute myeloid leukemia and B-cell chronic lymphocytic leukemia using in vitro and in vivo experimental systems. In addition to these directed investigations, we have performed unbiased

S18

EACR-23 Oral Presentations, Monday 7 July 2014 / European Journal of Cancer 50, Suppl. 5 (2014) S12–S20

functional studies, employing tools such as high-throughput RNA interference, to discover “secondary” pathway dependencies that arise in consequence of particular genetic alterations, such as oncogenic KRAS mutations, and we have devised strategies to target such context-specific liabilities for therapeutic benefit. We have extensively characterized the dynamic composition of the tumor-initiating cell compartment in xenograft models of human colorectal and pancreatic cancer, providing a unique resource for investigations into the functional requirements of “cancer stem cells” as well as the genetic basis of the functional heterogeneity observed in human cancers. Finally, we have established a platform for ex vivo treatment of genetically annotated primary human lymphoma specimens with a wide range of pathway inhibitors to systematically correlate mutational profiles with patterns of drug response. “Undruggable” genetic alterations were addressed in the context of gene fusions involving the H3K4 methyltransferase MLL, which define an aggressive subtype of acute myeloid leukemia (AML). Using a systematic functional genomic approach, we discovered that MLL fusion-driven AML cells are exceptionally reliant on the cell cycle regulator CDK6, but not its functional homolog CDK4, providing the rationale for a clinical trial of the smallmolecule CDK6 inhibitor palbociclib in patients with relapsed or refractory MLLrearranged leukemia. Moreover, we systematically investigate heterogeneity of drug response in primary chronic lymphocytic leukemia (CLL) and lymphoma using a cell-based high-throughput drug-screening platform and associate the response phenotype with genetics and other omics technology. The work offers a novel functional classification of CLL based on drug sensitivity and identifies drugs with preferential activity for TP53 mutant CLL. While cancer specific aberrations in adults tend to incur a multitude of genetic aberrations, the pediatric tumor genome is less complex and thus predestined for treatment by targeted agents. We identified mutations impacting MAPK signaling as the genetic hallmark in low-grade gliomas in children classifying this indication as a MAPK disease. In pediatric medullablastoma we identified the pivotal role of aberrant SHH signaling − which in turn serves as genetic predictor of response to SMO inhibition. The clinical implementation of a center-wide NCT MASTER (Molecularly Aided Stratification for Tumor Eradication) umbrella protocol provides all components of a clinical workflow for high-throughput molecular diagnostics. A pilot protocol for adult cancer patients with a particularly challenging disease course and surprise responders has enrolled more than 100 patients and identified actionable genetic lesions in a substantial proportion of cases. The nationwide INFORM study aims to enroll all children and adolescents with relapsed or refractory cancers in Germany to systematically enable personalized treatment based on comprehensive molecular profiling of individual tumors. No conflict of interest.

Monday 7 July 2014

14:00−15:45

Symposium

Inflammation and Cancer 72 Inflammation in colorectal and liver tumorigenesis M. Karin1 . 1 University of California San Diego, Laboratory of Gene Regulation and Signal Transduction Department of Pharmacology, La Jolla CA, USA Inflammation plays important roles in the pathogenesis of colorectal and liver cancers. Chronic inflammation of both tissues increases cancer risk in part through elevated expression of IL-6 and other pro-tumorigenic cytokines such as IL-1 and TNF. IL-6 exerts its tumorigenic activity by signaling via the receptor subunit gp130 to activate transcription factor STAT3. Curiously, activating gp130 mutations were found in human inflammatory hepatic adenoma, that when combined with activating mutations in the Wnt-b catenine pathway can lead to hepatocellular carcinoma (HCC). We expressed such a form of gp130 throughout the intestinal tract to determine its impact on development and tumorigenesis in the organ. Surprisingly, we found that activated gp130 has a strong impact on tissue homeostasis and leads to activation of a regenerative pathway responsible for the repair of mucosal injury, which if left unrepaired can lead to chronic intestinal inflammation. Interestingly, this pathway is independent of STAT3 and instead it relies on activation of YAP, a transcriptional co-activator that controls tissue growth and is frequently activated in HCC. These results suggest that IL-6 family members promote colon and liver tumorigenesis through multiple mechanisms, all of which need to be targeted to achieve an effective therapeutic effect. No conflict of interest.

73 Parainflammation in cancer A. Lasry1 , H. Hamza1 , E. Kadosh1 , E. Elyada1 , A. Pribluda1 , K. Alitalo2 , T. Stiewe3 , M. Oren4 , E. Pikarsky5 , Y. Ben-Neriah1 . 1 The Hebrew University of Jerusalem, School of Medicine-IMRIC-Immunology and Cancer Research, Jerusalem, Israel, 2 Biomedicum Helsinki and the Finnish Institute for Molecular Medicine, University of Helsinki, Helsinki, Finland, 3 Philipps-University, Marburg, Germany, 4 Weizmann Institute, Molecular Cell Biology, Rehovot, Israel, 5 Hebrew University, Immunology and Pathology, Jerusalem, Israel Inflammation has many faces, most commonly observed as an acute reaction in response to pathogen or another insult, or a chronic phase, accompanying chronic infection and chronic remittent inflammatory disease, such as inflammatory bowel disease [1]. Yet, there is another type of smoldering inflammation, harder to notice or monitor, which appears to underlie some of the major human diseases, cancer, diabetes type 2 and certain neurodegenerative diseases [2]. We have developed mouse models of cancer based on inducible CKIa knockout [3], which exhibit smoldering inflammation, and demonstrate how a low grade, infiltrate-free inflammatory reaction to persistent DNA damage response translates to an aberrant growth. We determined this unusual inflammatory repertoire denoted parainflammation [4] in the knockout mice and gut explants, demonstrated its association with cellular senescence and showed how in the absence of p53, parainflammation is converted from a growth inhibitory to growth promoting mechanism, both in vitro and in vivo. Anti-inflammatory reagents capable of blocking parainflammation reverse a tumor-related crypt proliferative phenotype of mutant intestinal organoids in vitro and prevent carcinogenesis in mutant mice. Our studies may explain the anti-carcinogenic effects of nonsteroidal anti-inflammatory drugs in human cancer patients [5]. We will discuss mechanisms and evolutionary aspects connecting inflammation and growth. Reference(s) [1] Medzhitov, R. Origin and physiological roles of inflammation. Nature 454, 428−35 (2008). [2] Ben-Neriah, Y. & Karin, M. Inflammation meets cancer, with NF-kappa B as the matchmaker. Nature Immunology 12, 715–723 (2011). [3] Elyada, E., Pribluda, A., et al. CKI alpha ablation highlights a critical role for p53 in invasiveness control. Nature 470, 409-U208 (2011). [4] Pribluda, P., Elyada, E., et al. A senescence-inflammatory switch from cancer-inhibitory to cancer-promoting mechanism. Cancer Cell 24, 242–256 (2013). [5] Chan, A.T. & Lippman, S.M. Aspirin and colorectal cancer prevention in Lynch syndrome. Lancet 378, 2051–2052 (2011). No conflict of interest. 74 Proffered Paper: Investigation of the spatial heterogeneity of specific immune cell phenotypes in the tumor microenvironment of follicular lymphoma J. Mansfield1 , L. Nelson2 , B. Wendik3 , C. Van der Loos4 , C. Rose2 , R. Byers2 . 1 CRi, Woburn, USA, 2 University of Manchester, Manchester, United Kingdom, 3 PerkinElmer, Hopkinton, USA, 4 Academic Medical Center, Amsterdam, Netherlands Background: Tumor-infiltrating lymphocytes (TILs) are present in the tumor microenvironment of many cancers, with consequent tumor immunogenicity and association with survival. In particular, increased levels of regulatory T cells (Tregs) are associated with poorer prognosis in some cancers. However, given the complexity of TIL phenotype their visualization in situ is difficult, and impossible without multiplex immunophenotyping. An understanding of both the phenotype and spatial distribution of TILs in situ within the tumor microenvironment would be advantageous to understanding their role in tumour immunobiology, especially given growing interest in tumour immunotherapy. Here we present a multi-marker, computer-aided method for analysing the distributions of CD3/FOXP3 (Treg) and CD3/CD69 (Tact) T cells in follicular lymphoma sections using a multispectral imaging (MSI) and automated analysis approach. An hypothesized interaction distance (HID) analysis was used to determine whether the spatial patterns of Tregs and Tacts were prognostically significant. Design: A single section of a tissue microarray containing triplex follicular lymphoma cores from 40 subjects [24 male, 16 female, age 35 to 75 years at diagnosis, median 55 years, 2–171 months follow-up] was stained for CD3, FOXP3, CD69 and hematoxylin. Each core was imaged using MSI and the individual staining of each marker separated from each other using spectral unmixing. CD3+ TILs were located using automated image analysis. The FOXP3 and CD69 status of each CD3+ TIL was then determined and the spatial distributions of the CD3/FOXP3 and CD3/CD69 cells were used as input into the HID analysis. Results: Multiplexed IHC staining, MSI and automated per-cell quantitative analysis was successful. Kaplan–Meier analysis demonstrated favourable outcome with higher numbers of CD3+, CD3+/FOXP3+ and CD3+/CD69+ cells. HID analysis demonstrated the association of favourable outcome with a