Scientific Program

Conference Series Ltd invites all the participants across the globe to attend 5th European Immunology Conference Berlin, Germany.

Day 2 :

Keynote Forum

Kathleen B Schwarz

Johns Hopkins University School of Medicine, USA

Keynote: Hypothesis testing: A role for inflammasome activation in pediatric autoimmune hepatitis

Time : 10:00-10:30

Conference Series Euro Immunology 2016 International Conference Keynote Speaker Kathleen B Schwarz photo
Biography:

Kathleen B Schwarz was graduated from Washington University School of Medicine, USA. She is a Professor of Pediatrics at the Johns Hopkins University School of Medicine where she is the Director of the Pediatric Liver Center. She is the President of the Federation for International Societies of Pediatric Gastroenterology, Hepatology and Nutrition. She has published more than 165 papers in peer-reviewed journals.

Abstract:

Introduction: An infectious trigger in a genetically susceptible host has been proposed as etiopathogenic in autoimmune hepatitis (AIH). Often formalin-fixed paraffin-embedded liver biopsies (FFPE) are the only hepatic tissues available for pathogenetic investigations but retrieval of mRNA from small biopsies is problematic. Our overall goal was to develop methods to obtain high quality mRNA from FFPE liver biopsies in order to perform unbiased high throughput sequencing of transcriptomes.rnrnMethods: 45 FFPE liver biopsies, 25 from AIH type 1 patients’ and 21 from controls were used to generate RNA libraries and analyzed using RNAseq. Total RNA was extracted from two cored tissue samples, cDNA libraries were constructed using the Illumina TruSeq Stranded Total RNA Sample Preparation Kit with Ribo-Zero to remove cytoplasmic and mitrochondrial rRNA and indexed paired-end sequencing was performed on an Illumina HiSeq2000 with 90 million paired reads per sample.rnrnResults: There was up-regulation in AIH patients; vs. controls of a number of intrahepatic genes; among these were genes related to B cell development and several consistent with activation of the NLRPEi inflammasome. The approach to further investigating a role of NLRPi inflammasome activation in AIH will be discussed including PCR and immunohistochemistry of liver biopsies and serum ELISA assays for inflammasome components as well as PBMC assays.rnrnConclusions: Possible therapeutic implications, including monoclonal antibodies against inflammasome components as well as anakinra (anti IL-1), beta hydroxybutyrate and the sulfonyl urea MCC950 which block NLRPEi will be discussed.rn

Keynote Forum

Yaffa Mizrachi Nebenzahl

Ben Gurion University of the Negev, Israel

Keynote: Natural killer receptor 1 dampens the development of allergic eosinophilic airway inflammation

Time : 10:30-11:00

Conference Series Euro Immunology 2016 International Conference Keynote Speaker Yaffa Mizrachi Nebenzahl photo
Biography:

Yaffa Mizrachi Nebenzahl has completed her PhD at The Weizmann Institute, Israel in 1985. She is the Head of the Molecular Microbiology Laboratory in The Shraga Segal Department of Microbiology, Immunology and Genetics at the Faculty of Health Sciences in Ben Gurion University of the Negev, Israel.

Abstract:

The function of NCR1 was studied in a model of experimental asthma, classified as a type 1 hypersensitivity reaction, in mice. IgE levels were significantly increased in the serum of OVA immunized NCR1 deficient (NCR1gfp/gfp) mice in comparison to OVA immunized wild type (NCR1+/+) and adjuvant immunized mice. Histological analysis of OVA immunized NCR1gfp/gfp mice revealed no preservation of the lung structure and overwhelming peribronchial and perivascular granulocytes together with mononuclear cells infiltration. OVA immunized NCR+/+ mice demonstrated preserved lung structure and peribronchial and perivascular immune cell infiltration to a lower extent than that in NCR1gfp/gfp mice. Adjuvant immunized mice demonstrated lung structure preservation and no immune cell infiltration. OVA immunization caused an increase in PAS production independently of NCR1 presence. Bronchoalveolar lavage (BAL) revealed NCR1 dependent decreased percentages of eosinophils and increased percentages of lymphocytes and macrophages following OVA immunization. In the OVA immunized NCR1gfp/gfp mice the protein levels of eosinophils’ (CCL24) and Th2 CD4+ T-cells’ chemo attractants (CCL17 and CCL24) in the BAL are increased in comparison with OVA immunized NCR+/+ mice. In the presence of NCR1, OVA immunization caused an increase in NK cells numbers and NCR1 ligand expression on CD11c+GR1+ cells but a decreased NCR1 mRNA expression in the BAL. OVA immunization resulted in significantly increased IL-13, IL-4 and CCL17mRNA expression in NCR1+/+ and NCR1gfp/gfp mice. IL-17 and TNFα expression increased only in OVA-immunized NCR1+/+ mice. IL-6 mRNA increased only in OVA immunized NCR1gfp/gfp mice. Collectively, it is demonstrated that NCR1 dampens allergic eosinophilic airway inflammation.

Break: 11:00-11:15 Coffee Break @ Main Lobby
  • Cancer and Tumor Immunobiology
    Vaccines and Vaccination
    Immunotherapy
    Auto Immunity
    Diagnostic Immunology
    Inflammation
    Innate Cancer immunology
    Immunity Vitamins
Location: Berlin, Germany

Chair

Kathleen B. Schwarz

Johns Hopkins University School of Medicine, USA

Co-Chair

Ildiko Molnar

EndoMed, Hungary

Session Introduction

Jessy S Deshane

University of Alabama at Birmingham, USA

Title: Suppression of B lymphopoiesis by myeloid-derived suppressor cells in tumor-bearing mice

Time : 11:15-11:35

Biography:

Jessy S Deshane is a pulmonary Immunologist with expertise in immune regulation in asthma. She investigates myeloid-derived regulatory cell biology and free radical mechanisms that regulate their differentiation and function. She pioneered these investigations both in mouse models and human asthma. She has authored 46 peer-reviewed publications, including high impact journals like Journal of Experimental Medicine, Journal of Clinical Investigations, Journal of Allergy and Clinical Immunology, Immunity and Cancer Research. She serves on the Editorial Boards for the journals Allergy and American Journal of Respiratory Cell and Molecular Biology and serves on grant review committees.

Abstract:

Rationale: Myeloid-derived suppressor cells (MDSCs) have been well established as regulators of anti-tumor immunity. MDSCs modulate amino acid metabolism in the tumor microenvironment and suppress T-cell function. However, it is less clear whether MDSCs regulate B-cell responses during tumor progression. Methods: Using a syngeneic orthotopic model for lung cancer with murine Lewis Lung Carcinoma cells, we evaluated B-cell subsets in tumor bearing mice by multi parameter flow cytometry. The amount of serum IgG or IL-7 was determined by ELSIA. Phospho-STAT5 and total STAT5 were detected by immunoblotting. To investigate MDSC-mediated suppression of B cell lymphopoiesis, we adoptively transferred MDSCs derived from bone marrow of CD45.2+ tumor bearing mice intratibially into congenic CD45.1+ mice. B-cell subsets in recipient mice at day 7 post MDSC transfer were enumerated as above. In vitro B-cell inhibitory assay was performed by co-culturing CFSE-labeled pre-activated splenocytes with MDSCs purified from bone marrow of tumor-bearing mice at a ratio of 1:1 in the absence or presence of arginase inhibitor nor-NOHA (20 M), iNOS inhibitor 1400W (500 nM) or IDO inhibitor 1-D-MT (1 mM) for 48 hours. The percentage of CD19+CFSElow cells (proliferating cells) was determined by FACS analysis. Results: Percentages and absolute numbers of Pro-, Pre- and mature B-cells were reduced in bone marrow (BM) of tumor bearing mice. Moreover, percentage and absolute number of follicular B cells were reduced, while immature B-cells increased in the spleen of tumor bearing mice. Levels of serum IgG were reduced in tumor-bearing mice. Furthermore, IL-7 and downstream STAT-5 signaling were impaired in tumor bearing mice. Transfer of BM-derived MDSCs from tumor bearing mice into congenic recipients resulted in significant reduction in both percentages and absolute numbers of immature and mature B-cells in peripheral blood of recipient mice. Pre-B cells and immature B-cells also decreased in BM of MDSC transferred recipients. Additionally, MDSCs suppress B-cell proliferation and IgG production by B-cells in an arginase and iNOS dependent but IDO independent manner. Conclusions: In the present study, we demonstrate that B-cell differentiation in vivo is impaired in the BM and spleen of mice with lung cancer. Adoptive transfer studies with congenic mice demonstrate that MDSCs derived from Lewis Lung Carcinoma bearing mice may suppress B-cell differentiation in tumor naive mice. These results together suggest that tumor-related MDSCs may potentially regulate humoral immune responses to promote tumor survival.

Biography:

Ildiko Molnar has completed her PhD in the special field of Graves’ Ophthalmopathy at the Hungarian Academy of Science. Her work and research connected her to Kenezy County and Teaching Hospital from 1977 to 2008. Her research activities are on field of internal medicine, endocrinology, immunology and allergology. Currently she is the Chief of EndoMed, Immunoendocrinology and Osteoporosis Centre, Private Outpatient Clinic from 2008. She is an expert in laboratory methods (ELISA, blotting, allergy testing) and DXA measurement. She has published more than 53 papers in reputed journals, 16 chapters and 2 books.

Abstract:

Type II 5’-deiodinase enzyme (DII) activity is responsible for T4 conversion to T3 resulting in the majority of intracellular T3 concentration. DII is a membrane-anchored protein characterized by tissue-specificity; highly expressed in thyroid, pituitary, skeletal, eye and cardiac muscles, brain, adipose tissue and bone. Decreased DII activity leads to hypothyroidism in euthyroid sick syndrome. We demonstrated DII expression in thyroid, eye and skeletal muscle tissues by immunohistochemistry using immunized guinea pig and patients sera with Graves’ orbitopathy. Decreased DII activities were measured after adding proinflammatory cytokines and patients sera with hyperthyroid Graves’ orbitopathy and systemic sclerosis. Antibodies to DII inhibited the mitogen-activated protein kinase (MAPK) activation in thyroid tissue. Proinflammatory cytokines (IL-6, TNFα, IFNγ) inhibited thyroid DII activities in dose-dependent manner (Vmax: 4.1×10-3 pmol/mg/min for IL-6; 0.18 pmol/mg/min for TNFα; 0.23 pmol/mg/min for IFNγ). Hyperthyroid patient sera with Graves’ orbitopathy decreased better thyroid DII activities than eye muscle DII ones (3.99±5.79 vs. 7.66±10.49 pmol/mg/min, P<0.05, n=26). Patient sera with systemic sclerosis (SSc, n=19) decreased DII activities compared to those in controls (n=16) (4.99±1.04 vs. 2.88±0.61 pmol/mg/min, P<0.0001). Immunized guinea pig and Graves’ patient sera with anti-DII antibodies resulted in relevant inhibition of MAPK activation. In conclusion, DII protein can be a new autoantigen in thyroid autoimmunity, particularly in Graves’ orbitopathy. DII activity blocking cytokines could be responsible for low FT3 levels causing euthyroid sick syndrome in systemic sclerosis. The difference in tissue-specific DII activities could be implicated in the development of orbitopathy in hyperthyroid Graves’ disease.

Biography:

Rongtuan Lin is an Associate Professor in the Department of Medicine at McGill University and a Project Director of the Molecular Oncology Group at the Lady Davis Institute for Medical Research. He has received his PhD from Concordia University and completed Post-doctoral training at the Lady Davis Institute for Medical Research. He made important contributions in the fields of interferon signaling and innate antiviral immunity. He has a highly successful laboratory research program with 100 scientific publications, which have been cited more than 5,500 times. He was a recipient of a Chercheur-boursier Senior and Junior 2 from Fonds de la Recherche en Sante du Quebec. In 1996 and 1998, he has received the Milstein Young Investigator Award from the International Society for Interferon and Cytokine Research.

Abstract:

Oncolytic viruses (OVs) are novel anticancer agents that infect and effectively kill cancer cells but not normal cells. Although tumor growth is delayed or eliminated in numerous animal models following treatment with OVs, several cancer models remain partially or completely resistant to viral oncolysis. To overcome this resistance, experimental strategies are now combining OVs with different cytotoxic compounds to improve OV efficacy. Our laboratory has previously demonstrated that OV replication can be bolstered by co-administration of other chemical agents such as Triptolide, a natural molecule derived from the medicinal herb. In the current study, we investigated the capacity of sulforaphane (SFN); an anti-cancer compound naturally occurred in cruciferous vegetables with demonstrated potent antioxidant and possible anti-inflammatory actions to enhance vesicular stomatitis virus (VSV) oncolysis in OV-resistant cancer cells. We ultimately demonstrate that the resistant PC3 prostate cancer cell line can be sensitized to VSV by addition of SFN. Indeed, SFN dose-dependently enhances the replication of VSV. Neither VSV (MOI 0.1) nor SFN (20 uM) alone are toxic against PC3 cells in vitro; however, in combination they greatly increased the oncolytic capacity of VSV by reducing cancer cell viability and promoting apoptosis-mediated cell death. Furthermore, the potentiation of VSV oncolysis by SFN is dependent on the production of ROS and is associated with the induction of autophagy. SFN is known to induce phase II antioxidant genes via Nrf2 activation, which regulates ROS levels and stimulates autophagy in prostate cancer cells. Mechanistically, SFN inhibited the innate antiviral response by blocking the type-1 interferon (IFN) signaling pathway, through the activation of the Nrf2 transcription factor. Exogenous Nrf2 expression inhibits Interferon-Stimulated Response Element (ISRE) promoter activity in a dose dependent manner following virus infection or IFN treatment. Taken together, these results demonstrate for the first time the synergic effect of SFN and VSV and indicate that SFN treatment increases VSV replication and the subsequent apoptosis of tumor cells by inhibiting IFN signaling. We are currently investigating the molecular mechanism involved in VSV-induced oncolysis by Nrf2 activators and evaluating the therapeutic potential of the combination of OV and Nrf2 activators in a mouse model of prostate cancer.

Biography:

Meltem Elitas is a Faculty Member in Mechatronics Program at Sabanci University. Her background is in Electrical and Mechatronics Engineering. She has obtained her Doctorate from Bioengineering and Biotechnology Department at Ecole Polytechnique Federale de Lausanne. She has performed her Postdoctoral studies at Yale University Biomedical Engineering Department. She has published more than 25 papers in reputed journals. Her research interests are biomechatronics, cellular heterogeneity, cellular interactions, tumor microenvironment, live cell imaging and development of microfabricated tools for quantitative biology.

Abstract:

Understanding the interactions between tumor cells and immune cells in a quantitative manner will provide valuable information to reveal the mechanism of diseases, immune defense and development of new treatment reagents and strategies for the diseases. Today one of the biggest limitations relies on the traditional methods and tools that we use to investigate the rare cells and specific events in biology particularly in immunology. Since these techniques are not adequate enough to be selective, specific and quantitative, the rare cells such as the metastatic or drug resistant ones or the events such as onset symptoms of tumors and infections are being masked by majority of the cells or events in the population. Therefore, we cannot diagnosis on time or provide successful strategies. As a consequence, our approaches might not target the right cells at the right time in the right place. To overcome these limitations, we might profit from engineering approaches and tools. We can develop quantitative, accurate, reproducible and precise methods and use microfabricated tools to understand the nature and behavior of rare cells and events. The improvements from microfabricated tools in conjunction with microscopy might provide statistics from large numbers of single cells, short assay time, less sample consumption, less waste production, quantitative and reproducible data, single-cell resolution images, high-throughput, spatio-temporal tracking and real-time assays, etc. This talk will present recently developed microfabricated tools to understand the immune cell-tumor cell interactions. I will present our microfluidic applications and their preliminary data from my research group.

Biography:

Amina Dahmani is currently a PhD candidate in Microbiology-Immunology at Université de Montréal, Canada. She has completed her Master degree in Immunology at Univérsité Laval, Canada, in Cellular Therapy Lab directed by Dr Jacques P Tremblay, where she studied the development of immunological tolerance to allogeneic myoblasts transplantation as potential therapy for Duchenne Muscular Dystrophy. Later she has joined Dr Jean-Sébastien Delisle team's, dedicated to cancer and viral adoptive immunotherapy to complete her PhD. She is currently working to improve adoptive immunotherapy protocols.

Abstract:

Adoptive immunotherapy (AI) has emerged as a potentially curative therapy for advanced cancer and infections. Recent findings suggest that the transfer of T-cells with “early” memory features may improve the therapeutic potential of AI. TGF-β is a pleiotropic cytokine that controls a large spectrum of biological and pathological processes. In T-cell biology, TGF-β is mostly known for its immunoregulatory properties, but recent evidence has revealed a novel role of TGF-β in T-cell memory differentiation and maintenance. Thus, we investigated whether TGF-β could promote features of memory in ex vivo stimulated human T-cells to further improve the efficacy of clinical protocols for AI. Here we show that agonistic TGF-β stimulation leads to the expression of central memory markers without significantly altering T-cell expansion or polyfunctional cytokine secretion following stimulation. Furthermore, TGF-β exposure decreased expression of transcription factors responsible for effector differentiation (T-BET, GATA3 and BLIMP1) and increased those associated with memory differentiation, notably ID3. The knock-down of ID3 by specific siRNA revealed that TGF-β-driven T-cell memory differentiation largely depends on ID3. Moreover, TGF-β-exposed T-cells showed enhanced persistence, expansion and alloreactivity after adoptive transfer into NSG mice. Finally, using clinically relevant culture methods to generate T-cell lines against viral and tumor antigens, we found that TGF-β programmed the expression of early memory markers without significantly curtailing T-cell expansion or antigen-specificity. This finding provides a rationale for clinical use of TGF-β to optimize memory phenotype of ex vivo pathogen/antigen-specific T-cells expanded for AI.

Break: 12:55-13:40 Lunch Break @ Element I+II Restaurant

Omar El Bounkari

Klinikum der Universität München, Germany

Title: The innate chemokine MIF goes adaptive in atherosclerosis

Time : 13:40-14:00

Biography:

Omar El Bounkari worked in Klinikum der Universität München, Germany

Abstract:

Macrophage migration inhibitory factor (MIF) proteins (MIF and MIF-2) are chemokine-like inflammatory mediators with unique structural properties distinct from classical chemokines. MIF proteins play arole in the control of both physiological and pathophysiological immune responses. With MIF-2 only very recently identified, most evidence currently is available for MIF. In fact, MIF is a pivotal upstream mediator of innate immunity, while some contribution to the adaptive response has been reported. When dysregulated, MIF is an exacerbating promoter of several inflammatory diseases including atherosclerosis, a chronic inflammatory condition of large and medium-sized arteries and the major underlying cause of cardiovascular morbidity and mortality worldwide. MIF orchestrates the atherogenic recruitment of monocytes/macrophages and T lymphocytes through non-cognate interaction with the CXC chemokine receptors CXCR2 and CXCR4, respectively, and contributes to the inflammation and destabilization in atherosclerotic lesions. These processes have been considered as the effects of an innate chemokine on innate inflammatory cells in the atherosclerotic lesion area.Here we show that MIF also controls adaptive immune cells in atherosclerotic pathogenesis. We present data that MIF is a novel B cell chemokine that promotes B cell migration and proliferationvia the chemokine receptors CXCR4 and CXCR7 as well as CD74, the surface form of MHC class II invariant chain. MIF-driven B cell responses are mediated through the ZAP-70 and ERK1/2 signaling pathways and encompass the activation of calcium transients. We also studied the impact of Mif gene deletion in the proatherogenicApoE-/- genetic background in mice and have unraveled a surprising atherogenic phenotype with a previously unrecognized link between MIF and B cell pathobiology. This suggeststhat MIF could be a potential therapeutic target to induce protective B cell responses in such diseases.

Biography:

Denis Soulet has completed his PhD in Neuroimmunology from Laval University, Canada and Postdoctoral studies from Ycee Claude Bernard, Sweden. He is an Associate Professor at the Medicine Faculty of Laval University, Canada. He has published more than 40 papers in reputed journals and has been serving as an Editorial Board Member of SM Journal of Gastroenterology and Hepathology. He is leading a research team dedicated to study the role of peripheral inflammation in the enteric nervous system and its contribution to Parkinson’s disease. The ultimate goal of his research program is to design immunomodulatory based disease modifying drugs for PD.

Abstract:

Parkinson’s disease (PD) is a neurological disorder characterized by motor symptoms which are often preceded by non-motor symptoms, including gastrointestinal dysfunctions. Common treatments are only symptomatic; there is still no disease modifying drug available to cure patients. Since numerous pro-inflammatory markers have been measured in the central and peripheral nervous system, this deleterious immune response seems to be a potential target to develop new therapeutic strategies. Therefore, a better understanding of the role of the immune response in the etiology and progression of PD is essential. During my talk, I will present original data about the impact of the innate immune response on enteric neuronal damage in PD models. At first, we characterized the immune response induced by the neurotoxin MPTP in the enteric nervous system of partially immunodeficient mice. We demonstrated the timeline of inflammatory events occurring prior to the neuronal demise and the critical role of monocytes and macrophages in the gut. Thereafter, we tested various estrogenic compounds for their immunomodulatory and neuroprotective properties in PD models both in vivo and in vitro, delineating the major contribution of various estrogenic receptors, mainly the G Protein-coupled Estrogen Receptor 1 (GPER1). More recently, we successfully explored the therapeutic potential of a clinically approved selective estrogen receptor modulator, Raloxifene, for drug repurposing in PD. In conclusion, our data highlight the critical role of the immune response at early stages of PD and the immunomodulatory and neuroprotective potential of estrogen-based hormonotherapy at the pre-clinical level.

Speaker
Biography:

Ganapathi Bhat Mugulthimoole is a Senior Consultant Medical Oncologist & Stem Cell Transplant Physician at Jaslok Hospital & Research Centre. He has completed his graduation in Medicine in 1993 and Post graduation in General Medicine in 2000. He has further trained in the field of oncology in various institutions in India and later gained expertise in stem cell transplant while working in Kuwait Cancer Control Centre (2002-2006). He has gained specialized training in stem cell transplantation as part of the ESH-EBMT (2007), 2011 (Labaule, France) and ICAS training program (2009) from Ulm University, Germany. He has numerous academic articles published in Indian, international journals and textbooks to his credit.

Abstract:

The tumor microenvironment is a principal feature of cancer biology that supports the initiation and progression of the tumor and response to therapy. Cells and molecules of the immune system are the essential elements of the tumor microenvironment. Therapeutic stratagems can harness the immune system to precisely target tumor cells by possibly stimulating tumor-specific immunological memory, which may lead to long term regression. Understanding the complexity of immunomodulation by tumors is important for the expansion and promotion of immunotherapy. Several approaches are being carried out to augment the anti-tumor immune responses. Among them are immunotherapeutic vaccines, adoptive cell transfer therapies and checkpoint blocking drugs. However, cancer cells try to evade the immune watchdogs by reducing surface tumor antigen or inducing cells that express certain proteins that affect immune cell inactivation or by promoting tumor proliferation and survival. Gaining a deeper understanding of the tumor immunogenicity by employing advanced techniques such as sequencing of the tumor DNA, will help to better address the challenges and gain an appreciation of the delicate association between cancer and our immune systems.

Biography:

Moshe Giladi has joined Novocure in 2005 and served as the Head of the NovoBiotic project until 2008. He was then promoted to Head of Novocure's Preclinical Research leading a team of experts of various fields: Cancer, immunology, cell biology and also responsible for research collaboration with academic institutes. He leads research activities studying tumor treating fields mechanism of action. He has received his PhD in Life Sciences from the Department of Molecular Microbiology and Biotechnology, Faculty of Life Sciences and his MBA from the Leon Recanati Graduate School of Business Administration both at the Tel Aviv University, Israel.

Abstract:

Tumor treating fields (TTFields) are an effective anti-neoplastic treatment modality delivered via non-invasive application of low intensity, intermediate frequency, alternating electric fields. This therapy is approved for the treatment of patients with glioblastoma. Previous investigations have shown that TTFields disrupt microtubules and septin filaments, both of which govern key roles in mitosis. Some of the outcomes of mitosis under TTFields application include abnormal chromosome segregation and ER stress which trigger different forms of cell death. The goal of this study was to evaluate whether TTFields induced cancer cell death can be perceived as immunogenic by the immune system and to explore the possibility of combining TTFields with immune-modulating drugs. Murine Lewis lung carcinoma (LLC) and ovarian surface epithelial (MOSE) cells were treated with TTFields using the inovitro system. The exposure of calreticulin (CRT) on the surface of treated cells was evaluated using flow cytometry. High-mobility group box 1 protein (HMGB1) secretion was measured using ELISA assay. For in vivo experiments, immunocompetent C57BL/6 mice were orthotopically implanted with LLC cells and treated with TTFields, anti-PD-1 or combination of the two modalities. Changes in tumor volume were monitored and flow cytometry analysis was performed for phenotypic characterization of tumor infiltrating immune cells. We demonstrate that application of TTFields leads to the exposure of CRT on the cell surface and also promotes release of HMGB1 from cancer cells in vitro. In vivo, the combined treatment of TTFields and anti-PD-1 led to a significant decrease in tumor volume compared to control group and to animals treated with anti-PD-1 alone. An increase in CD45+ tumor infiltrating cells was observed in both anti-PD-1 and TTFields+anti-PD-1 groups although statistical significance was reached only in the combination treatment group. Interestingly, CD45+ cells from the combination treatment group also demonstrated a significant upregulation of PD-L1 expression on the cell surface. Specifically, this upregulation in the PD-L1 expression was observed in both F4/80+CD11+ cells (macrophages) and CD11c+ cells (dendritic cells) whereas no significant effect on the infiltration pattern of these immune cell populations was noted. Taken together, our results demonstrate that TTFields application potentiates immunogenic cell death in cancer cells and that combining TTFields with specific immunotherapies such as anti-PD-1 might achieve tumor control by further enhancing antitumor immunity.

Tali Feferman

Weizmann Institute of Science, Israel

Title: Hypoxia limits CTL functions: Lessons from live intratumoral imaging

Time : 15:00-15:20

Biography:

Tali Feferman has completed her PhD in Molecular Biology at the Macquarie University and Postdoctoral Training at The Heart Research Institute in Sydney. She has then joined the Weizmann Institute of Science in 2000. Her initial interest involved exploring the mechanism of action and immunomodulation of Myasthenia Gravis (MG) and its model experimental autoimmune MG (EAMG). In recent years her interest is mainly focused on addressing questions important for optimizing cancer immunotherapy using cutting-edge imaging microscopic techniques in live mice.

Abstract:

Poor tumor vascularization is an obstacle to immunotherapy by CTLs. It impairs tumor infiltration but also introduces hypoxia, known to interfere with T-cell migration. It is yet unknown how suboptimal vascularization affects CTL migration and function within tumors. To study this question, we combined immunohistochemistry of human melanoma samples with two-photon imaging in live mice. Orthotopically implanted B16-OVA tumors were studied after adoptive transfer of in vitro matured antigen-specific OT-I CTLs. In patients, CD8 T-cells concentrated around peripheral vessels in the tumor and sparsely infiltrated avascular areas. In mice, CTLs crawled rapidly in oxygenated areas within 50 µm of flowing blood vessels. Occluding intratumoral blood vessels triggered immediate arrest of CTL motility, which was quickly reversed when flow was resumed. Immunohistology indicated that CTLs avoided hypoxic tumor areas. Live CTL imaging in vitro showed deceleration under hypoxic conditions and when oxidative phosphorylation was blocked. To circumvent intratumoral CTL dysfunction we attempted to increase vascular density by implanting tumors in matrices containing bFGF. bFGF-laced tumors were more easily rejected after transfer of CTLs and displayed delayed growth in untreated mice but were not affected in mice deficient in CD8 T-cells. CTLs infiltrated such tumors in normal numbers but displayed enhanced motility in highly vascular tumors, suggesting that enhanced rejection resulted from improved intratumoral CTL migration. Taken together, the results suggest that hypoxia limits CTL function away from blood vessels and that alleviating it may synergize with immunotherapy.

Biography:

Hisham Abd El Dayem is presently working in Ain Shams University, Egypt.

Abstract:

Purpose: To compare expression of multidrug-resistant protein 1/P-glycoprotein (MDR1/Pgp) in retinoblastoma in eyes treated by primary enucleation due to advanced tumor at initial presentation and those enucleated after being resistant to chemotherapy. Methods: This study was a prospective study. Twenty retinoblastoma patients presented to Retinoblastoma Clinic at Ophthalmology Department, Ain Shams University Hospitals. All patients had enucleation and were divided into 2 groups. Patients in group-1 underwent primary enucleation due to advanced tumor at presentation. Patients in group-2 underwent secondary enucleation after failure of conservative treatment. Immunohistochemical studies were performed searching for expression of multidrug-resistant protein 1/P-glycoprotein (MDR1/Pgp) in the two groups. Results: Analysis of the primary enucleation group showed high positive, low positive and negative expression in 1 (10%), 2 (20%) and 6 cases (70%) respectively. In secondary enucleation group: 5 cases (50%), 3 cases (30%) and 2 cases (20%) showed high positive, low positive and negative expression respectively. Conclusions: This pilot study though, not being able to demonstrate statistical significance in MDR1 expression in primary enucleated vs. secondary enucleated resistant cases, demonstrated p-value low enough to indicate a trend for more MDR1 expression in resistant cases (P=0.068). Further study with a larger sample size is warranted.

Break: 15:40-15:55 Coffee Break @ Main Lobby
Biography:

Nan-Shan Chang is currently the Professor and Director of the Molecular Medicine Institute, National Cheng Kung University (NCKU) in Taiwan, and the Adjunct Professor with the SUNY Upstate Medical University and the NYS Institute for Basic Research in Developmental Disabilities, New York. Dr. Chang is most noted for his discovery of tumor suppressor WWOX in 2000. Recent Awards: Breast cancer and neurofibromatosis research awards from the Department of Defense, USA, in 2008 and 2010; NCKU Distinguished Professor Award 2010, 2013 and 2016; Distinguished Scientist Award 2011 from the Society of Experimental Biology & Medicine, USA.

Abstract:

Whether tumor suppressor WWOX stimulates immune cell maturation is largely unknown. Here, we determined that Tyr33-phosphorylated WWOX physically binds non-phosphorylated ERK and IκBα in immature acute lymphoblastic leukemia MOLT-4 T cell and in the naïve mouse spleen. The IκBα/ERK/WWOX complex was shown to localize, in part, in the mitochondria. WWOX prevents IκBα from proteosomal degradation. Upon stimulating MOLT-4 with ionophore A23187/phorbol myristate acetate (IoP), endogenous IκBα and ERK undergo rapid phosphorylation in less than 5 min, and subsequently WWOX is Tyr33 and Tyr287 de-phosphorylated and Ser14 phosphorylated. Three hr later, IκBα starts to degrade and ERK returns to basal or non-phosphorylation, and this lasts in the next 12 hr. Finally, expression of CD3 and CD8 occurs in MOLT-4, along with re-appearance of the IκBα/ERK/WWOX complex near 24 hr. Inhibition of ERK phosphorylation by U0126 or IκBα degradation by MG132 prevents MOLT-4 maturation. By time-lapse FRET microscopy, IκBα/ERK/WWOX complex exhibits an increased binding strength by 1-2 fold after exposure to IoP for 15-24 hrs. Meanwhile, a portion of ERK and WWOX relocate to the nucleus, suggesting their role in the induction of CD3 and CD8 expression in MOLT-4. (Supported by MOST and NHRI, Taiwan, and DoD, USA)

Speaker
Biography:

Miao Dong is currently pursuing PhD in City University of Hong Kong. She has obtained her Bachelor degree from Liaoning University, China, majoring in Ecology and completed her Master degree from Shandong Agricultural University. During three years study, she investigated antioxidant defense system in zebrafish and has 6 publications (3 of 6 are as first author). During her PhD study, she has done research on interaction between innate immune proteins in fish blood and bacteria, also the collection of natural occurring antimicrobial peptides from medaka fish blood. The related manuscript has been submitted to JBC.

Abstract:

The excessive use of antibiotics in aquaculture contributes to the uprising of antibiotic resistance that threatens human health. We explore the innate immunity of fish for naturally occurring antimicrobial factors that can be developed into potential antibiotic agents. Antimicrobial peptides (AMPs) have been studied in many organisms but efforts on the systematic identification of AMPs in fish have been lacking. In this study, we systematically identified naturally occurring peptides in medaka plasma. Blood collected from medaka of different gender, age and infection status were combined and thereby providing a resource of plasma macromolecules under various possible physiological conditions. Peptides under the molecular weight of 3 kDa were fractionated and purified followed by mass spectrometry analysis. In total, 6483 unique peptides were identified against the medaka genome, constituting a database of circulating peptides in this organism. After evaluation with a combination of web based prediction tool and conserved physicochemical properties of AMPs, 83 potential antimicrobial peptides were predicted. One of them, a 13-residue peptide named VPS13D3241-3253, showed broad spectrum toxicity on fish and human pathogenic bacteria (Gram positive or Gram negative) without significant cytotoxicity on mammalian cell lines. Scanning electron microscopy indicated that VPS13D3241-3253 disrupted the cell wall of both Gram positive and negative bacteria. SOS response assay showed that this peptide efficiently induced DNA damage in bacteria. The identification of VPS13D3241-3253 illustrates the feasibility of the proteomic approach in the discovery of potentially novel AMPs from fish. These AMPs will form an important basis for the development of new antibacterial agents in the fishery.

Biography:

Anne Stinn is currently a PhD student at the Max Planck Institute for Infection Biology in Berlin, Germany. From 2008 to 2013 she studied Biology at the Justus Liebig University in Giessen, Germany. After completing her study she went to London as an Intern in the Research Group of Cell Death, Cancer and Inflammation (CCI) at the UCL Cancer Institute. She has started her PhD in the year 2014.

Abstract:

Acute myeloid leukemia (AML) is a highly malignant cancer of the myeloid cell lineage that is characterized by the rapid growth of abnormal white blood cells. Although AML is a relatively rare disease accounting for about 1% of all cancer cases (US), it is the type of leukemia showing the lowest survival rate. In the majority of all AML cases mutations in the kinase domain of the FMS-like tyrosine kinase III receptor (FLT3; CD135) are reported. Besides treatment based on chemo and radiation therapy as well as bone marrow transplantation, bispecific antibodies are studied for the use in immunotherapy against AML. These antibodies recognize tumor-associated antigens (TAAs) as well as the agonistic T-cell receptor/CD3 complex (TCR/CD3) and should thereby lead to a tumor cell-restricted activation of immune cells and specific lysis of cancer cells. In this study a structural analysis of the bispecific antibody NF-CU, the first known to date was performed. In addition to the structure of the NF-CU itself, the crystallographic structure of the antibody bound to FLT3 and CD3 was investigated. Deciphering the crystal structure of the antibody-antigens complex should give an inside into epitope recognition as well as the molecular mechanism leading to T-cell activation and tumor cell death.

Biography:

Maryam Golshani was graduated from the Pasteur Institute with a degree in Medical Bacteriology. Presently, she is a Post doctorate fellow and Junior Research Group Leader at IPI working on new Brucella vaccine candidates. Her research mainly focuses on in silico investing the immunogenicity of new vaccine targets and in vivo evaluating their protective efficacy against Brucella infection. She has involved in more than 11 projects and published 11 papers in reputed journals.

Abstract:

Objectives: Brucellosis is the most common bacterial zoonosis worldwide and no safe and effective vaccine is available for the prevention of human brucellosis. In humans, brucellosis is mostly caused by Brucella melitensis and Brucella abortus. According to our in silico studies, Omp2b is predicted to be potentially immunogenic antigen conserved in main Brucella pathogens. The aim of this study was to design truncated form of Omp2b and to evaluate the immunogenicity and protective efficacy of a recombinant protein vaccine encoding tOmp2b. Methods: Bioinformatics tools were used to design the truncated protein based on conserved domains and regions of epitopes with strong affinity for MHC molecules. The humoral/cellular immune response and protection levels against challenge with wild B. melitensis and B. abortus infections were evaluated in rtOmp2b+ adjuvants immunized mice and control groups. Results: Vaccination of BALB/c mice rtOmp2b provided the significant protection level against both B. melitenisis and B. abortus. Moreover, rtOmp2b elicited a strong specific IgG response (higher IgG2a titers) and significant IFN-γ/IL2 production. Conclusion: According to the results, rtOmp2b is able to induce cross-protection against B. melitensis and B. abortus infections. Therefore, it could be a new potential candidate for the development of Brucella subunit vaccines.

Biography:

Jalil Mehrzad has completed his PhD at the age of 32 years from Ghent University, Faculty of Veterinary Medicine, and postdoctoral studies from McGill University. He is an assciate professor of Immunology in Ferdowsi University of Mashhad (next year will move to Tehran University as full-time scientific member of departmet of Microbiology and Immunology). With H-index and citations of 16 and 1315, respectively, he has published more than 45 papers in reputed journals and has been serving as regular reviewer for many journals in the area of immunobiology, molecular biotechnology and medicine.

Abstract:

Caspases-mediated apoptosis/cell death activation is key regulatory response in many physiopathological conditions. Application of bioluminescence and the reaction of luciferase would provide a powerfully novel in vitro/vivo assay for apoptosis detecion. As key brain immune cells, astrocytes and microglials, are vital part of the central nervous system (CNS); they are the main responder to inflammation in CNS; any disruption on their function would lead to CNS damage. Aflatoxin B1 (AFB1) is commonly found in foodstuffs, and can be the cause of many diseases including cancer. AFB1 and its metabolites cause oxidative stress in especially the CNS-derived cells, adversely affecting their normal activities, thus leading to the neurodegenerative diseases including multiple sclerosis (MS), Alzheimer’s and Huntington’s diseases. Considering the importance of astrocytes and the inevitable existence of AFB1 in the feed/foods, worldwide, the study of astrocytes-AFB1 interactions is valuable. We therefore investigated the impact of AFB1 on the apoptosis of one of the key accessory supportive CNS, astrocytes, using several biochemical experimentations including intracellular ATP and caspases 3/7 measured by bioluminescence and luciferase reactions. The release of cytochrome c and apoptosis/necrosis of AFB1-treated astrocytes with various concentration of AFB1 and exposure time was also tested using Western blotting and flow cytometry techniques, respectively. Bioluminescence results revealed decreased intracellular ATP, increased caspases 3/7 activities, cytochrome-c release and apoptotic/necrotic of astrocytes particularly at higher timepoints and doses of AFB1. Considering the broad roles of astrocytes in CNS, this finding deepens our understanding of the molecular mechanisms and functional consequences of the neural cells damage neurotoxicity triggered by AFB1 exposure in mammals.