Research team

Expertise

Flow cytometry is a widely used technique that allows the simultaneous and multi-parameter analysis of physical and biochemical characteristics of a population of living cells or particles in a heterogenous sample. The Laboratory of Experimental Hematology (LEH) has 20+ years of demonstrable experience in flow cytometry (200+ published manuscripts), as well as experience in guidance and support of both internal and external research groups with flow cytometric experiments (30+ joint manuscripts). As a flow cytometry and cell sorting core facility at the AUHA, our ambition is to make basic and advanced flow cytometry available for all active and prospective users at the AUHA in order: (i) to leverage qualitative cell biological and (pre)clinical cellular research, (ii) to provide qualitative education covering flow cytometry and its applications over multiple faculties (FGGW, FBD and FWET), and (iii) to provide external service using flow cytometry as a basis, both intellectually as well as practically.

Where multiple sclerosis strikes: discovery of target antigens involved in MS pathogenicity, with emphasis on molecular mimicry and epitope spreading. 15/10/2023 - 14/10/2025

Abstract

More than 2.8 million people worldwide are living with multiple sclerosis (MS). This disabling disease is caused by an autoimmune reaction directed against myelin proteins. However, the specific myelin epitopes that MS targets are not yet known. This poses an obstacle to the development of antigen-specific therapies. Currently, most therapies for MS are immunomodulatory, and although they alleviate the symptoms of the disease, they also increase the risk of opportunistic infections. Therefore, this project seeks to identify the target antigens involved in MS to pave the way for the development of antigen-specific therapies. For this purpose, we will use human dendritic cells (DCs) loaded with myelin lysate or lysate from Epstein Barr Virus (EBV)-infected cells. The DCs are loaded via phagocytosis, reflecting the in vivo process of antigen-loading of DCs. After antigen loading, the lysate will be processed and presented by the DCs on major histocompatibility complexes (MHC). The epitopes presented will be identified by immunopeptidomics, a recently developed technique using mass spectrometry. The pathogenicity of the identified epitopes will then be studied in an in vivo mouse model, while the presence of T cells specific for these epitopes in the blood of MS patients will be examined with IFN-gamma ELISpot assay.

Researcher(s)

Research team(s)

Project type(s)

  • Research Project

A controlled phase II clinical trial evaluating efficacy of myelin peptide-loaded tolDC as treatment for MS. 01/10/2023 - 30/09/2027

Abstract

Multiple sclerosis (MS) is an inflammatory neurodegenerative disease of the central nervous system for which no cure is available. It is the leading cause of non-traumatic disabling neurological disease in young adults with more than 6.500 people affected in Flanders. Since MS strikes during the primary productive time of one's personal and professional life, it leads to a major physical and socio-economic burden to the patient, family, and society. Therefore, new therapeutic interventions with improved efficacy over existing drugs and good tolerability are needed. As chronic inflammatory processes drive the neurodegeneration, we hypothesize that improved clinical outcome depends on restoring the balance between inflammation and the remaining capacity of neuronal self-renewal. Therefore, cell therapy that specifically targets the damaging immune reactions that cause MS and induce disease-specific tolerance without affecting protective immunity against pathogens and cancer is a promising approach. Recently, a collaborative network of European centers joined efforts to bring antigen-specific therapy for MS to the clinic. Two single-center phase I clinical trials evaluating the use of antigen-specific tolerance-inducing dendritic cells (tolDC) in MS patients were conducted (previously funded by IWT- TBM and H2020). No serious adverse events were observed. Next, we aim to demonstrate efficacy of tolDC treatment in a phase II clinical trial in patients with MS. Coordinated patient and MRI monitoring, including radiological correlates of neurodegeneration, and immunomonitoring will enable us to demonstrate efficacy of tolDC administration and to support future efforts in the field of MS therapy. An effective therapy that lowers morbidity with reduced occurrence of side effects and less frequent hospitalizations will enhance quality of life of patients as well as dramatically reduce economic burden. This would represent a breakthrough for healthcare in MS.

Researcher(s)

Research team(s)

Project type(s)

  • Research Project

Development of a CD19 CAR T cell therapy for multiple sclerosis. 01/01/2023 - 31/12/2024

Abstract

There is a clear unmet need for innovative treatments that can suppress ongoing inflammatory disease activity in treatment refractory RRMS patients. The grant of the Belgian Charcot Foundation enables us to start developing CD19 CAR T cells for the use in MS and related autoimmune diseases that are driven by pathogenic B cells. While this project is focused on laboratory research, we are committed to paving the road towards a first-in-man clinical trial in the future.

Researcher(s)

Research team(s)

Project website

Project type(s)

  • Research Project

Towards an Off-the-Shelf Therapy: Tolerogenic Dendritic Cell-derived Extracellular Vesicles for the Treatment of Multiple Sclerosis. 01/12/2022 - 30/11/2024

Abstract

MS is a chronic auto-immune disorder of the central nervous system (CNS) and the leading cause of non-traumatic disabling disease in young adults. Although the exact cause of MS remains to be elucidated, it is currently accepted that both genetic and environmental factors affect complex immunological responses, both in the periphery and the CNS. To date, there is still no cure for MS, but several immune-modifying and/or -suppressive treatments, especially targeting the peripheral immune system, have been developed over time. However, these have varying efficacy, have limited long-term effectiveness, and sometimes life-threatening side effects, thereby underscoring the urgent need for novel therapeutic approaches to be developed and evaluated. Tolerogenic dendritic cells (tolDCs) are professional antigen-presenting cells with immunosuppressive properties, priming the immune system into a tolerogenic or unresponsive state against various (self)antigens. TolDCs are essential in maintenance of central and peripheral tolerance through induction of T cell clonal deletion, T cell anergy, generation and activation of regulatory T-cells (Tregs), as well as the direct modulation of pro-inflammatory environments. For that reason, tolDCs show considerable promise as candidates for specific cellular therapy for treatment of allergic diseases, autoimmune diseases or transplant rejections. In this context, the Laboratory of Experimental Hematology (LEH, Cools' team) recently recruited nine patients for a phase I clinical trial evaluating the potential safety and feasibility of tolDCs for the treatment of multiple sclerosis (MS) (NCT02618902). While no serious adverse events are observed in all treated patients, much remains to be understood about the exact molecular and cellular interactions these therapeutic cells provoke in vivo. Accumulating evidence shows that extracellular vesicles (EVs) secreted by immune cells play a key role in intercellular communication. In this project, we aim to determine the immune-regulatory mechanism by tolDCs, hypothesising that it would be mediated by EVs and their immunosuppressive cargo. To achieve this goal, we will apply an unbiased multi-omics approach, both in vitro and in vivo, to unravel the therapeutic potential of tolDC-derived EVs. We hereby anticipate that our findings will lead to ground-breaking insights on current understanding of EVs in immune-regulatory therapy and ultimately will lead to the development of a novel (non-cellular) off-the-shelf therapeutic compound to be evaluated in patients suffering from detrimental auto-immune disorders, including MS. This highly innovative application addresses the use of tolDC-derived EVs as disease-modifying treatment in MS and is expected to provide new insights into how immune tolerance is initiated following interaction of key immune-regulatory cargo of the EVs with peripheral and CNS immune cells. With this project, we present a clinically relevant project relying on inventive fundamental research with a high translational value and valorization potential. The integrative multiomics analysis will give a better insight in the molecular pathways involved in the induction of tolerance and immunoregulation by tolDCs. Furthermore, this project could lead to the development of a cell-free therapy based on tolDC-EV for the treatment of MS, which can surpass drawbacks associated with cell therapy. In addition, the possibility of allogenic exosome therapy would result in a more positive cost-benefit ratio since an "off-the-shelf" product is less expensive than an individualized cell therapy. Hence, the study proposed here is merely the beginning of numerous possible new research questions as well as clinical translation.

Researcher(s)

Research team(s)

Project type(s)

  • Research Project

Towards an Off-the-Shelf Therapy: Tolerogenic Dendritic Cell-derived Extracellular Vesicles for the Treatment of Multiple Sclerosis. 01/11/2022 - 30/09/2025

Abstract

Multiple Sclerosis is a complex neurodegenerative disease of the central nervous system (CNS), currently affecting almost 15 000 people in Belgium. To date, there is still no cure for MS, but several immune-modifying treatments have been developed. The use of tolerogenic dendritic cells (tolDC) for the treatment of MS is currently being investigated. These tolDC can modulate the immune response and (re)establish self-tolerance. However, their exact working mechanism has not been fully elucidated yet. In this project, we hypothesize that tolDC modulate the auto-reactive response via extracellular vesicles (EV). EV are nanosized membrane vesicles that are released by almost every cell type and have been reported to be involved in immune regulation. In particular, the cargo carried by these EV can influence the immune response. Indeed, the cargo compromising of functionally active compounds such as RNAs, lipids, metabolites, and proteins can alter the phenotypic and functional properties of the recipient cells. Hence, we anticipate a role of immunomodulatory cargo-containing EVs in the mode-of-action of tolDC. For this, we aim to explore the immunosuppressive properties of tolDC-derived EV and their capacity to establish tolerance. This research would contribute to a better understanding of the working mechanism of tolDC. In addition, results could lead to the development of a cell-free therapy for MS surpassing the drawbacks associated with cell therapy.

Researcher(s)

Research team(s)

Project type(s)

  • Research Project

Flow Cytometry and Cell Sorting Core Facility Uantwerpen (FACSUA) 01/01/2022 - 31/12/2026

Abstract

Flow cytometry is a widely used technique that allows the simultaneous and multi-parameter analysis of physical and biochemical characteristics of a population of living cells or particles in a heterogenous sample. The Laboratory of Experimental Hematology (LEH) has 20+ years of demonstrable experience in flow cytometry (200+ published manuscripts), as well as experience in guidance and support of both internal and external research groups with flow cytometric experiments (30+ joint manuscripts). With this application, we now aim to maintain and expand a flow cytometry and cell sorting core facility at the AUHA. The ambition of LEH is to make basic and advanced flow cytometry available for all active and prospective users at the AUHA in order: (i) to leverage qualitative cell biological and (pre)clinical cellular research, (ii) to provide qualitative education covering flow cytometry and its applications over multiple faculties (FGGW, FBD and FWET), and (iii) to provide external service using flow cytometry as a basis, both intellectually as well as practically.

Researcher(s)

Research team(s)

Project type(s)

  • Research Project

VAXINFECTIO-PD _ VAXINFECTIO: Vaccine & Infectious Disease Institute _ PD: Product Development. 01/01/2021 - 31/12/2026

Abstract

VAXINFECTIO-PD is an established Industrial Research Fund (IOF) consortium, well equipped to build an ecosystem offering research, valorisation, innovation and development to answer existing and new challenges in the field of infectious diseases and vaccinology. These domains fall within one of the valorisation domains of the Antwerp University, and the newly established business unit Antwerp Valorisation & Development (AVD) of the UAntwerp. The VAXINFECTIO-PD consortium built up a unique and extensive track record through research, services, spin-off creation and innovative pathways, in generating product concepts/prototypes and research platforms that form the basis of medical innovation. The various core research units have had an important international image in the recent years with publications in leading journals, coordination of several European projects, as well as active presence and involvement in international scientific and policy forums. For the 6-year period the IOF-consortium will further focus on 5 interlinked valorisation avenues, all creating or guaranteeing growth on the parameters P3, P4, P5 and P6: translational vaccination platform for improved and new preventive and therapeutic vaccines, prognostic and diagnostic platforms, core facilities (for cellular vaccines, human challenge studies and biobanks), infectious disease and immune modelling and prediction, and improved vaccine delivery and medical devices through product development.

Researcher(s)

Research team(s)

Project type(s)

  • Research Project

Approaching multiple sclerosis from a computational perspective through bioinformatic analysis of the T-cell repertoire. 01/11/2020 - 31/10/2024

Abstract

Recent developments in the field of sequencing technology allow for the characterization of adaptive immune receptor repertoires with unprecedented detail. T-cell receptor (TCR) sequencing holds tremendous promise for understanding the involvement and dynamics of adaptive immune components in autoimmune disorders. As the field is rapidly evolving from pre-processing of TCR-seq data to functional analysis of adaptive immune repertoires, new opportunities emerge for the development of comprehensive approaches for the post-analysis of immune receptor profiles. These approaches can offer comprehensive solutions to address clinical questions in the research on autoimmune disorders. An important example is multiple sclerosis (MS), a neuroinflammatory disease of the central nervous system, for which very little is known about the specific T-cell clones involved in its pathogenesis. By analysing the adaptive immune repertoire of MS patients, we postulate it is possible to uncover key drivers of the MS disease process. The identified T-cell clones will present themselves as highly specific biomarkers and therapeutic targets. This translational research project will lead to novel approaches for the identification of condition-associated T-cell clones, to new monitoring tools to evaluate the efficacy of MS-therapies and to a model to predict the disease course of MS.

Researcher(s)

Research team(s)

Project type(s)

  • Research Project

Engaging the immune system for remyelination in the brain using BDNF-engineered regulatory T cells. 01/10/2020 - 30/09/2024

Abstract

Multiple sclerosis (MS) is a neurodegenerative disease of the central nervous system (CNS), characterized by inflammatory attacks against the myelin sheath. Today, over 10 disease-modifying therapies are approved, predominantly focusing on immunomodulation. However, remyelination remains a major unmet clinical need in (progressive) MS therapy. Today, efforts are made to unravel de- and remyelinating mechanisms. Therefore, brain-derived neurotrophic factor (BDNF) seems an interesting protein, as it promotes neuroprotection and (re)myelination. Interestingly, BDNF levels are reported to be reduced in MS. While neurons are the principal source of BDNF in the CNS, key-immune cells can also secrete BDNF, suggesting that BDNF mediates the cross-talk between the immune- and nervous system. Recently, a growing body of research underscoring the key role of regulatory T cells (Treg) in MS, has emerged. Interestingly, a novel pro-regenerative function of Treg was revealed, mediated by the secretion of pro-myelinating factors. Nevertheless, the relation between immune cell-mediated BDNF expression and its accompanying effects in the CNS, such as remyelination, remains elusive in MS. Therefore, we aim to investigate the influence of immune cell-induced BDNF expression on remyelination using state-of-the-art techniques and patient samples. Our findings may result in the development of novel strategies to improve remyelination, predominantly focussing on progressive MS treatment.

Researcher(s)

Research team(s)

Project type(s)

  • Research Project

INnovative Training in Myeloid Regulatory Cell Therapy (INsTRuCT). 01/01/2020 - 30/06/2024

Abstract

Cell therapy is an active area of immunological research and represents a highly innovative and rapidly expanding sector of pharmaceutical industry. The INsTRuCT Consortium answers an unmet need in the field for postdoctoral researchers experienced in scientifically excellent research and cell therapy development. INsTRuCT draws upon complementary expertise of its academic and industrial partners to offer a unique research and training programme. INsTRuCT proposes 15 distinctive research projects based at European companies or universities recognized for their scientific achievements and innovation. INsTRuCT is structured to promote interdisciplinary and intersectoral cooperation between partners, thereby accelerating pharmaceutical development and clinical application of novel myeloid regulatory cell (MRC)-based therapies. INsTRuCT is a primarily research-based training programme, which will be complemented by theoretical and practical training opportunities. INsTRuCT will encourage a translational view of research, which will be reinforced by intersectoral secondments. Teaching transferrable and communication skills is a high priority for INsTRuCT. ESR will gain a comprehensive overview of the drug development process in Europe as it applies to cell-based therapies; hence, INsTRuCT's graduates will be fitted for future roles as innovative leaders in the field. INsTRuCT will strengthen interactions between cooperating research groups at junior and senior levels, thereby promoting dissemination of standardized research approaches and data-sharing. Overall, INsTRuCT constitutes an original research and training concept that responds to the specific needs of a growing sector for postdoctoral scientists trained in Basic Immunology and cell therapy development. Consequently, INsTRuCT has a very high impact potential, both in terms of its scientific and technical advancements, and its future contribution to innovation and economic development within the European Union.

Researcher(s)

Research team(s)

Project type(s)

  • Research Project

Integrative omics approach to identify the mechanism of tolerance induction by tolerogenic dendritic cell­ derived extracellular vesicles in multiple sclerosis. 01/01/2022 - 31/12/2023

Abstract

While first generation tolDC-based therapies have shown considerable clinical promise, a better understanding of tolDC immunobiology will open many possibilities for enhancing or redirecting their therapeutic activities. In this project, we aim to investigate mechanisms linking metabolic activity of tolDC to their functional polarization. We hypothesize that tolDC-derived EV have the potential to regulate tolerance-inducing molecular pathways. We aim to identify metabolites involved in the mode-of-action of tolDC immunoregulation. For this, the following objectives have been set forth: (1) To purify tolDC-derived EV from MS patients and healthy controls and to assess their immunoregulatory function using in vitro systems (2) To identify key metabolomic and lipidomic biomarkers in patients and healthy control tolDC-derived EV using omics analysis (3) To engineer and validate the key factors in tolerance induction and therapeutic repair in tolDC-derived EV (4) To investigate the therapeutic effectiveness of immunometabolite-containing EV in vivo In summary, this research project will contribute to a better understanding of the mode-of-action of vitD3-treated tolDC, focusing on EV and metabolite/bioactive lipid components. We envisage that our results will provide proof of the immunoregulatory capacities of EV and provide new insights in the use of EV or modified form for the treatment of MS.

Researcher(s)

Research team(s)

Project type(s)

  • Research Project

BDNF-engineered regulatory T cells, a future regenerative cell-therapeutic strategy in progressive MS. 01/01/2021 - 31/12/2022

Abstract

The general goal of this research project is to develop a clinically safe cell-based vaccine for the treatment of (progressive) MS, based on BDNF-expressing Tregs. By using state-of-the art techniques, we will develop "designer" Tregs that are engineered to express high levels of BDNF. We hypothesize that these Tregs will excel in their pro-regenerative properties, driving oligodendrocyte differentiation and remyelination, beyond immunomodulation with the aim to induce remyelination in MS.

Researcher(s)

Research team(s)

Project type(s)

  • Research Project

Targeted tolerance in multiple sclerosis: development of transgenic T cell receptor-engineered regulatory T cells recognizing myelin-derived antigens. 01/01/2020 - 31/12/2023

Abstract

Cell therapy is one of the most promising future clinical options in the medical arsenal for the treatment of patients suffering from serious conditions where unmet medical needs exist. Breakthroughs in cell and molecular biology have enabled the development of cell-based vaccines, and to date cell therapies are being evaluated in the first clinical trials aiming to treat autoimmune diseases, including multiple sclerosis (MS). Although the therapeutic landscape of MS is constantly evolving, none of the currently available treatments results in a permanent stabilization of the disease, and most of them indiscriminately suppress the immune system. In this perspective, immune-modulatory cell therapy has the potential to target underlying disease mechanisms in a more specific way. In particular, regulatory T cells (Tregs) offer the opportunity to target cells that are potentially involved in the induction and progression of the disease. In current proposal, we aim to develop TCR-engineered Tregs to enforce their interaction with cells that are key in the disease pathogenesis. In doing so, we ultimately aim to control autoimmunity.

Researcher(s)

Research team(s)

Project type(s)

  • Research Project

Deep Sequencing of myelin-reactive T-cells to elucidate new disease mechanisms and identify correlates for treatment responsiveness. 01/01/2020 - 31/12/2020

Abstract

Whereas antigen-specific activation of autoreactive T-cells is considered essential in the initiation and maintenance of MS, how to identify the broad repertoire of unique receptors expressed by these autoreactive T-cells from blood remains unclear. Nonetheless, with improved T-cell receptor (TCR)-sequencing technological development, efforts in identifying immune T-cell signatures in blood, CSF and brain lesions of MS patients have been initiated. Although accurately evaluating TCR clonal expansion using high throughput sequencing in bulk DNA/RNA has been challenging, single-cell sequencing allows to establish TCR repertoires of autoreactive T-cells on a cell-by-cell basis, obtain full-length V(D)J sequences, pair α and β sequences and combine TCR with 5' transcriptome sequencing in the same cells, and this for 1000s of cells. The combined expertise of our interuniversity team in immunology and characterization of autoreactive T-cells (N. Cools, U Antwerp) on the one hand and in genetics and single-cell sequencing (A. Goris, KU Leuven) on the other makes it now feasible, timely and innovative to investigate the pathogenic characteristics of autoreactive T-cells in MS. For this, the following three aims have been set forth: 1. What is the TCR repertoire of autoreactive T-cells in MS? 2. What are the transcriptional characteristics of autoreactive T-cells? 3. Can the autoreactive T-cell clonotype repertoire be used as a correlate for therapy responsiveness?

Researcher(s)

Research team(s)

Project type(s)

  • Research Project

Targeted tolerance in multiple sclerosis: development of transgenic T cell receptor-engineered regulatory T cells recognizing myelinderived antigens. 01/01/2019 - 31/12/2022

Abstract

The therapeutic landscape of MS is constantly evolving, and one could pose the question if we still have unmet needs for the treatment of MS? Nevertheless, despite the availability of improved therapies and the significant advances in the understanding of what triggers disease, patients continue to experience relapses and, in some cases, are exposed to potential life-threatening side-effects. Hence, current challenge is to balance the need to modify the underlying disease pathogenesis and the long-term risks. In this perspective, immunemodulatory cell therapy has brought a new hope for a wide spectrum of diseases. Tregs offer the opportunity to target cells that are potentially involved in the disease progress. Nevertheless, whether Tregs act in an antigen-specific manner remains elusive. Hence, despite the potential that Treg therapy holds, 2 there are still some challenges, not in the least to direct the interaction of Tregs with key disease-associated immune cells in an antigen-specific manner. To address these, the following objectives have been set forth in current project proposal: Our first objective is to select antigen-specific effector T cells by means of tetramer analysis, thereby identifying and cloning a myelin-recognizing TCR. Secondly, we will optimize a clinically safe mRNA electroporation protocol to induce expression of mRNA encoding the TCR in freshly-isolated and expanded Tregs from MS patients. Thirdly, we ensure the stability of the phenotype and suppressive function of TCR-engineered Tregs. In doing so, we will deliver in vitro proof-of-concept of the safety of the approach which is especially important when administering the cells in an inflammatory disease-driven microenvironment. Finally, we will investigate if TCR-transgenic Tregs can modulate ongoing disease processes by investigating their effect on the phenotype and function of DCs from healthy volunteers and MS patients. Ultimately, we envisage that this will foster a durable clinical application of this technology without the risk for general immunosuppression.

Researcher(s)

Research team(s)

Project type(s)

  • Research Project

Towards patient-tailored treatment in multiple sclerosis: a dendritic cell-based vaccine for the treatment of multiple sclerosis. 01/10/2018 - 30/09/2020

Abstract

Multiple sclerosis (MS) is an inflammatory disease of the central nervous system in which the body 's own immune system attacks the myelin sheath. This leads to disruption in signaling in the brain and spinal cord and to loss of brain tissue. MS is the most common cause of non-traumatic disability in young adults. To date, many aspecific immunomodulatory and general immunosuppressive treatments are used to slow down the disease course, but these treatments have several side effects, ranging from mild to severe and life-threatening issues, including other autoimmune diseases and infections. Thus, there remains an unmet need for specific treatments with a good safety profile. Restoring antigen specific tolerance is an interesting approach to tackle these problems. Theoretically, a limited number of vaccinations with tolerogenic dendritic cells (tolDC) could reeducate the patient's own immune system in the longterm. Based on our previous research in the laboratory on MS and clinical studies in other autoimmune diseases we are ready to bring tolDC treatment to MS patients. The aim of this project is to assess safety and feasibility of autologous myelin-peptide-loaded tolDC in active MS patients, who will receive 6 vaccinations in a phase I clinical trial. Safety will be evaluated by recording of adverse events. Feasibility will be determined by successful production of tolDC. Positive results can lead to clinical trials evaluating efficacy.

Researcher(s)

Research team(s)

Project type(s)

  • Research Project

Support EU-project LEGO. 08/05/2018 - 07/11/2018

Abstract

This project allowed us to consult a scientific writer for the support in the preparation of a European Horizon 2020 project, entitled LEGO (Leveraging qualitative Engineering of GMP-Operations for advanced therapeutics).

Researcher(s)

Research team(s)

    Project type(s)

    • Research Project

    Neuronal self-renewal by antigen-specific tolerization in multiple sclerosis reinstalling the balance between inflammation and regeneration (RESTORE). 01/01/2018 - 30/06/2023

    Abstract

    Multiple sclerosis (MS) is an inflammatory neurodegenerative disease of the central nervous system (CNS) for which no cure is currently available. It is the leading cause of non-traumatic disabling neurological disease in young adults with more than 500,000 people affected in Europe. Since MS strikes during the primary productive time of one's personal and professional life, it leads to a major physical and socio-economic burden to the patient, family and society. Therefore, new therapeutic interventions with improved efficacy over existing drugs and good tolerability are warranted. As chronic inflammatory processes drive the neurodegeneration, we hypothesize that improved clinical outcome depends on restoring the balance between inflammation and the remaining capacity of neuronal self-renewal. In this perspective, cell therapy that specifically targets the damaging immune reactions that cause MS and induce disease-specific tolerance without affecting protective immunity against pathogens and cancer is a promising approach. Recently, we set-up a collaborative network of European centers working in cell therapy (COST Action BM1305). From this, centers from four different EU countries with two additional partners now aim to take the next step and join efforts to bring antigen-specific therapy for MS to the clinic. Our objectives are to evaluate safety, clinical practicality and demonstrate first proof-of-principle of therapeutic efficacy of antigen-specific tolerance-inducing dendritic cells (tolDC) in MS patients in two single-center clinical trials while comparing different modes of tolDC administration. Coordinated patient monitoring and centralized MRI monitoring, including radiological correlates of neurodegeneration, and immunomonitoring will enable us to directly compare results between trials and enable consented biobanking, data safeguarding and accessibility to support future efforts in the field of MS therapy. Antigen-specific cell therapy has the potential to provide this chronic inflammatory disease with a personalized and effective treatment option and therefore fits within current program. An effective therapy that lowers morbidity by uniting efficacy with reduced occurrence of side effects and less frequent hospitalizations will enhance quality of life of patients as well as dramatically reduce economic burden. This would represent a breakthrough for healthcare in MS.

    Researcher(s)

    Research team(s)

    Project type(s)

    • Research Project

    Targeted tolerance in multiple sclerosis: development of transgenic T cell receptor-engineered regulatory T cells recognizing myelin basic proteins. 01/01/2018 - 31/12/2018

    Abstract

    The therapeutic landscape of MS is constantly evolving, and one could pose the question if we still have unmet needs for the treatment of MS? Nevertheless, despite the availability of improved therapies and the significant advances in the understanding of what triggers disease, patients continue to experience relapses and, in some cases, are exposed to potential life-threatening side-effects. Hence, current challenge is to balance the need to modify the underlying disease pathogenesis and the long-term risks. In this perspective, immunemodulatory cell therapy has brought a new hope for a wide spectrum of diseases. Tregs offer the opportunity to target cells that are potentially involved in the disease progress. Nevertheless, whether Tregs act in an antigen-specific manner remains elusive. Hence, despite the potential that Treg therapy holds, 2 there are still some challenges, not in the least to direct the interaction of Tregs with key disease-associated immune cells in an antigen-specific manner. To address these, the following objectives have been set forth in current project proposal: Our first objective is to select antigen-specific effector T cells by means of tetramer analysis, thereby identifying and cloning a myelin-recognizing TCR. Secondly, we will optimize a clinically safe mRNA electroporation protocol to induce expression of mRNA encoding the TCR in freshly-isolated and expanded Tregs from MS patients. Thirdly, we ensure the stability of the phenotype and suppressive function of TCR-engineered Tregs. In doing so, we will deliver in vitro proof-of-concept of the safety of the approach which is especially important when administering the cells in an inflammatory disease-driven microenvironment. Finally, we will investigate if TCR-transgenic Tregs can modulate ongoing disease processes by investigating their effect on the phenotype and function of DCs from healthy volunteers and MS patients. Ultimately, we envisage that this will foster a durable clinical application of this technology without the risk for general immunosuppression.

    Researcher(s)

    Research team(s)

      Project type(s)

      • Research Project

      Isolation, freezing and storage of peripheral blood mononuclear cells (PBMC). 28/08/2017 - 30/06/2019

      Abstract

      This project represents a formal research agreement between UA and on the other hand the client. UA provides the client research results mentioned in the title of the project under the conditions as stipulated in this contract.

      Researcher(s)

      Research team(s)

      Project type(s)

      • Research Project

      From bench to bedside: accelerating the clinical development of cell therapy innovations by "lean" gmp manufacturing of ATMP. 01/05/2016 - 30/11/2016

      Abstract

      To specifically advance translation initiatives of cell therapeutic advanced therapeutic medicinal products (ATMP) into commercially viable and safe treatments and hence, overcome regulatory, developer-related and value chain-related challenges, a consortium of stakeholders along the value chain, will, in collaboration with the sponsor, drive the ATMP through the early clinical trials with the objective to generate a lean manufactured and effective ATMP. The key concept is to task each of the specific necessary steps to the entity that is best suited for the task. It is important for all team members to understand, to some degree, the whole process involved in product development to adequately balance what needs to be done and more importantly when to do it as time and resources are not unlimited. Concrete tasks of this partnership are (i) upgrade the research 'product and process' to a lean manufacturing process with built-in quality measures, (ii) revise the whole process to generate an efficient, scalable process from patient to lab and back to patient, (iii) interact from an early stage with the Committee for Advanced Therapies (CAT) in the process of marketing authorization licensing and (iv) will support the sponsor in the licensing/spin-out of their ATMP project. In collaboration with an external consultant, different business models will be evaluated from which one concrete business case will be designed and serve as the basis to apply for further funding.

      Researcher(s)

      Research team(s)

        Project type(s)

        • Research Project

        Towards a "negative" cellular vaccine for the treatment of multiple sclerosis: counter-acting epitope spreading for long-lasting tolerance induction using mRNA-electroporated tolerogenic dendritic cells. 01/01/2016 - 30/09/2018

        Abstract

        Multiple sclerosis (MS) is an inflammatory disease that affects 1 out of 1000 people in Europe. MS is mediated by an autoimmune response to components or antigens of the insulating cover of the nerves of the central nervous system, i.e. myelin antigens, which are no longer recognised as being 'self'. This causes progressive destruction of the nerves of the brain and spinal cord, presenting as a wide range of motor and sensory disturbances. On the basis of our recent research, we hypothesise that specific cells of the immune system, i.e. dendritic cells (DC), regulate the immune response by inducing immunity or tolerance towards specific antigens. Therefore these DC might be used to suppress abnormal immune responses in autoimmune diseases such as MS. The final aim will be to induce tolerance to myelin antigens by administering DC to MS patients, which might lead to a change in the natural course of the disease. Several techniques exist for the loading of DC with specific antigens, which makes them tolerance inducing for specific autoimmune responses rather than being overall immunosuppressive. We will compare two of these techniques, in combination with a dose escalation vaccine study in MS patients in order to ensure patient safety when administrating a vaccine with tolerance-inducing DC. Patients will be divided in two groups, comparing both antigen-loading strategies by evaluating adverse events, relapse-free interval and immunological response after vaccination.

        Researcher(s)

        Research team(s)

          Project type(s)

          • Research Project

          Identification and design of dendritic cells with blood-brain barrier-crossing capacity: moving targets to treat multiple sclerosis (MS) 01/10/2015 - 30/09/2019

          Abstract

          Multiple sclerosis (MS) is the leading cause of non-traumatic disability in young adults. Although growing insights into disease mechanisms underlying MS have resulted in the development of new therapeutic strategies, none of the currently available treatments results in permanent stabilization or cure of MS. Current research efforts are focused on further unraveling MS immunopathogenesis as well as on finding ways to specifically manipulate disease-causing immune cells in order to treat MS. In this context, dendritic cells (DC) are set forth as interesting cellular targets. Post-mortem studies of MS brains as well as studies in animal models suggest that migration of DC from the bloodstream through the blood-brain barrier (BBB) and subsequent accumulation of these cells in the brain parenchyma represent crucial events in MS pathogenesis. Hence, DC and the process of DC migration are interesting targets for the development of new therapeutic strategies. Here, we will study the transmigratory capacity of circulating DC from MS patients using an in vitro BBB model. By studying differences in phenotype and function between migrating and non-migrating DC from MS patients and healthy controls, we aim to identify new therapeutic targets in order to interfere with DC recruitment to the brain. Ultimately, this will allow us to generate tolerogenic DC exhibiting enhanced migratory capacity, with the potential to suppress ongoing myelin-specific responses in the central nervous system.

          Researcher(s)

          Research team(s)

          Project type(s)

          • Research Project

          A "negative" dendritic-cell based vaccine for the treatment of multiple sclerosis: a first-in-man multicenter trial. 15/01/2015 - 14/01/2019

          Abstract

          This project represents a research agreement between the UA and on the onther hand IWT. UA provides IWT research results mentioned in the title of the project under the conditions as stipulated in this contract.

          Researcher(s)

          Research team(s)

          Project type(s)

          • Research Project

          Dendritic cell recruitment in the central nervous system: moving targets to treat multiple sclerosis. 01/01/2014 - 31/12/2014

          Abstract

          This project represents a formal research agreement between UA and on the other Charcot Stichting. UA provides Charcot Stichting research results mentioned in the title of the project under the conditions as stipulated in this contract.

          Researcher(s)

          Research team(s)

            Project type(s)

            • Research Project

            Tolerogenic dendritic cells: a silencing strategy to suppress autoimmunity. 01/01/2013 - 31/12/2013

            Abstract

            This project represents a formal research agreement between UA and on the other Charcot Stichting. UA provides Charcot Stichting research results mentioned in the title of the project under the conditions as stipulated in this contract.

            Researcher(s)

            Research team(s)

              Project type(s)

              • Research Project

              Dendritic cells as a potential target for the development of an atherosclerosis vaccine. 01/10/2012 - 30/09/2016

              Abstract

              Acute cardiovascular syndromes (e.g. myocardial infarction and stroke) are a major cause of morbidity and mortality in industrialized countries. They result from rupture and subsequent thrombosis of an atherosclerotic plaque that has built up in the wall of large and middle-sized arteries. Chronic inflammation, mediated by dendritic cells (DCs) drives the development of atherosclerosis. DCs are present in healthy arteries in areas predisposed to atherosclerotic plaque formation, and accumulate within plaques where they can be localized in close vicinity to T cells. Recent work has revealed important functions of DCs in regulating inflammatory and immune mechanisms in atherogenesis. In addition to antigen presentation to T-cells with subsequent activation, DCs themselves will secrete inflammatory cytokines (e.g. IL-12), further exacerbating atherosclerosis. Because of their unique properties (capable of inducing either immune responses or immune tolerance), DCs can be harnessed to suppress unwanted responses, in the form of vaccines. Vaccination strategies using DCs are currently being explored in various diseases. In fact, the approval of Provenge® (the first "DC vaccine" for prostate cancer) by the FDA in 2010 has paved the way for further development of DC vaccines. Our central hypothesis is that a vaccine-based approach to manage atherosclerotic cardiovascular disease is a potentially viable strategy. In fact, the first proof of concept that this approach could be very useful in combatting cardiovascular disease came very recently from the group of Goran Hansson. Treatment of mice with in vitro generated tolerogenic DCs attenuated atherosclerotic plaque development. However, these DCs have an unstable phenotype. Therefore, in this project we aim to (1) identify biomarkers for plaque DCs for targeted immunotherapy, (2) generate stable, tolerogenic DCs using RNA interference to suppress autologous T-cell activation and (3) study the effects of siRNA mediated DC targeted IL-12 silencing on atherosclerotic plaque progression and stability in mice. This research program is part of a global research effort to develop new therapeutic approaches for atherosclerotic plaque stabilization. Specifically, the current program aims to elucidate whether or not modulation of immune responses can stabilize atherosclerotic plaques.

              Researcher(s)

              Research team(s)

              Project type(s)

              • Research Project

              The effect of cellular mediators on the modulation of innate pathogenic responses in multiple sclerosis (MS). 01/10/2012 - 30/09/2015

              Abstract

              In this project, we want to further investigate and exploit the capacity of DC and Treg to correct or modulate pathogenic responses in MS patients. Current research will provide the foundation for the eventual development of a cellular vaccine for the treatment of MS. Depending on the results of this study it can be envisaged to treat patients suffering from MS with tolerogenic DC and/or immunosuppressive Treg in order to eliminate or inactivate autoreactive T cells.

              Researcher(s)

              Research team(s)

                Project type(s)

                • Research Project

                The extent of bone marrow dysfunction and the potential to reverse underlying mechanisms in patients with ischemic cardiomyopathy. 01/01/2012 - 31/12/2015

                Abstract

                The proposed project approach will create unique opportunities to describe the extent of bone marrow dysfunction in patients with ischemic heart disease and to explore underlying mechanisms. As such, it might be possible to identify reversible pathways that should be targeted in order to functionally rejuvenate "sick" autologous bone marrow-derived stem cells. The combination of gene and cell therapy has the potential to create tailored therapy, ultimately improving morbidity and mortality of patients with cardiovascular disease.

                Researcher(s)

                Research team(s)

                  Project type(s)

                  • Research Project

                  The immune-modulating role of vitamin D3-treated dendritric cells in multiple sclerosis. 01/01/2011 - 31/12/2013

                  Abstract

                  This project represents a formal research agreement between UA and on the other hand a private institution. UA provides the private institution research results mentioned in the title of the project under the conditions as stipulated in this contract.

                  Researcher(s)

                  Research team(s)

                    Project type(s)

                    • Research Project

                    Characterization of circulating dendritic cells (DC) in multiple sclerosis (MS): evaluation of subtypes and functionality. 01/01/2010 - 31/12/2011

                    Abstract

                    To date, knowledge with regard to the distribution of myeloid and plasmacytoid dendritic cells in the circulation of MS patients is lacking. To support the hypothesis that a disturbance in the distribution of DC subtypes and/or DC function contributes to an imbalance in the regulation of immunity and tolerance, the number, phenotype and function of DC in the peripheral blood of MS patient and healthy controls will be determined.

                    Researcher(s)

                    Research team(s)

                      Project type(s)

                      • Research Project

                      The effect of cellular mediators on the modulation of pathogenic responses in multiple sclerosis (ms) patients. 01/10/2009 - 30/09/2012

                      Abstract

                      In this project, we want to further investigate and exploit the capacity of DC and Treg to correct or modulate pathogenic responses in MS patients. Current research will provide the foundation for the eventual development of a cellular vaccine for the treatment of MS. Depending on the results of this study it can be envisaged to treat patients suffering from MS with tolerogenic DC and/or immunosuppressive Treg in order to eliminate or inactivate autoreactive T cells.

                      Researcher(s)

                      Research team(s)

                        Project type(s)

                        • Research Project

                        Clinical and preclinical research of the effect of cellular mediators on the modulation of pathogenic responses in multiple sclerosis. 01/01/2009 - 31/12/2012

                        Abstract

                        In this project, we want to further investigate and exploit the capacity of DC and Treg to correct or modulate pathogenic responses in MS patients. Current research will provide the foundation for the eventual development of a cellular vaccine for the treatment of MS. Depending on the results of this study it can be envisaged to treat patients suffering from MS with tolerogenic DC and/or immunosuppressive Treg in order to eliminate or inactivate autoreactive T cells.

                        Researcher(s)

                        Research team(s)

                          Project type(s)

                          • Research Project

                          Functional chracterisation of regulatory cell populations: In vitro induction of tolerance using dendritic cells. 01/01/2008 - 31/12/2009

                          Abstract

                          In the current project, our aim is to further investigate and exploit the capacity of dendritic cells and regulatory T cells to suppress antigen-specific immune responses. A better understanding of their interactions, as well as the mechanisms involved in the induction of peripheral T cell tolerance will ultimately lead to the development of new therapies for immune-mediated pathologies.

                          Researcher(s)

                          Research team(s)

                            Project type(s)

                            • Research Project

                            Development of a dendritic cell based vaccine against human papilloma virus induced cervical cancer. 01/10/2003 - 30/09/2005

                            Abstract

                            Cervical cancer is the third most common cancer among women worldwide. Despite the progress in current therapies, these common treatments still have several disadvantages. On the one side is it impossible to remove all occurred micrometastases with surgery. On the other hand do radio- and chemotherapy not exclusively attack tumour tissue, but also exert a toxic effect on normal tissues and suppress the immune system. Therefore current research aims novel and more effective therapies. Loading dendritic cells with tumour antigens for the induction of anti-tumour immunity is at this moment a 'hot topic' of experimental immunotherapy. The principle of immunotherapy is to specifically stimulate the immune system for generating a tumour specific immune response. Cervical cancer is linked with human papilloma virus (HPV) infection. Although there are over 20 oncogenic HPV genotypes, HPV type 16 and 18 are the most prevalent in cervix carcinoma. The principle of a therapeutic vaccine consists in presenting HPV related antigens, such as E6 and E7 viral proteins, to professional antigen presenting cells (APC), especially dendritic cells (DC), to induce a strong T-cell response. The shape, in which the HPV antigens are presented to the DC, determines for a great part the type and the strength of the resulting T-cell response. The current challenge is determining the most efficient way in using modified DC in immunotherapeutic protocols . The generation of an optimal cellular anti-tumour response implies the activation of tumour specific CD8+ cytotoxic T-cells. However, more and more studies underline the important role of CD4+ T-cells in the regulation of the immune response. We aim in this project at the generation of a strong HPV specific CD4-positive as well as a CD8-positive immune response. For this purpose, we will generate cDNA constructs for the in vitro transcription of HPV 16 E6 and E7 mRNA, if necessary in combination with MHC class II signalling sequences (e.g. LAMP-I). The functionality of the produced mRNA shall be tested after transfection of DC with this mRNA. We will demonstrate protein expression with Western Blotting, while MHC class I and/or class II presentation of the antigenic peptides shall be tested with peptide-specific CD8+ or CD4+ T-cell clones. We shall also start with the development of an in vitro T-cell activation protocol for the generation of HPV 16 E6 and E7 specific T-cells with peptide pulsed DC. Furthermore, we shall start up an ex vivo study, more specific in the second term of the project, on the basis of the developed strategy during the first term. We will determine whether in vitro an autologous immune response can be elucidated against the tumour cells of a patient. For this purpose, we shall generate DC from peripheral blood samples of cervix carcinoma patients and modify these DC with mRNA coding for HPV antigens. After in vitro activation of autologous T-cells, we shall determine if there is sufficient reactivity against autologous tumour cells of the patients. The ultimate goal of these experiments is the development of an efficient strategy that can be used in a clinical model.

                            Researcher(s)

                            Research team(s)

                              Project type(s)

                              • Research Project

                              Development of a dendritic cell based vaccine against human papilloma virus induced cervical cancer. 01/10/2001 - 30/09/2003

                              Abstract

                              Cervical cancer is the third most common cancer among women worldwide. Despite the progress in current therapies, these common treatments still have several disadvantages. On the one side is it impossible to remove all occurred micrometastases with surgery. On the other hand do radio- and chemotherapy not exclusively attack tumour tissue, but also exert a toxic effect on normal tissues and suppress the immune system. Therefore current research aims novel and more effective therapies. Loading dendritic cells with tumour antigens for the induction of anti-tumour immunity is at this moment a 'hot topic' of experimental immunotherapy. The principle of immunotherapy is to specifically stimulate the immune system for generating a tumour specific immune response. Cervical cancer is linked with human papilloma virus (HPV) infection. Although there are over 20 oncogenic HPV genotypes, HPV type 16 and 18 are the most prevalent in cervix carcinoma. The principle of a therapeutic vaccine consists in presenting HPV related antigens, such as E6 and E7 viral proteins, to professional antigen presenting cells (APC), especially dendritic cells (DC), to induce a strong T-cell response. The shape, in which the HPV antigens are presented to the DC, determines for a great part the type and the strength of the resulting T-cell response. The current challenge is determining the most efficient way in using modified DC in immunotherapeutic protocols . The generation of an optimal cellular anti-tumour response implies the activation of tumour specific CD8+ cytotoxic T-cells. However, more and more studies underline the important role of CD4+ T-cells in the regulation of the immune response. We aim in this project at the generation of a strong HPV specific CD4-positive as well as a CD8-positive immune response. For this purpose, we will generate cDNA constructs for the in vitro transcription of HPV 16 E6 and E7 mRNA, if necessary in combination with MHC class II signalling sequences (e.g. LAMP-I). The functionality of the produced mRNA shall be tested after transfection of DC with this mRNA. We will demonstrate protein expression with Western Blotting, while MHC class I and/or class II presentation of the antigenic peptides shall be tested with peptide-specific CD8+ or CD4+ T-cell clones. We shall also start with the development of an in vitro T-cell activation protocol for the generation of HPV 16 E6 and E7 specific T-cells with peptide pulsed DC. Furthermore, we shall start up an ex vivo study, more specific in the second term of the project, on the basis of the developed strategy during the first term. We will determine whether in vitro an autologous immune response can be elucidated against the tumour cells of a patient. For this purpose, we shall generate DC from peripheral blood samples of cervix carcinoma patients and modify these DC with mRNA coding for HPV antigens. After in vitro activation of autologous T-cells, we shall determine if there is sufficient reactivity against autologous tumour cells of the patients. The ultimate goal of these experiments is the development of an efficient strategy that can be used in a clinical model.

                              Researcher(s)

                              Research team(s)

                                Project type(s)

                                • Research Project