Ongoing projects

The study of alpha-synuclein pathology and related neuroinflammation in a human brain-like context: a human neurospheroid approach. 01/04/2024 - 31/03/2025

Abstract

Synucleinopathies, encompassing diseases like Parkinson's disease and dementia with Lewy bodies, are a group of neurodegenerative disorders characterized by the formation of alpha-synuclein (αSyn) aggregates that are able to propagate in a prion-like manner between cells of the nervous system. However, the exact role of αSyn pathology in the disease progression of these synucleinopathies remains to be elucidated. Moreover, microglia have been pointed to as a major player in synucleinopathy pathophysiology, but how these cells affect αSyn pathology remains unclear. Current in vitro models are limited in their ability to replicate human responses to pathological αSyn with sufficient fidelity. Recently developed (microglia-containing) brain organoids represent a promising new tool to study αSyn pathology in a human-brain like environment. In this study, we will use human brain organoids or 'neurospheroids' (NSPHs) to enhance our understanding of αSyn pathology, focusing on propagation and associated pathophysiological pathways. By using NSPHs with and without microglia (annotated as tri- and bipartite NSPHs), we aim to determine the role of microglia and neuroinflammation in these processes. To this end, pre-formed αSyn fibrils will be added to NSPHs. Staining of NSPHs for pathological αSyn, by phosphorylated αSyn antibody and thioflavin, at different timepoints allows to monitor internalization, accumulation and propagation of αSyn pathology. Altered pathophysiological pathways will be determined by RNA-sequencing and validated at the protein level by means of immunocytochemistry (ICC). In summary, this project will help to identify major alterations associated with αSyn pathology and clarify the role of microglia in a human brain-like context.

Researcher(s)

Research team(s)

Project type(s)

  • Research Project

Characterize specific human TCRs via TCR Seq including both NGS protocols and in silico analysis and in vitro experimental assays for T cells stimulations with tumor associated and viral antigens pools, with the final aim of TCR-T cells development. 01/03/2024 - 28/02/2026

Abstract

Next Generation Sequencing (NGS) has emerged as a suitable tool to evaluate and characterize the T Cell Receptor (TCR) immune repertoire. This approach paves the way for the use of the TCR repertoire study as a novel complex biomarker to track the exposure of individuals to non-self antigens and to develop new therapeutic strategies by improving our comprehension of the adaptive immune response against infectious agents and cancer antigens. The collaboration between Italy's Istituto Romagnolo per lo Studio dei Tumori "Dino Amadori" (IRST) and Belgium's University of Antwerp involves both these fields and aims to identify and characterize specific human TCRs via NGS to develop therapeutic TCR-T cells, exploiting experimental protocols, in silico analysis, and in vitro assays for T cell stimulation with antigens' pools. The first objective is to study the T cell response to mRNA-based COVID vaccines in healthy subjects and lymphoma patients from Emilia Romagna region through RNA samples sequencing and subsequent analysis of TCR specificity against SARS-CoV-2 proteins. The correlation between TCR diversity and strength with the amount of neutralizing antibodies will also be examined in consideration of the predicted HLA context, leading to an actionable SARS-Cov2 bulk TCR-seq database. Genomic instability, which is a common feature in cancer cells, often leads to the generation of chromosomal rearrangements and aneuploidy. Many are the examples today of gene fusions that promote cell transformation in different oncological settings. Those events lead to the unique opportunity to generate neoantigens that could be presented to the immune system in an HLA-restricted manner. Therefore, the second objective is the identification and validation of neoantigens originated from genetic fusion events in cancer patients already available in IRST. Subsequently, experiments will be focused on the identification of antigen-specific T cells and TCRs against neoantigens originating from selected genetic fusion events. To these ends, computational protein reconstruction and prioritization from the already available fusion genes database will be performed to retrieve fusion transcripts in hematological tumors. These peptides will be later synthesized and pooled to stimulate T cells, with subsequent expansion and characterization of the expressed receptors. Before that, a proof-of-concept T cell expansion and following TCR-identification experiments with model protein(s) for a known neoantigen will be set up.

Researcher(s)

Research team(s)

Project type(s)

  • Research Project

FWO Research Sabbatical 2023-2024 (Prof. P. Ponsaerts) 01/02/2024 - 31/01/2025

Abstract

The research team of prof. Peter Ponsaerts at the Laboratory of Experimental Hematology (University of Antwerp, UA) has established during the past 5 years multiple 2D and 3D murine and human iPSC-derived neuro-immune cell culture models (including neurons, astrocytes, macrophages and microglia), with a specific focus on inflammation, viral infection, stroke and Parkinson's Disease. Having established these models, the research team of prof. Peter Ponsaerts has already implemented standard cytokine profiling, immunocytochemistry (ICC) and bulk RNA-Seq analyses for cell type characterization and cellular responses to inflammation, trauma or infection. Nevertheless, there is an unmet need to advance their experimental toolbox to characterize 2D and 3D neuro-immune cell culture models. The envisaged research sabbatical at the "Mass Spectrometry Lab" of the "Analytical Biochemistry and Proteomics Research Unit" from the "Center for Advanced Studies and Technology (CAST)" at the "University G. d'Annunzio" in Chieti-Pescara in Italy aims at two educational (E) and two scientific (S) action points: (E1) To learn and understand the base principles of high sensitivity proteomics and metabolomics (including metallomics and lipidomics) analyses using state-of-the-art mass spectrometry instruments and methods. (E2) To learn and apply MaxQuant, Perseus, Ingenuity Pathways Analysis (IPA) and MetaboAnalyst software for omics data analysis. (S1) To perform a longitudinal proteomics and metabolomics study of human (h)iPSC-derived bi- and tri-partite neurospheroids during development and maturation. (S2) To perform a proteomics and metabolomics study of mature hiPSC-derived bi- and tri-partite neurospheroids following viral infection, hypoxic/hypoglycemic stress (stroke) and Parkinson's Disease (PD) mimicking conditions. By completion of this research sabbatical, prof. Peter Ponsaerts will not only have gained the necessary bioinformatics skills to analyse proteomics data, but also will be able to integrate novel proteomics and metabolomics data with scRNA-Seq, ICC and electrophysiological analyses performed by his research team at UA, within current and future research projects.

Researcher(s)

Research team(s)

Project type(s)

  • Research Project

BOF Sabbatical 2024 - P. Ponsaerts. 01/02/2024 - 31/01/2025

Abstract

The research team of prof. Peter Ponsaerts at the Laboratory of Experimental Hematology (University of Antwerp, UA) has established during the past 5 years multiple 2D and 3D murine and human iPSCderived neuro-immune cell culture models (including neurons, astrocytes, macrophages and microglia), with a specific focus on inflammation, viral infection, stroke and Parkinson's Disease. Having established these models, the research team of prof. Peter Ponsaerts has already implemented standard cytokine profiling, immunocytochemistry (ICC) and bulk RNA-Seq analyses for cell type characterization and cellular responses to inflammation, trauma or infection. Nevertheless, there is an unmet need to advance their experimental toolbox to characterize 2D and 3D neuro-immune cell culture models. The envisaged research sabbatical at the "Mass Spectrometry Lab" of the "Analytical Biochemistry and Proteomics Research Unit" from the "Center for Advanced Studies and Technology (CAST)" at the "University G. d'Annunzio" in Chieti-Pescara in Italy aims at two educational (E) and two scientific (S) action points: (E1) To learn and understand the base principles of high sensitivity proteomics and metabolomics (including metallomics and lipidomics) analyses using state-of-the-art mass spectrometry instruments and methods. (E2) To learn and apply MaxQuant, Perseus, Ingenuity Pathways Analysis (IPA) and MetaboAnalyst software for omics data analysis. (S1) To perform a longitudinal proteomics and metabolomics study of human (h)iPSC-derived bi- and tri-partite neurospheroids during development and maturation. (S2) To perform a proteomics and metabolomics study of mature hiPSC-derived bi- and tri-partite neurospheroids following viral infection, hypoxic/hypoglycemic stress (stroke) and Parkinson's Disease (PD) mimicking conditions. By completion of this research sabbatical, prof. Peter Ponsaerts will not only have gained the necessary bioinformatics skills to analyse proteomics data, but also will be able to integrate novel proteomics and metabolomics data with scRNA-Seq, ICC and electrophysiological analyses performed by his research team at UA, within current and future research projects.

Researcher(s)

Research team(s)

Project type(s)

  • Research Project

Functional and molecular profiling of immune-competent neurospheroids under stroke-like conditions. 01/11/2023 - 31/10/2024

Abstract

Due to the enormous impact of stroke on the patient's quality of life and on society, decades of research resulted in the identification of thousands of candidate neuroprotective drugs. Unfortunately, none have led to an effective therapy to date. This can partially be attributed to the lack of in vitro systems able to accurately recapitulate human ischemic responses. Fortunately, the advent of induced pluripotent stem cell (iPSC)-technology has provided novel tools for generating human-based in vitro brain models, namely neurospheroids. Extending the host laboratories' preceding research efforts to generate bi-partite (neurons + astrocytes) and tri-partite (neurons + astrocytes + microglia) human iPSC-derived neurospheroids, I hypothesize that subjection of mature tri-partite neurospheroids to stroke-like conditions (oxygen/glucose-deprivation), in combination with advanced single cell analysis tools and measurement of electrophysiological network activity, will aid to unravel biologically relevant cellular and molecular events in the context of stroke pathology. In this way, the combined cellular and molecular toolbox for neurospheroid culture, manipulation and analysis will in short term pave the way for novel fundamental studies unravelling new pathways and/or potential targets for neuroprotection or repair, and in long-term novel therapeutic approaches for patients with cerebral ischemia.

Researcher(s)

Research team(s)

Project type(s)

  • Research Project

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

Development of a novel chimeric antigen receptor (CAR)-T cell therapy for acute myeloid leukemia. 01/10/2023 - 30/09/2027

Abstract

Chimeric antigen receptor (CAR)-T cell therapy has revolutionized the treatment of certain hematologic cancers. In this immunotherapy, the patient's T-cells are "armoured" ex vivo with a CAR that targets certain antigens on the tumor cell surface. Once administered, the CAR-T-cells will recognize the tumor cells and mediate lysis of the cells However, CAR-T-cell therapy is not yet a breakthrough for acute myeloid leukemia (AML), a highly aggressive blood cancer with dismal prognosis, due to various reasons. One of the reasons is the lack of a suitable CAR-target antigen on the AML cell surface. Another contributing factor is that the T-cells in AML, which are usually taken from peripheral blood, are deemed suboptimal. It is possible that tumor-infiltrating lymphocytes (or TIL) represent a superior cell population, but little research in the CAR therapy field is focused on TIL. To our knowledge, no research has been conducted on the use of TIL for the development of CAR-T cell therapy in AML. The aim of this project is twofold, with the ultimate goal of developing a new CAR-T-cell therapy for AML. Firstly, in this project, a new CAR targeting a promising target antigen that is highly expressed on the AML cell surface will be tested. Secondly, we wish to determine whether bone marrow-derived TIL may be suitable as a source for CAR-based therapy for AML. In a model of AML, we will perform an extensive phenotypic, transcriptional, and functional characterization of anti-AML CAR-engineered TIL compared to conventional peripheral blood lymphocytes.

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

Self-amplifying RNA-engineered T cells for Anti-cancer Redirected Therapy (START): Pioneering the development of non-integrating and long-lasting CAR-T and TCR-T cells. 01/10/2023 - 30/09/2026

Abstract

Recently, interest has grown in using self-amplifying RNA (saRNA) in vaccines for infectious diseases and cancer. SaRNA is a type of messenger RNA (mRNA) that contains the non-structural proteins of an alphavirus replicase complex that amplifies the original strand of RNA and allows the expression of proteins of interest in the host cell without risk of infection. Compared to conventional mRNA, saRNA-mediated expression of proteins of interest may last for 28 days, while lacks the risks of genomic integration or cell transformation of integrative technologies such as viral vectors, transposons, and CRISPR-based knock-in. Moreover, saRNA vaccines under clinical investigation show that saRNA is safe and elicits robust immune responses. However, this technology has not been explored yet to genetically engineer effector immune cells, such as T cells, ex vivo. Therefore, the START project aims to investigate and optimize saRNA transfection as an innovative and potent technology for genetically engineering T cells with chimeric antigen receptors (CARs) and T-cell receptors (TCR) against different hematological and solid cancer antigens, with a thorough evaluation of antitumor activity, T cell fitness and potential transcriptomic and cell metabolic changes that could be related to saRNA replication activity. The START project will provide the basis for the future generation of non-integrative and long-lasting CAR-T and TCR-T cell therapies for hematological and solid malignancies.

Researcher(s)

Research team(s)

Project type(s)

  • Research Project

PrioriTCR - Prioritization of T-cell receptors for development of T-cell therapy using an immunoinformatics approach. 01/09/2023 - 31/08/2024

Abstract

The emergence of immunotherapy has improved cancer treatment in many different ways. One of the specific approaches is T-cell receptor (TCR)-T-cell therapy, in which potent TCRs are introduced into patient T cells in the laboratory, after which they can specifically destroy unwanted cells in the body. Although this therapy shows promising results, identification of potent TCRs remains a major hurdle. Due to the immense diversity of TCR repertoires, it is challenging to efficiently detect tumor-reactive T cells in the blood. In addition, TCRs are antigen-specific, meaning that different TCRs are required for different (sub)cancer types. The aim of project PrioriTCR is to develop an immunoinformatics platform that simplifies and accelerates the identification of potent TCRs. This proof-of-concept project is designed for the Wilms' Tumor 1 (WT1) antigen, which is overexpressed in a variety of solid tumors and blood cancers. WT1 is considered a virtually universal cancer marker, promising to target with specific immunotherapy. By combining limited laboratory experiments with blood samples from cancer patients with artificial intelligence, this project will result in the identification of new candidate potent WT1-specific TCRs for development of next-generation T-cell therapies. The developed computer models can be further extended for TCRs against other cancer markers.

Researcher(s)

Research team(s)

Project type(s)

  • Research Project

Unravelling the role of antigen-specific T cells in NMOSD and MOGAD. 15/03/2023 - 31/12/2025

Abstract

Neuromyelitis Optica Spectrum Disorders (NMOSD) and Myelin Oligodendrocyte Glycoprotein Antibody Associated Disease (MOGAD) are rare autoimmune diseases of the central nervous system (CNS) that are distinct from multiple sclerosis (MS), a more prevalent CNS autoimmune disease. Even though there is evidence for a key role of T cells in the pathogenesis of NMOSD and MOGAD, the focus of research has been directed more towards unravelling the role of autoantibodies. Detection of antigen-specific T cells in the peripheral blood of people with aquaporine-4 positive NMOSD and even more so in MOGAD, has been challenging so far. Identification and functional characterization of antigen-specific autoreactive T cells in the peripheral blood is a necessary step to demonstrate and strengthen the evidence for the pivotal role of T cells in the pathogenesis of NMOSD and MOGAD and may pave the way towards future development of antigen-specific T cell modulatory treatments.

Researcher(s)

Research team(s)

Project website

Project type(s)

  • Research Project

BCMA immunoPET to predict and monitor treatment response to CAR-based cellular therapies in multiple myeloma. 01/01/2023 - 31/12/2026

Abstract

Multiple myeloma is a rare form of white blood cell cancer of the bone marrow. While there is no cure, multiple myeloma can be managed successfully in many patients for years because of the growing availability of new drugs. Despite these advances, most current treatment strategies follow a one-size-fits-all approach and novel techniques to select patients for specific therapies are needed, especially considering the potential toxicity and cost of emerging immunotherapies (like CAR-based cellular therapies). Moreover, pockets of myeloma cells can exist within a patient with different sensitivity for a specific treatment, and single-site bone marrow biopsy may be less reliable to identify heterogeneous disease. Positron emission tomography (PET) provides a powerful platform to characterize tumors non-invasively by modifying and radio-labeling antibodies to image the tumor phenotype. In this preclinical project, we will develop, validate, and assess the predictive potential of a new antibody-based PET tracer to assess BCMA, a protein that is highly and selectively expressed on myeloma cells, offering our radiopharmaceutical unique specificity. Finally, a mouse model expressing human characteristics will be used to assess our tracer in a clinically relevant setting using CAR-based cell therapy. If successful, our tracer will help physicians select patients who can benefit from CAR therapy and avoid risking the severe side-effects in patients with a low likelihood of response.

Researcher(s)

Research team(s)

Project type(s)

  • Research Project

Towards the use of in vitro and in silico T-cell response prediction to guide the development of vaccines, using mRNA-based rabies vaccine as a proof-of-concept. 01/01/2023 - 31/12/2024

Abstract

The traditional process of designing and developing vaccines has been challenged dramatically by the COVID-19 pandemic, with the adoption of mRNA-based vaccines and a reduction of lengthy development pipelines from 10-15 years to 1-1.5 years. This creates a push for further innovation in vaccine development, in particular for diseases with a high unmet need. As an example, mortality due to rabies (a lyssavirus) remains unacceptably high. Although safe, effective vaccines are available for human and animal use, human vaccines are too expensive and generally inaccessible for widespread use in regions where the risk of bites from rabid animals is highest. mRNA approaches offer an opportunity to provide affordable vaccines with the possibility of manufacturing in low and middle income countries, with optimised design affording broader protection. The aim of such an approach would be to drive down cost and broaden supply and equity of access. Novel in silico approaches such as those analysing the T cell receptor response may permit insights into the immune response elicited by rabies vaccines, aid understanding of the mode of action and guide future use. The objective of the current project is to investigate if detailed analysis of the T-cell receptor response can be valuable to inform vaccine design and improve the vaccine development process, using a rabies mRNA vaccine as a proof-of-concept. We will combine in vitro (UAntwerp) and in silico (ImmuneWatch) techniques to gain insights into the T cell response against a range of experimental rabies mRNA vaccine constructs (Quantoom). The project is a public-private partnership aiming to explore novel approaches in vaccine development and to prepare future collaborations between the project partners.

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

SECRET - Evaluation of the neuro-protective properties of perinatal derivatives using murine iPSC-derived neurospheroids. 01/12/2022 - 30/11/2024

Abstract

Perinatal tissues encompass a large and diverse cell/tissue family obtained from term pregnancies. Perinatal derivatives (PnDs) include amniotic membrane, chorion, Wharton's jelly, amniotic fluid, and the cells isolated from these tissues, as well as the factors that these cells release. PnD have drawn much attention due to their immune-modulatory and tissue-protecting properties, making them attractive candidates in regenerative medicine. Focussing on neuro-protection, we will here investigate whether different types of PnDs are able to inferfere with inflammation-associated neuro-degenerative processes in murine induced pluripotent stem cell (iPSC)-derived neurospheroids. Neurospheroids cultured from iPSC represent an important research tool to study neuron-astrocyte interactions, during development, homeostasis and stress. Hereto, we developed a 5-week old murine iPSC-derived neurospheroid model containing mature neurons and astrocytes in order to evaluate the therapeutic potential of several neuro-protective/modulating compounds in vitro preceding animal experiments. Following development and characterization of this new murine iPSC-derived neurospheroid model, its sensitivity to immune signal-induced stress (a.o. stimulation with IL1b, TNF and/or LPS) has been demonstrated by monitoring astrocyte activation (a.o. production of IL6 and CXCL10). In this new project in collaboration with the 'Universita Cattolica del Sacre Cuore' (Rome, Prof. Ornella Parolini) and the 'Centro di Ricerca E. Menni' (Brescia, Dr. Antonietta Silini), we will now investigate the neuro-protective/modulating activity different PnDs on astrocyte activation in murine iPSC-derived neurospheroids. These studies will then allow to preselect the most potential PnDs for subsequent animal studies and/or human clinical trials in the field of neuro-degenerative disease.

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

Targeting the NF-κB pathway with targeted protein degradation for the treatment of hematological malignancies. 01/09/2022 - 31/08/2026

Abstract

Hematologic malignancies are cancers that primarily affect the blood, bone marrow and lymph nodes. Among the different subtypes, B-cell non-Hodgkin lymphoma (B-NHL) and Acute Myeloid Leukemia (AML) are the most prevalent indications. Common to both indications, alterations in the Nuclear Factor kappa-light-chain-enhancer of activated B cells (NF-κB) pathway are frequently observed, leading to constitutive activation and oncogenic signalling. Known key effectors within the pathway such as Bruton's tyrosine kinase (BTK), interleukin-1 receptor associated kinases (IRAKs), Myeloid differentiation primary response 88 (MYD88), and Mucosa-associated lymphoid tissue lymphoma translocation protein 1 (MALT1) represent promising therapeutic targets for these indications and have been at the center of significant drug discovery efforts. In order to tackle common limitations associated to canonical small molecule inhibitors (SMI) (e.g., resistance mutations, lack of response due to scaffolding functions, …), this project is aimed at exploring the therapeutic potential of selective target degradation through PROteolysis-TArgeting Chimera (PROTAC). PROTAC represents an innovative protein degradation technology able to induce protein degradation by taking advantage of the ubiquitin proteasome pathway. The goal of the project is to provide a better understanding of the therapeutic potential of PROTACs specific for BTK, IRAK1 and IRAK4 in comparison to their respective SMI counterparts. To this end, we will evaluate the molecular and functional consequence of target degradation or inhibition in relevant models of AML and B-NHL as well as reflect the work on AML primary patient material. In addition, potential synergistic activity between PROTACs and clinically relevant SOC/SMIs options will be evaluated. Last, potential on-target/off-tumor activity in immune cell sub-types in which the NF-kB pathway is known to play a role (e.g., T-cells) will be evaluated.

Researcher(s)

Research team(s)

Project type(s)

  • Research Project

Refining the efficacy of systemic administration of bioactive molecules for Parkinson's Disease (RePark) 01/09/2022 - 31/08/2025

Abstract

Parkinson's Disease (PD) is a neurodegenerative disease primarily linked to ageing affecting the psychomotor functions. An impairment in the activity of dopaminergic neurons (aggregation and intracellular accumulation of alfa-synuclein), is at the basis of this functional loss. Current therapies do not revert the PD progression but stand on the relief of the symptoms. Importantly, the efficacy of new therapeutic approaches is hampered by the lack of in vitro models that mimic the PD's hallmarks. Moreover, the effectiveness of therapeutic approaches is hampered by the low ability of the drugs to cross the blood- brain barrier (BBB). RePark will develop an in vitro PD model that combines a mimic of the BBB, the brain's extracellular matrix (ECM) and brain cells. With this system it will be possible to overcome the disadvantages of using animal models and to recapitulate in vitro the cellular characteristics of PD, as well as to assess the delivery and efficacy of new therapeutic drugs.

Researcher(s)

Research team(s)

Project type(s)

  • Research Project

Bioreactor infrastructure for upscaled culture of organoids and tumoroids. 01/06/2022 - 31/05/2024

Abstract

In this application, we request financing for three benchtop CERO 3D Cell Culture Bioreactor units for the culture of 3D cell cultures, including spheroids and organoids, that are increasingly being used in biomedical research. Currently, 3D organoids and spheroids are cultured in traditional cell culture plates under static or shaking (using orbital shaker) conditions in a standard CO2 cell culture incubator, which is suboptimal for long-term and large-scale culture of spheroids and organoids. A bioreactor system would take organoid and spheroid culture at the campus to a next level in terms of quality (improved viability, maturation and homogeneity) as well as quantity. Each CERO 3D Cell culture bioreactor unit can maintain four 50 mL organoid cultures, including monitoring and control of temperature, pH and carbon dioxide levels. In total, the envisaged bioreactor infrastructure will be able to accommodate twelve simultaneous organoid cultures under highly controlled conditions. The envisaged CERO 3D Bioreactor units will be applied for multiple research domains at the University of Antwerp, and more specifically for upscaled culture of stem cell-derived spheroids and organoids, tumoroids derived from primary tumour material of patients, stem cell-derived cardiomyocytes, stem cell-derived cartilage tissue and intestinal organoids. Furthermore, based on our own experience in upscaled organoid culture, the instalment of bioreactor units has become an urgent need to progress towards future valorisation activities.

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

Preclinical development of novel chimeric antigen receptor (CAR)-modified cell-based immunotherapy approaches for hematological malignancies. 01/10/2021 - 30/09/2025

Abstract

Most CAR-T cell therapies for treatment of multiple myeloma (MM) are directed towards BCMA, a target antigen that is highly expressed on malignant plasma cells. It has become evident that the extracellular BCMA-binding domain of a CAR, derived from a monoclonal antibody (mAb), is an important determinant of clinical efficacy of anti-BCMA CAR-T cell therapies. In this project, we want to further investigate and compare anti-BCMA mAbs from different animal species for their competence for incorporation in CAR-T cell therapies against MM. In addition, most CAR-T cells get their genetic material delivered via viral transduction or transposons. A major disadvantage of viral loading methods are the need for highly specialized infrastructure and the long time needed for production. The speed up the production process, make it safer and to counteract virus-mediated insertion of DNA in the genome, we want to use episomal vectors. These vectors exist extrachromosomal, yet are duplicated and thus passed along to daughter cells. Next, we want to turn on/off specific CARs by (de)methylating the genes, thereby regulating transcription. It also enables speeding up and optimization of the production process by incorporating different CARs in a single T-cell, whereby only the CAR of choice is switched on. This could be incorporated into allogenic cell therapy in which different CARs are present in an off-the-shelf allogenic T-cell/NK-cell.

Researcher(s)

Research team(s)

Project type(s)

  • Research Project

MulTplex project: Development of a combinatorial multi-epitope T-cell receptor (TCR), non-signaling chimeric antigen receptor (NSCAR) and immunosuppressive immune checkpoint (IICP)-disrupted adoptive T-cell therapy against leukemia. 01/10/2021 - 30/09/2025

Abstract

Acute myeloid leukemia (AML) is one of the most common leukemias in adults with a 5‐year overall survival rate of only 30%. Despite therapeutic advances in the last decade, novel adoptive T-cell immunotherapies using anti-tumor chimeric antigen receptors (CARs) and T-cell receptors (TCRs) are not fully developed for AML. Moreover, expression of immunosuppressive immune checkpoints (IICPs) hinder the success of these T-cell therapies. To address this issue, the aim of this project is to develop an innovative multi-epitope Wilms' tumor 1 (WT1)-specific TCR, CD200-specific non-signaling chimeric antigen receptor (NSCAR) and IICP-disrupted (mulTplex)-engineered adoptive T-cell therapy for AML. We will combine TCRs with different human leukocyte antigen (HLA) restrictions and specificities against diverse epitopes of WT1, a key intracellular antigen, in a multi-epitope strategy. To avoid the interaction between native and introduced TCRs, native TCRs will be disrupted by CRISPR-Cas9 technology. The NSCAR, which lacks the typical CAR's signaling domain, will act as an "anchor" for the T cells by locking onto AML cells through CD200, a novel extracellular AML antigen, and without triggering T-cell activation. By doing so, we expect to improve TCR-mediated anti-AML cytotoxic capacity of mulTplex-engineered T cells. To further harness the anti-leukemic activity of engineered T cells, AML-associated IICPs will also be disrupted using CRISPR-Cas9 methods. Both in vitro and in vivo evaluation of mulTplex-engineered T cells will ensure translation of our innovative combinatorial approach into clinical studies.

Researcher(s)

Research team(s)

Project type(s)

  • Research Project

Post-transplant infusion of CD19 CAR-armored donor lymphocytes in adult patients with B-cell hematological malignancies: a phase I/IIa study. 01/10/2021 - 31/10/2024

Abstract

Allogeneic hematopoietic stem cell transplantation (HSCT) is an established treatment modality for patients with relapsed/refractory (r/r) CD19+ B-cell hematological malignancies, such as acute lymphoblastic leukemia (ALL) or non-Hodgkin's lymphoma (NHL). The prognosis of patients in whom the disease is not under control or has relapsed after HSCT, is particularly grim. For these patients, the therapeutic options are limited. One of the few available salvage strategies involves the use of donor lymphocyte infusions (DLI). The mechanism of action of DLI relies on the administration of immune effector cells, predominantly T cells, from the stem cell donor, with the ultimate goal to elicit a "graft-versus-leukemia" or "graft-versus-tumor" effect. Unfortunately, DLI have only modest clinical activity and can evoke or exacerbate serious transplant-related side effects such as "graft-versus-host" disease. CD19-targeted chimeric antigen receptor (CAR)-T cell therapy offers new hope for patients with r/r B-cell hematological malignancies. Here, T-cells derived from the patient (autologous) are genetically modified to express a CD19 CAR, a synthetic receptor enabling binding of the cells to the CD19-expressing target cells. Upon engagement, the CAR will trigger activation of the T cells which will then become cytotoxic towards the target cells. In this project, DLI products will be loaded with an in-house developed and optimized CD19 CAR. By using allogeneic cells derived from healthy donors as source for CAR-T cell manufacturing, which are usually "fitter" than autologous, patient-derived T cells, we aim to enhance the anti-tumoral activity of the CAR-T cells.

Researcher(s)

Research team(s)

Project type(s)

  • Research Project

Identification and validation of T cell receptor (TCR) pairs for TCR-T cell therapy for AML and NHL (HEME TCR). 01/06/2021 - 31/12/2024

Abstract

Confidential .

Researcher(s)

Research team(s)

Project type(s)

  • Research Project

Immunological control of Varicella zoster virus (VZV)-infected iPSC-derived brain models by steady-state and immune-compromised astrocytes and microglia. 01/01/2021 - 31/12/2024

Abstract

Varicella zoster virus (VZV) is a member of the herpesvirus family and is a highly successful and ubiquitous human pathogen. Both in children and in adults, varicella-related complications may lead to hospitalisation. While in children direct neurological complications may occur following primary infection (varicella), in adults vasculitis and neurological complications are not uncommon following reactivation of latent VZV (herpes zoster). With a clear link between VZV and neuropathology, it is inevitable that the immune system of the central nervous system (CNS) will be challenged by VZV. However, to date little is known about how astrocytes and microglia behave upon encounter of VZV in the CNS. In this project, we will address this question using an established human in vitro model of axonal infection of human induced pluripotent stem cell (hiPSC)-derived CNS neurons with fluorescent reporter VZV stains. Using this model, we will first longitudinal monitor how hiPSC-derived astrocytes and microglia influence the processes of VZV infection, latency and reactivation. Next, using iPSC models derived from VZV patients with mutations in POLRIII, we will investigate whether immune compromised astrocytes and/or microglia can control neuronal VZV infection. Altogether, these studies will help us understanding innate immune control of VZV in the CNS, and will allow - beyond the scope of this project – to develop novel strategies to prevent VZV spreading in the CNS.

Researcher(s)

Research team(s)

Project type(s)

  • Research Project

Deep phenotyping of cellular heterogeneity and maturation in human iPSC-derived brain organoids and cardiomyocytes. 01/01/2021 - 31/12/2024

Abstract

Pluripotent stem cell (PSC) technology is increasingly gaining interest for modelling diseases and developing precision therapeutics. However, the immaturity and heterogeneity of PSC-derived cell populations impinge on the reproducibility and preclude their use for comparative and diagnostic analyses. Recognizing these caveats for their own human induced (hi)PSC-based research, the Laboratories of Experimental Hematology, Cardiogenetics and Cell Biology and Histology have teamed up to develop a pipeline that enables accurate phenotypic staging of hiPSC-derived cell culture models. Specifically, to create mature and standardized 2D and 3D-models for cardiomyocyte cultures and brain organoids, our consortium proposes to optimize the culture conditions and refine the interrogation methods. Longitudinal follow-up and validation of functional activity in the differentiation products will be achieved by means of high-throughput multi-electrode array recordings and live cell calcium and voltage imaging. In parallel, cellular heterogeneity will be mapped with quantitative immunofluorescence and transcriptome analyses. The correlation of functional and molecular readouts will allow establishing a biomarker panel for mature hiPSC-derived cardiomyocyte models and brain organoids. Thus, with this work, we intend to develop more reproducible models and to allow selecting only those with the highest functional maturity, a prerequisite for our future stem cell research aimed at studying and treating neuro-inflammatory and cardiogenetic disorders.

Researcher(s)

Research team(s)

Project type(s)

  • Research Project

Optimization of chimeric antigen receptor (CAR) design for improved cellular immunotherapy of hematological diseases. 01/11/2020 - 31/10/2024

Abstract

Chimeric antigen receptor (CAR)-T cell therapy has demonstrated unprecedented clinical activity in patients with hematological diseases, but a large proportion of them will ultimately relapse. Further optimization of this new treatment modality is therefore required to unlock its full therapeutic potential. In this project, in addition to using readily available cell line models, we will use our mRNA electroporation technology for CAR loading of immune cells. This will provide a rapid and efficient way to explore new research paths that can lead to optimized CAR-based cellular therapies for hematological diseases. Will assess the value of a multi-targeted approach incorporating two established CAR targets (CD19 and B-cell maturation antigen) and the novel CAR candidate CD200. Next, the hinge and co-stimulatory domains in the CAR structures will be sequentially modified, comparing conventional hinge and co-stimulatory domains with our recently discovered 4-1BB-hinge and CD26 co-stimulatory domains. Exhaustion will be prevented by introducing programmed death (PD-1) silencing RNA in the CAR-modified cells to reduce PD-1-mediated co-inhibitory signaling. Finally, positive findings will be translated from our cell line models to conventional T cells, NK cells and gdT cells.

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

Therapeutic dendritic cell vaccination to improve frontline treatment of adult patients with glioblastoma multiforme (GBM) and pediatric patients with GBM and diffuse intrinsic pontine glioma (DIPG). 01/01/2020 - 31/12/2025

Abstract

Based on the strong need for more targeted, tolerable and durable treatment strategies that could postpone or even prevent recurrence of disease in the most common adult malignant brain tumor, we embarked on a phase I/II clinical trial assessing frontline treatment with autologous dendritic cell (DC) vaccines loaded with glioblastoma-associated tumor antigen Wilms' tumor 1 in conjunction with conventional chemoradiation following surgery in adults with glioblastoma multiforme (GBM; NCT02649582). Childhood high-grade glioma (HGG, including GBM) and diffuse intrinsic pontine glioma (DIPG) are rare aggressive brain tumors. In the absence of a standard of care, treatment is mostly adapted from adult schedules, resulting in 5-year survival rates of less than 5% and 1% after diagnosis, respectively. With limited advanced investigational treatment options for this vulnerable patient population, we strive to extend our clinical study to the pediatric application. Ultimately working towards the clinical valorization of an adjuvant DC-based immunotherapy approach, health care evaluation is warranted. To this extent, we will include collection of patient-reported outcome on how the study therapy is experienced throughout time in the response evaluation of all study patients. As the search for biomarkers is gaining momentum in the rapidly evolving cancer immunotherapy landscape, we are also continuously expanding the screening assays on clinical patient material. The present project proposal is designed to allow completion of the intended adult GBM patient recruitment number and to extend the trial, innovating on the pediatric application of DC vaccination, health care evaluation and emerging therapeutic biomarker research. Within the context of hard-to-treat brain tumors, this study and its specific design will add a new dimension to our translational and clinical DC vaccine programs by investigating whether DC vaccination can be combined with first-line chemoradiation treatment of adult GBM and childhood HGG and DIPG patients and whether this combination leads to tumor-specific immune responses and improved survival. Exploration of patient-reported outcomes will help to improve symptom management, functional status and overall quality of life and will provide necessary information for future clinical valorization of this type of personalized medicine. In depth research on clinically valuable biomarkers will allow us to make a significant contribution to the broader (immunotherapy-oriented) scientific community.

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

Immune effector cell therapy for hematological malignancies with a focus on acute myeloid leukemia and multiple myeloma. 01/10/2019 - 30/09/2024

Abstract

Acute myeloid leukemia (AML) and multiple myeloma (MM) are two types of blood cancers with a high unmet therapeutic need. The knowledge that cells of our immune system can recognize and kill cancer cells has laid the foundation for immune effector cell (IEC) therapy. This involves the infusion of immune cells that are "armored" outside the body with a T-cell receptor (TCR) or a chimeric antigen receptor (CAR). Such TCR- or CAR-loaded immune cells can execute a targeted attack against cancer cells. The aim of the present project is to improve the therapeutic efficacy of IEC therapy for AML and MM, while reducing the risk of side effects and costs of treatment. More specifically, immune cells will be weaponed with AML-directed TCRs or MM-directed CARs via a technique called electroporation. This involves the application of an electrical pulse to the cells, making temporary holes in their surface and enabling their loading with the TCR or CAR. When compared to the current IEC therapies, this novel procedure will allow for the generation of IECs with reduced costs, improved safety profile and enhanced anti-tumor activity. It is therefore expected that this research project will make an important contribution to the development of the next-generation IEC products for AML and MM.

Researcher(s)

Research team(s)

Project type(s)

  • Research Project

Safety and feasibility evaluation of first-in-human interleukin-15-transpresenting Wilms' tumor (WT1)-targeting autologous dendritic cell vaccination in cancer patients. 01/01/2019 - 31/12/2024

Abstract

Improvement of first-line treatment for cancer patients with a high tumor recurrence rate and low effective treatment options, such as pancreatic cancer (PC), is warranted. Pancreatic cancer is a devastating disease with a 5-year survival rate below 5%, depending on the specific stage of disease when it is diagnosed, rendering it the 4th most common cause of cancer-related death worldwide. Even those who are eligible for curative-intent resection and conventional adjuvant treatment will nearly all die of their disease due to the high tendency towards recurrence. Adjuvant treatment with gemcitabine after resection of PC decreases recurrence rate, but the disease-free survival of these patients stays dismal with a 5-year survival rate below 21%, underscoring the need for new adjuvant regimens. The combination of gemcitabine with immunotherapy might improve outcome as suggested by some studies, but available data is so far limited to a few early-phase uncontrolled clinical trials. Interleukin (IL)-15-transpresenting dendritic cells (DCs) are a promising armament for immunotherapy of PC. Complementary to current treatments, DCs as quintessential antigen-presenting cells of the immune system can activate the antitumor immune system to attack pancreatic cancer cells. Preclinical data demonstrate the therapeutic potential of these innovative IL-15-transpresenting DCs evidenced by superior activation of the antitumor immune system to attack cancer cells. Since this will be the first-in-human use of IL-15-transpresenting DCs, the objectives are to test the safety, feasibility and immunopotency in patients with refractory solid tumors, the prototypic cancer patient population for phase I trials. This phase I clinical study is pivotal for future testing of this promising IL-15-transpresenting DC vaccine as adjuvant therapy to current anticancer regimens aiming to improve the standard of care of cancer patients with a high unmet medical need.

Researcher(s)

Research team(s)

Project type(s)

  • Research Project

Past projects

EHA Kick-off Grant - Self-amplifying RNA-engineered T cells for Anti-leukemic Redirected Therapy (START): Pioneering the development of non-integrating and long-lasting CAR-T-cell immunotherapies. 03/04/2023 - 02/04/2024

Abstract

Recently, interest has grown in using RNA in vaccines for infectious diseases and cancer. In this project, we will investigate a new type of messenger RNA (mRNA) for genetic engineering of T cells with chimeric antigen receptors (CARs) and T cell receptors (TCRs) against hematological and solid cancers. This novel mRNA allows prolonged expression of the protein of interest compared to conventional mRNA without the risks of insertional mutagenesis present in integrating engineering technologies such as viral transduction. We will conduct an in-depth analysis of T cell fitness after RNA engineering and thoroughly evaluate the antitumor activity of the RNA-engineered CAR-T and TCR-T cells. This project will provide the basis for the future generation of safer and more durable cellular therapies for hematological and solid cancers.

Researcher(s)

Research team(s)

Project type(s)

  • Research Project

BDNF-engineered regulatory T cells, a future regenerative cell-therapeutic strategy? 01/04/2023 - 31/03/2024

Abstract

The ultimate goal of this research project is to develop a clinically safe and regenerative cell-based vaccine for the treatment of (progressive) multiple sclerosis (MS), since remyelination remains a major unmet need. We recently succesfully developed "designer" Tregs that are engineered to express high levels of BDNF. Here, we are ready to assess the remyelinating capacity of these transgenic Tregs, hypothesizing that these Tregs will excel in their pro-regenerative properties, driving oligodendrocyte differentiation and remyelination, beyond immunomodulation. This would represent a breakthrough for MS healthcare.

Researcher(s)

Research team(s)

Project type(s)

  • Research Project

Computer-Aided Design of optimized Chimeric Antigen Receptors for hematological malignancies (CADCAR project) 29/03/2023 - 31/12/2023

Abstract

Chimeric antigen receptor (CAR)-T-cell therapy has achieved remarkable clinical response rates in relapsed/refractory B-cell malignancies. Unfortunately, the frequency of relapse remains high as a result of decreased cellular fitness, poor anti-tumor activity or a lack of persistence of the CAR-T-cell product. While the CAR architecture is a foundational driver of CAR-T-cell responses, its design is poorly understood, in particular for the structural hinge domain. Current hypothesis-driven workflows are low in throughput, expensive and laborious, and complicate pattern recognition. This project aims to define CAR hinge domain design rules by studying the relationship between hinge properties and CAR-T-cell responses in the context of hematological malignancies. We employ high-throughput cellular assays to phenotypically and functionally evaluate a large library of novel hinge domain candidates. A machine learning algorithm will be trained to correlate hinge domain characteristics with the obtained cellular outputs. We anticipate to create an algorithm that is capable of predicting superior hinge domains from a naïve set of candidates against a multitude of target antigens. We envision that in the future this model can be further trained to include other CAR domains and domain combinations to assist in further personalization of CAR-T-cell therapy.

Researcher(s)

Research team(s)

Project type(s)

  • Research Project

Modulation of astrocyte activation in murine iPSC-derived neurospheroids. 01/01/2023 - 31/12/2023

Abstract

Neurospheroids cultured from induced pluripotent stem cells (iPSC) represent an important research tool to study neuron-astrocyte interactions, during development, homeostasis and stress. In course of the EU MSCA ITN PMSMatTrain project, the UAntwerp partner developed a 5-week old murine iPSC-derived neurospheroid model containing mature neurons and astrocytes in order to evaluate the therapeutic potential of several neuro-protective/modulating compounds in vitro preceding animal experiments. Following development and characterization of this new murine iPSC-derived neurospheroid model, its sensitivity to immune signal-induced stress (a.o. stimulation with IL1b, TNF and/or LPS) has been demonstrated by monitoring astrocyte activation (a.o. production of IL6 and CXCL10). This SEP grant will be applied to support the 4th PhD year of Julia Di Stefano in which the candidate will investigate the neuro-protective/modulating activity of APRIL (delivered by means of AAV vectors) and placenta stem cell-derived factors (delivered by means of extracellular vesicles) on astrocyte activation in murine iPSC-derived neurospheroids. Finally, it is expected that the technology developed here will become an important additional tool in (i) fundamental research to study neuro-development and functioning, as well as (ii) to preselect interesting therapeutic molecules before proceeding to animal studies and/or human clinical trials.

Researcher(s)

Research team(s)

Project type(s)

  • Research Project

Learning the design language of CAR structural components using a high-throughput screening method. 01/09/2022 - 28/02/2023

Abstract

Chimeric antigen receptor (CAR)-T cell therapy is an innovative form of cellular immunotherapy that utilizes T-lymphocytes that are genetically engineered to express a CAR. While initial response rates are often outstanding, the majority of patients suffers from a relapse which is often cause by a lack of sustained effector functionality or persistence of the CAR-T cells. The importance of the CAR architecture to therapeutic outcome is becoming increasingly clear. However, it is unlikely that the full potential of CAR-T-cell therapy will be reached by using a combination of domains derived from only a small subset of immune-related proteins, as is the case today. The main reason behind this limited selection of building blocks is the use of slow and labor-intensive low-throughput methods for the evaluation of novel candidate domains. Only few groups, such as the Birnbaum lab, have developed a high-throughput workflow for the functional evaluation of up to 1 million CAR designs. While those workflows provide invaluable information novel costimulatory domain combinations, it has never been applied to other structural domains of CARs. The aim of this research stay at the Birnbaum lab is to acquire the practical know-how on CAR combinatorial library construction and a high-throughput CAR screening workflow. By applying this knowledge directly under the supervision of the host group, we will attempt to decipher the design language of severely understudied CAR components by screening a sizeable library of potentially valuable CAR domains.

Researcher(s)

Research team(s)

Project type(s)

  • Research Project

Replic-ON project – Pioneering the investigation of self-amplifying messenger RNA electroporation for long-lasting ex vivo transfection of immune cells. 01/04/2022 - 31/03/2023

Abstract

Recently, there has been growing interest in the use of self-amplifying mRNA (saRNA) in therapeutic vaccines for infectious diseases and cancer. SaRNA is a type of messenger RNA (mRNA) that contains the non-structural proteins (nsP1-4) of an alphavirus replicase that copies the original strand of mRNA upon delivery into the cell. The nsP1-4 replicon is followed by a subgenomic promoter and the sequence of a gene of interest, allowing the expression of proteins of interest in the host cell. The self-replicating property means that proteins of interest encoded in the transfected saRNA will be expressed for a longer period of time compared to conventional mRNA. However, since there is no integration into the genome of the host cell, insertional mutagenesis is prevented. Thus, saRNA-based strategies combine the best of stable viral- or non-viral-based and transient mRNA-based engineering strategies. SaRNA is usually delivered in vivo as "naked" saRNA with or without intradermal electroporation or formulated into nanoparticle vaccines, with which expression of the protein of interest may last for 28 days. However, the exploitation of this technology for ex vivo modification of T cells in a therapeutic product has never been explored thus far. The primary objective of the Replic-ON project is to explore saRNA transfection as an innovative technology for genetically engineering immune cells in the context of the development of cell-based therapies. If successful, this project will provide groundbreaking data for the further development of ex vivo saRNA transfection technology as an amenable approach for T-cell genetic engineering in larger fundamental research project applications. We expect that this project will be the cornerstone for the much-needed development of more efficient and long-lasting non-integrating cellular immunotherapies while straddling the boundary between short-lived conventional mRNA technologies and integrating technologies such as viral transduction. Finally, this pioneering research would consolidate our leadership on ex vivo saRNA-based cellular therapies within the research community.

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

Encouraging grant 2021 'Rosa Blanckaert' for young researchers: (Bio)Medical Sciences. 01/12/2021 - 31/12/2023

Abstract

Chimeric antigen receptor (CAR)-T cell therapy has demonstrated unprecedented clinical activity in patients with hematological diseases, but a large proportion of them will ultimately relapse. Further optimization of this new treatment modality is therefore required to unlock its full therapeutic potential. In this project, in addition to using readily available cell line models, we will use our mRNA electroporation technology for CAR loading of immune cells. This will provide a rapid and efficient way to explore new research paths that can lead to optimized CAR-based cellular therapies for hematological diseases. Will assess the value of a multi-targeted approach incorporating two established CAR targets (CD19 and B-cell maturation antigen) and the novel CAR candidate CD200. Next, the hinge and co-stimulatory domains in the CAR structures will be sequentially modified, comparing conventional hinge and co-stimulatory domains with our recently discovered 4-1BB-hinge and CD26 co-stimulatory domains. Exhaustion will be prevented by introducing programmed death (PD-1) silencing RNA in the CAR-modified cells to reduce PD-1-mediated co-inhibitory signaling. Finally, positive findings will be translated from our cell line models to conventional T cells, NK cells and gdT cells.

Researcher(s)

Research team(s)

Project type(s)

  • Research Project

MulTplex project: Innovating on combinatorial genetically engineered adoptive Tcell therapies against hematological diseases. 01/11/2021 - 31/10/2022

Abstract

Genetic engineering of lymphocytes for adoptive cell transfer has marked a turning point in personalized immunotherapy, especially in the treatment of cancer. Adoptive T-cell immunotherapies using antitumor chimeric antigen receptors (CARs) and T-cell receptors (TCRs) have, however, not met expectations yet for the majority of malignancies, including acute myeloid leukemia (AML). Moreover, expression of immunosuppressive immune checkpoints (IICPs) hinders the success of these therapies. To address the shortcomings of current T-cell therapies, the aim of this research project is to develop an innovative combinatorial and genetically engineered adoptive T-cell therapy focusing on AML as a disease model. In this project four important issues will be covered. First, cancer cells capitalize on processes such as downregulating peptide-major histocompatibility complex (pMHC) ligands to lower their immunogenicity and, by doing so, evade immune detection. Second, TCRs that target tumor self-antigens are scarce and usually have low affinities, having difficulties in binding target tumor antigens. Finding ways to improve interaction between pMHC ligands and low affinity TCRs, such as those that target self-antigens, would improve the chance of success in TCR-engineered T-cell therapies. Third, adoptive T-cell therapies are confronted with immunosuppressive environments that hinder their efficacy via engagement of IICPs, such as PD-1, TIM-3, or LAG-3. Determining the most relevant IICPs is key for developing effective adoptive T-cell therapies. Fourth, these therapies must be tumor-specific and efficacious once translated into a clinical setting. Taken together, combinatorial and flexible approaches for TCR-engineering will mark the next-generation of T-cell immunotherapies, by addressing (a) improved interaction between T cells and cancer cells, (b) immune evasion through IICPs, (c) cost-effectiveness of an all-in-one therapy, and (d) safety using RNA-based methods. In summary, improved adoptive T-cell therapies that overcome CAR and TCR challenges as well as the immunosuppressive environment that hinders antileukemic T-cell action will facilitate innovative solutions for cancer treatment.

Researcher(s)

Research team(s)

Project website

Project type(s)

  • Research Project

ANCHOR project - Antigenspecific non signaling CARs as hemato-oncological remedy. 22/06/2021 - 22/06/2022

Abstract

Acute myeloid leukemia (AML) is a rare type of cancer that predominantly affects people in the third age. The 5‐year overall survival rate of AML patients is only 30%, a figure that has not substantially changed despite enormous therapeutic advances in the last decade. Novel immunotherapies, such as T-cell receptor (TCR) T-cell and chimeric antigen receptor (CAR) T-cell therapies, are difficult to adopt in the context of AML. This is because most AML-related antigens are intracellular self-antigens that are expressed on the AML cell surface as peptides via major histocompatibility complexes (MHC); TCRs specific for these self-antigens are difficult to obtain since self-reactive T cells undergo thymic negative selection. In contrast to CD19 which is a very suitable extracellular target antigen for CAR-T cell therapy in acute lymphoblastic leukemia (ALL), the very few extracellular antigens expressed on AML cells that can serve as targets for CAR-T cell-based therapies, such as CD33 and CD123, are also expressed on normal hematopoietic stem/progenitor cells entailing a risk of intolerable myeloablation. The aim of this innovative project is to combine the best of two worlds, namely to redirect T-cells towards the key intracellular AML antigen Wilms' tumor protein 1 (WT1) using WT1-specific TCRs, combined with an innovative non-signaling CAR (NSCAR) towards a novel candidate extracellular AML antigen.

Researcher(s)

Research team(s)

Project type(s)

  • Research Project

Antigenspecific non-signaling CARs as hemato-oncological remedy (ANCHOR project)(GILEAD award). 17/02/2021 - 31/12/2021

Abstract

Acute myeloid leukemia (AML) is a rare type of cancer that predominantly affects people in the third age. The 5‐year overall survival rate of AML patients is only 30%, a figure that has not substantially changed despite enormous therapeutic advances in the last decade. Novel immunotherapies, such as T-cell receptor (TCR) T-cell and chimeric antigen receptor (CAR) T-cell therapies, are difficult to adopt in the context of AML. This is because most AML-related antigens are intracellular self-antigens that are expressed on the AML cell surface as peptides via major histocompatibility complexes (MHC); TCRs specific for these self-antigens are difficult to obtain since self-reactive T cells undergo thymic negative selection. In contrast to CD19 which is a very suitable extracellular target antigen for CAR-T cell therapy in acute lymphoblastic leukemia (ALL), the very few extracellular antigens expressed on AML cells that can serve as targets for CAR-T cell-based therapies, such as CD33 and CD123, are also expressed on normal hematopoietic stem/progenitor cells entailing a risk of intolerable myeloablation. The aim of this innovative project is to combine the best of two worlds, namely to redirect T-cells towards the key intracellular AML antigen Wilms' tumor protein 1 (WT1) using WT1-specific TCRs, combined with an innovative non-signaling CAR (NSCAR) towards a novel candidate extracellular AML antigen.

Researcher(s)

Research team(s)

Project type(s)

  • Research Project

COVID19 - ONCO-VAC, Vaccination against Covid-19 in cancer patients under active treatment (Belgium, B-VOICE) 01/02/2021 - 31/12/2022

Abstract

COVID-19 is a disease caused by an infectious outbreak of SARS-CoV-2. This viral SARS-CoV-2 infection can present itself in a broad spectrum of clinical features, ranging from asymptomatic, sensation of a mild cold or flu to severe bilateral pneumonia and death. Cancer patients are at high risk to develop serious illness after infection with SARS-CoV-2. Therefore, it is of high importance to protect these patients by following hygiene measurements and social distancing. But, as indicated by the guidelines of the Belgian and European Society for Medical Oncology, it is also important to vaccinate cancer patients. Although, not many studies that elevated the vaccine efficacy of COVID-19 vaccines in cancer patients have been performed. Due to the cancer or the treatment, it could be possible that the efficacy of the vaccines is lower in cancer patients or that they develop more side effects as a result of vaccination. To investigate this, we will monitor the reaction of the immune system of current and ex oncological and haematological patients on the different COVID-19 vaccines (Pfizer, Moderna, AstraZeneca, Janssen Pharmaceutica). The adverse effects as a reaction on vaccination will be investigated in this population as well. Clinical data of the patients will be collected and a blood drawn will be performed at different time points: before vaccination and 4, 6 and 12 months after receiving the first vaccination dose. The primary endpoint of the study is the quantification of different anti SARS-CoV-2 specific IgG antibodies per study cohort at 4 months after the first vaccination. The secondary endpoints of the study are to measure the SARS-Cov-2 specific T cell response and to investigate the evolution and duration of the cellular immune response after vaccination in the patient cohort. Another secondary endpoint is to analyse the titers of neutralizing antibodies both 4,6 and 12 months after receiving the first vaccination dose. Furthermore, it is aimed to investigate the efficacy of the immune response in the patient cohort for each different vaccine. This will be assessed by the SARS-CoV-2 infection rate based on information collected through questionnaires on incidence of (PCR-confirmed or chest CT scan confirmed) SARS-CoV-2 infection within a time frame of 12 months after the start of the study. At last, we will investigate the safety of the different COVID-19 vaccines that are commercially available in Belgium. Safety will be reported in terms of incidence and severity of adverse effects (AEs) using a questionnaire. Patients will be asked to report their adverse events over a period of 3 days after the vaccination day. This research project will provide knowledge on how the immune reaction after vaccination develops in cancer patients and patients with oncological or haematological history. The team of prof Lion of the Laboratory of Experimental Hematology, focusses on the SARS-Cov-2 specific cellular immunity research.

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

Evaluation of a novel hinge and co-stimulatory domain in BCMA-targeted chimeric antigen receptor T cells. 01/04/2020 - 31/03/2021

Abstract

The field of chimeric antigen receptor (CAR)-T-cell immunotherapy has evolved tremendously over the past decades. One of the milestones in CAR development was the incorporation of co-stimulatory domains, providing the necessary signaling to trigger full T-cell activation. Given their improved performance, these so-called second-generation CAR-T cells equiped with CD28- or 4-1BB -based intracellular domains dominate the clinical trial landscape. For multiple myeloma, the second most common type of blood cancer, early-phase clinical trials with B-cell maturation antigen (BCMA)-targeted CAR-T cells have demonstrated promising results, but relapses are frequently observed. Therefore, a great deal of research attention is currently being paid at improving the efficacy of the CAR-T cells. In the proposed project, we want to evaluate the potential of CD26 as a co-stimulatory domain in BCMA-targeted CARs. CD26 has been shown to improve persistence and anti-tumor activity of T cells, and is associated with a memory phenotype. In addition, the impact of the hinge domain on CAR-T-cell efficacy has been poorly studied, although the hinge domain has been shown to be important for antigen recognition. Therefore, we developed a novel 4-1BB-based hinge in collaboration with colleagues at Fudan University, Shanghai, China. This new hinge was already validated in the context of anti-HER-2, -GPC3 and -CD19 CAR-T cells, and will be further evaluated here in multiple myeloma.

Researcher(s)

Research team(s)

Project type(s)

  • Research Project

Applying bioinformatics in dendritic cell-mediated immune cell activation research: automated analysis of high-dimensional flow cytometry datasets. 01/04/2020 - 31/03/2021

Abstract

Can the bioinformatics R package FlowSOM – for high-dimensional single-cell flow cytometry datasets – assist research on dendritic cell-mediated immune cell activation? The primary objective of this Small Project is to evaluate the R package FlowSOM for the analysis of high-dimensional flow cytometry data and to explore its use in the preclinical and clinical evaluation of immunogenicity of next-generation anticancer dendritic cell vaccine candidates that are currently under investigation at the Laboratory of Experimental Hematology (UAntwerp) and the Center for Cell Therapy and Regenerative Medicine (Antwerp University Hospital). With increasing dimensionality of biological data and technical advances, manual flow cytometry data analysis will become inadequate. Applying bioinformatics, automated and unbiased comparisons between in vitro/ex vivo-stimulated immune effector cells with novel dendritic cell vaccine candidates will assist further development of potent dendritic cell preparations with the most superior immune-stimulating capacities and will be essential in unraveling therapy responsive immune profiles in longitudinal studies. FlowSOM is a powerful algorithm that builds self-organizing maps (SOMs) to provide an overview of marker expression on all cells and reveal cell subsets that could be overlooked with manual gating. Ultimately, our aim is to develop an advanced immune profiling platform for evaluation of preclinical and clinical dendritic cell-mediated immune responses.

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

Preclinical development of a next-generation CAR-T cell therapy for multiple myeloma. 01/11/2019 - 31/10/2020

Abstract

In Belgium, two patients receive the diagnosis of multiple myeloma (MM) each day. Despite considerable therapeutic advances over the past decades, MM remains incurable. Drug resistance often leads to refractory disease and relapses. Therefore, there is an urgent need for novel treatment methods for MM. Immunotherapy has become an important asset in the treatment of various cancers, including MM. Chimeric antigen receptor (CAR)-T cell therapy has attracted much attention in recent years, most notably in B cell malignancies (BCMA). CAR-T cells are T lymphocytes that are genetically modified, predominantly by lentiviral or retroviral transduction, to express a CAR that can recognize virtually any surface epitope expressed on a cell. Results of early-phase clinical trials in MM, mainly targeted towards B cell maturation antigen, were promising with high clinical response rates, including complete responses. Unfortunately, responses are usually temporary and relapses have been described due to loss of BCMA expression following CAR-T therapy. In addition, serious adverse events that usually require hospitalization such as cytokine release syndrome are frequently reported. Hence, there is a general consensus that CAR-T cell-based immunotherapy can only become a "viable" therapeutic option in the future if these 3 challenges are adequately addressed: improving efficacy (challenge 1) while reducing toxicity (challenge 2) and costs (challenge 3). The general objective of this project is to develop a next-generation CAR-T cell treatment for multiple myeloma (MM). The hypothesis of this study is that targeting multiple antigens will broaden the anti-tumor immune response and, thus, enhance efficacy of the treatment by reducing the chance of immune escape. Incorporation of immune checkpoint downregulation and enhancement of their co-stimulatory and migratory function can potentially further augment the anti-tumoral properties of the CAR-T cells. Considering potential adverse events, we envisage mRNA electroporation and the use of gamma/delta (γδ) T cells as improvements to the safety and overall costs of CAR-T cell therapy. In summary, we envision a more effective, safer and economically viable CAR-T cell therapy.

Researcher(s)

  • Promoter: Van Tendeloo Vigor

Research team(s)

Project type(s)

  • Research Project

Preclinical development of a next-generation CAR-T cell therapy for multiple myeloma. 02/07/2019 - 01/07/2020

Abstract

Multiple myeloma (MM) is an malignancy that remains incurable to date. Chimeric antigen receptor (CAR)-T cell therapy has obtained impressive clinical results in leukemia and lymphoma, and is gaining momentum in MM as well. This project aims to tackle shortcomings in efficacy and safety in current CAR-T cell therapies by targeting multiple MM antigens, reducing T cell exhaustion and exploring alternative T cell subsets. Importantly, transient T cell modification ensures patient safety, reduces manufacturing costs significantly and can serve as an early-phase testing platform for clinical translation of novel CAR-T cell therapies.

Researcher(s)

  • Promoter: Van Tendeloo Vigor

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

Safety and feasibility evaluation of first-in-human interleukin-15-transpresenting Wilms'tumor (WT1) targeting autologous dendritic cell vaccination in cancer patients. 01/01/2019 - 31/12/2022

Abstract

Improvement of frontline treatment for cancer patients with a high tumor recurrence rate and low effective treatment options is warranted. Dendritic cell (DC) vaccination is in this context a promising immunotherapeutic armament. Complementary to current treatments, DCs as quintessential antigen-presenting cells of the immune system, can activate the antitumor immune system to attack cancer cells. We previously established novel monocyte-derived DC generation protocols integrating the pleiotropic immune regulator interleukin (IL)-15 while downmodulating ligands for the inhibitory checkpoint programmed death (PD)-1. Our preclinical data demonstrate high therapeutic potential of these designer DCs, evidenced by superior immunogenic capacities. Further extending our DC immunotherapy program, this project is designed to enable the first-in-human clinical application of our novel designer DC vaccines, allowing development of clinical-grade production processes, human in vivo safety and feasibility testing and design of next-level combinatorial therapy approaches for cancer patients with a high unmet medical need.

Researcher(s)

Research team(s)

Project type(s)

  • Research Project

Development of anti-leukemia killer T-lymphocytes genetically modified to express T-cell receptors against the Wilms' tumor 1 antigen. 01/01/2019 - 31/12/2021

Abstract

Acute myeloid leukemia (AML) is an agressive type of blood cancer that still carries a dreadful prognosis. Even with treatment, only one out of 4 patients with AML will be alive 5 years after the diagnosis. This explains the urgent need for novel therapies. It is well known that our own immune system can fight cancer, laying the foundation for the development of immune-based therapies for cancer. One type of immunotherapy that has attracted much recent interest is T-cell therapy. Such therapy is based on the intrinsic ability of T cells - an important part of our immune system - to recognize and kill cancer cells. They do so via T-cell receptors, which are expressed on their cell surface. These T-cell receptors recognize certain substances, called antigens, that are presented by the cancer cells. In the context of AML, one antigen that serves as an attractive T-cell target is the Wilms' tumor protein 1 (WT1). In this research project, we will try to "weaponise" T cells to attack leukemia cells by genetically enforcing the expression of T-cell receptors against WT1. The ultimate goal is to exploit these anti-leukemia "killer" T cells for therapeutic purposes and to fulfill the unmet therapeutic need in AML.

Researcher(s)

Research team(s)

Project type(s)

  • Research Project

A human in vitro demyelination model for studying the effect of M2 macrophages/microglia. 01/01/2019 - 31/12/2020

Abstract

The host's laboratory's promising results of IL13 treatment in a mouse model for MS, encourage us to further investigate this potential therapeutic strategy in a human setup. Therefore, we propose to develop a human in vitro demyelination model to study the effect of IL13-induced M2 macrophages and microglia. We will do so by (I) differentiating hiPSC towards oligodendrocytes; (II) evaluating the myelinating potential by oligodendrocyte-neuron co-culture; (III) optimizing a toxin-induced demyelination procedure; and (IV) evaluating the effect of macrophage/microglia addition with or without IL13 stimulation.

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

The study of classical and alternative activation in human induced pluripotent stem cell-derived microglia and macrophages. 01/04/2018 - 31/03/2019

Abstract

Neuroinflammation occurs in all central nervous system (CNS) pathologies and can be defined as the activation of local and peripheral infiltrating immune cells, with the key players being brain-resident microglia and blood-borne infiltrating macrophages. While growing evidence ascribes different roles to microglia and macrophages in neuro-inflammation, the main interest in both phagocytes, with regard to therapeutic strategies, is their ability to obtain different activation states, ranging from pro-inflammatory (M1) to anti-inflammatory (M2) activation. These new revelations led to many studies nowadays, which are investigating immune modulation as a potential therapeutic strategy to treat CNS pathologies. However, since existing pre-clinical models for the study of neuro-inflammation are based on either human cell lines or rodent models, this new and potential therapeutic strategy creates the need for more reliable pre-clinical models for human neuro-immune research. Therefore, with this project, we aim to develop an in vitro assay to study and modulate activation states in human neuro-inflammation by using human induced pluripotent stem cell (hiPSC)-derived microglia and macrophages. For this, we will introduce and validate in vitro differentiation protocols for hiPSC-derived microglia and macrophages. Phenotypical characterization will be performed by using known markers for immunocytochemistry and flow cytometry. Next, functional analyses of the developed hiPSC-derived microglia and macrophages will include (i) migration assays for chemokines CX3CL1 and CCL2, known to attract, respectively, microglia and macrophages; (ii) phagocytosis assays; and (iii) M1-M2 priming experiments, determining the polarising capacity of both microglia and macrophages by flow cytometry and ELISA. With this research project, our main aim is to meet the urgent need for novel in vitro human neuro-inflammation models, but with a successful outcome, we will also achieve a major step forward towards less animal testing.

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

Development of next-generation 3D brain organoids for the study and modulation of immunemediated neurodegeneration in cerebrovascular disease. 01/01/2018 - 31/12/2022

Abstract

Developing novel neuroprotective and/or immune-modulating therapeutic strategies for almost every neurological disease or trauma requires, both for academia and pharmaceutical industry, the existence of robust in vitro cell culture models to mimic disease-associated pathological events. Unfortunately, a complex interplay between multiple central nervous system (CNS) cell types and multiple cell types from the body's peripheral immune system, cannot be easily recapitulated by currently used 2-dimensional (2D) co-culture assays. It is exactly therefore that successful pre-clinical experimental efficacy has proved to be very difficult to translate into clinical benefit, and as a consequence there is an increasing gap in knowledge and progress between bench and bed side. One highly promising novel approach to improve the predictive power of in vitro human neuro-immune research consists in developing modular 3D brain organoids that resemble brain tissue at the structural, cellular and functional level. Within this project we aim to develop and optimize a new method for generating isogenic 3D brain organoids, comprising human pluripotent stem cell (hPSC)-derived neurons, astrocytes and microglia. Furthermore, hPSC-derived astrocytes and endothelial cells will be used to create a blood-brain-barrier model for physical separation of hPSC-derived macrophages from the generated human 3D brain organoids. Together, this integrated cell system will represent a powerful new 3D human neuro-immune cell culture paradigm. Within this multidisciplinary IOF-SBO project, the methodological approach to generate 3D brain organoids, combined with the experience in the field of clinical research and the availability of patient samples, is truly unique and will - in first instance - highly contribute to the field of in vitro cerebrovascular disease modelling and treatment validation. Furthermore, our aims to install an integrated 3D brain organoid technology platform at the University of Antwerp, will - given the current scientific and economic interests – allow for both short-term and long-term valorisation of our combined efforts, with both intellectual (PhD-theses, A1 publications) as well as financial (contract research) revenues.

Researcher(s)

Research team(s)

Project type(s)

  • Research Project

Improved RNA-based engineering of T lymphocytes with leukemia-specific T cell receptors to redirect their effector functions: towards a clinically safe platform to evaluate efficacy and potential off-target toxicity. 01/01/2018 - 31/12/2021

Abstract

The extraordinary specificity of T lymphocytes for their antigen turns them into highly attractive and targeted immunotherapeutics. However, the scarcity of tumor-reactive T cells in cancer patients and the difficulty of their expansion in sufficient numbers for adoptive immunotherapy are substantial hurdles to broaden their clinical application. Transient introduction of a T cell receptor (TCR) specific for a pre-defined tumor-associated antigen by means of RNA-engineering into unselected bulk T cells would instantaneously confer redirected anti-tumor specificity to a large number of effector T cells for adoptive immune therapy with a built-in safety switch. This research project aims to investigate the generation, in vitro validation and preclinical testing of a set of Wilms' tumor 1 (WT1)-specific TCRs derived from leukemia patients that responded successfully to a therapeutic WT1 vaccine. On the short term, we are confident that this research project will provide a sound basis for exploratory and translational phase I trials using WT1-specific TCR mRNA-engineered T cells to study the safety (on- & off-target off-tumor effects) and feasibility of adoptive T cell therapy in patients with WT1-positive hematological malignancies. On the long term, adoptive T cell therapy using redirected T cells is poised to become a new treatment paradigm for both hematological and solid cancer patients at risk of relapse, if needed in combination with other antitumor therapies.

Researcher(s)

Research team(s)

Project type(s)

  • Research Project

Human and murine macrophages with a stable alternative activation as a therapeutic tool to promote neuroregeneration. 01/01/2018 - 31/12/2021

Abstract

Worldwide, spinal cord injuries (SCI) are a major cause of morbidity, mortality and reduced quality of life. Until now no regenerative therapy for SCI is available and standard care of SCI patients includes primarily the administration of immunosuppressant drugs and rehabilitation. Thus, new therapeutic strategies are desperately needed. After SCI, pro-inflammatory macrophages dominate the spinal cord and exert detrimental effects by secreting multiple pro-inflammatory factors, by stimulating the formation of the inhibitory fibrotic scar, and by attacking dystrophic axons. Previously, the applicants demonstrated that the implantation of IL-13-expressing mesenchymal stem cells or macrophages induces 'antiinflammatory' arginase-1 (Arg-1)-positive macrophages/microglia in the injured spinal cord, leading to improved functional recovery. Therefore, we investigate here whether high Arg-1 production by macrophages with a stable alternative activation leads to the suppression of pro-inflammatory macrophages/microglia after SCI via arginine depletion. In addition, for translation to the human situation, we will investigate whether Arg1-overexpressing human macrophages exert neuroprotective effects in an in vitro multicellular co-culture model of human stem cell-derived corticospinal-like motor neurons, astrocytes and microglia. Thus, in this project, we aim to analyse one key mechanism of how anti-inflammatory macrophages improve functional recovery after SCI.

Researcher(s)

Research team(s)

Project type(s)

  • Research Project

Application of human 3D brain organoids to evaluate the potency of interleukin 13 for modulation of detrimental microglia and macrophage immune response. 06/12/2017 - 31/12/2019

Abstract

Due to the current understanding that multiple sclerosis (MS) is a chronic autoimmune disease of the central nervous system (CNS), where mononuclear cell infiltration in brain and spinal cord is a major contributor to demyelination, gliosis, axonal loss and eventually loss of neuronal function, we investigated over the past 6 years whether local modulation of CNS lesions with the immune-modulating cytokine interleukin (IL)13 might ameliorate detrimental disease progression. While our pre-clinical studies in the cuprizone (CPZ)-induced CNS inflammation/demyelination mouse model for human MS have demonstrated proof-of-principle for this approach, currently we do not know whether human microglia and macrophages are equally well susceptible to IL13-mediated immunomodulation in the pro-inflammatory MS environment. Using advanced human induced pluripotent stem cell (hiPSC) derived 3D cell culture models, we aim to provide further pre-clinical rationale for the use of IL13 as an additional treatment approach in advanced stage MS.

Researcher(s)

Research team(s)

Project type(s)

  • Research Project

Development of isogenic human embryonic stem cell-derived 3D neuro-immune cell culture models: pre-clinical evaluation of interleukin 13 for microglia and macrophage immunomodulation under stroke-like pathology. 01/10/2017 - 30/09/2021

Abstract

Development of three-dimensional (3D) in vitro cell culture models for human neuro-immunological research is currently a hot topic in medical cell biology research. Although multiple protocols have been described for generating human 3D brain organoids starting from pluripotent stem cells, current models display several limitations, including the lack of extracellular matrix (ECM), the absence of multiple types of immune cells and a functional blood-brain-barrier (BBB). With this project we aim to develop and optimize a new method for generating 3D neuro-immune cell culture models to study and modulate human neuro-inflammatory responses. For this, isogenic 3D cell cultures comprising human embryonic stem cell (hESC)-derived neurons, astrocytes and microglia will be established on decellularized mouse brain sections in order to provide growth and organizational support by original brain ECM proteins. In addition, hESC-derived astrocytes and endothelial cells will be used to create a BBB model for physical separation of hESC-derived macrophages. Further inclusion of genetic engineering strategies, to allow for real time bioluminescence imaging and (live cell) confocal microscopy, will be applied to ensure profound validation and high throughput screening applications. Once established, we will use this technology to further extend our research efforts to optimize therapeutic strategies based on interleukin (IL)13-mediated immunomodulation, following hypoxic and hypoglycemic stress (i.e. stroke-like conditions). Once validated, we believe that implementation of the proposed 3D brain organoid technology by academia and/or pharmaceutical industry will not only have great impact on the reliability of pre-clinical drug screening, and consequently on the medical and social investments associated with patient care, but also will find application in advanced human toxicology research.

Researcher(s)

Research team(s)

Project website

Project type(s)

  • Research Project

Development of isogenic human embryonic stem cell-derived 3D neuro-immune cell culture models: pre-clinical evaluation of microglia and macrophage immunomodulation for stroke treatment. 01/10/2017 - 30/09/2020

Abstract

Development of three-dimensional (3D) in vitro cell culture models for human neuro-immunological research is currently a hot topic in medical cell biology research. Although multiple protocols have been described for generating human 3D brain organoids starting from pluripotent stem cells, current models display several limitations, including the lack of extracellular matrix (ECM), the absence of multiple types of immune cells and a functional blood-brain-barrier (BBB). With this project we aim to develop and optimize a new method for generating 3D neuro-immune cell culture models to study and modulate human neuro-inflammatory responses. For this, isogenic 3D cell cultures comprising human embryonic stem cell (hESC)-derived neurons, astrocytes and microglia will be established on decellularized mouse brain sections in order to provide growth and organizational support by original brain ECM proteins. In addition, hESC-derived astrocytes and endothelial cells will be used to create a BBB model for physical separation of hESC-derived macrophages. Further inclusion of genetic engineering strategies, to allow for real time bioluminescence imaging and (live cell) confocal microscopy, will be applied to ensure profound validation and high throughput screening applications. Once established, we will use this technology to further extend our research efforts to optimize therapeutic strategies based on interleukin (IL)13-mediated immunomodulation for cerebrovascular disease.

Researcher(s)

Research team(s)

Project type(s)

  • Research Project

Harnessing the expression of interleukin-15 and CD56 in immunotherapeutic strategies combating leukemia: a functional and mechanistic characterization 01/10/2017 - 12/05/2019

Abstract

The "Breakthrough of the Year" of 2013, awarded by Science, was the burgeoning field of cancer immunotherapy. In spite of some great results, the search continues towards an ideal functioning of our immune cells, leaving a paramount question unanswered: can we identify specific cellular attributes denoting optimal activation and immune performance in combating cancer? In this project we will investigate the interleukin (IL)-15-mediated activation of immune cells, more specifically dendritic cells (DCs) and gd T cells, and their expression of CD56 in a model of acute myeloid leukemia. First, IL-2 and IL-15 will be compared for their immunostimulatory effect on gd T cells, conceivably strengthening the use of IL-15 in, among other, adoptive immunotherapy protocols. Induction of activation and enhancement of effector functions of gd T cells by these cytokines will be correlated with their CD56 expression. Next, we will delineate the cross-talk between our CD56+ and IL-15 expressing IL-15 DCs and gd T cells. Furthermore, the role of CD56 on these immune cells will be unraveled. This will give us the opportunity to finally answer the question whether CD56 expression is being indicative of an activated state, whether it actively leads to tumor cell killing and whether or not homodimeric interactions do play a role. Finally, mechanistic insight will be gained into the individual contribution of the IL-15 signaling pathways in induction of CD56 expression and immune cell activation.

Researcher(s)

  • Promoter: Van Tendeloo Vigor
  • Co-promoter: Smits Evelien
  • Fellow: Van Acker Heleen

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

Multicenter randomized phase II trial of Wilms' tumor (WT1) antigen-targeted dendritic cell vaccination to prevent relapse in patients with acute myeloid leukemia. 01/01/2017 - 31/12/2020

Abstract

The five-year survival rate for acute myeloid leukemia (AML) is 26.6%, pointing to the need for new treatment options. We have recently provided the first clinical proof-of-concept evidence that vaccination with Wilms’ tumor 1 mRNA-electroporated dendritic cells (DC) can result in complete clearance of minimal residual disease. Our phase I/II study showed improved survival compared to historical data and demonstrable antileukemic effects. Now we want to confirm the results of our initial study in a multicenter randomized phase II clinical study in 138 patients with AML at high risk of relapse. The primary aim is to determine whether DC vaccination can significantly prevent relapse and increase survival. In addition, tumor marker levels and immune activation will be monitored. If the curative potential with low toxicity can be confirmed in this novel large randomized trial, our cell therapy can become a new standard postremission treatment for AML.

Researcher(s)

Research team(s)

Project type(s)

  • Research Project

Maximizing the anti-tumor potency of next-generation dendritic cell vaccines by combining interleukin-15 and in situ silencing of programmed-death ligands. 01/01/2017 - 31/12/2020

Abstract

Currently therapeutic cancer vaccines have taken center stage in cancer immunotherapy. Such cancer vaccines are designed to delay or prevent cancer relapse after standard treatment with chemotherapy or radiotherapy, and to attack distant metastatic cancer cells. We have already successfully tested a personalized cell-based cancer vaccine for leukemia patients and we demonstrated that the vaccine could prevent leukemia relapse in about 35% of the vaccinated leukemia patients. This cancer vaccine consists of specially cultured immune cells of the patient that upon injection in the skin starts off an anti-tumor immune response against residual or chemotherapy-resistant leukemia cells. In this project we aim to make this personalized leukemia vaccine even more powerful in the test tube by innovative manipulations and by implementing new emerging anti-cancer strategies that have already proven successful in solid tumors.

Researcher(s)

  • Promoter: Van Tendeloo Vigor
  • Co-promoter: Lion Eva
  • Fellow: Versteven Maarten

Research team(s)

Project type(s)

  • Research Project

Improving frontline treatment for malignant pleural mesothelioma with concomitant platinum/pemetrexed-based chemotherapy and Wilms' tumor protein 1 (WT1)-targeted dendritic cell immunotherapy. 01/01/2017 - 31/12/2020

Abstract

The prognosis of patients diagnosed with malignant pleural mesothelioma (MPM) remains dismal with a median overall survival from diagnosis of only 12 months. The steadily increasing incidence of MPM along with the limited efficacy of the currently available treatment options for MPM prompts a search for new, more effective therapeutic modalities and strategies. Dendritic cells · (DCs), the immune system’s quinte-ssential antigen-presenting cells, are a promising armament for immunotherapy of MPM. In this phase 1/11 clinical study designed to improve most common care of MPM, DCs loaded with the mesothelioma-associated tumor antigen Wilms’ tumor 1 protein (WTl) will be used in conjunction with conventiona,l platinum/pemetrexed-based chemotherapy for the frontline treatment of newly diagnosed resectable and non-resectable MPM. Primary objective is to provide the first-in-human experimental demonstration that combining chemotherapy with WTl-targeted DC therapy is feasible and safe a·nd enables induction of systemic and in situ mesotheliomaspecific immune responses in MPM patients.

Researcher(s)

Research team(s)

Project type(s)

  • Research Project

Generation of iPSC-derived 3D neuro-immune cell culture models for study and modulation of neuro-inflammatory processes. 01/01/2017 - 31/12/2020

Abstract

Development of three-dimensional (3D) in vitro cell culture models for neuroscience research is currently a hot topic in medical cell biology research. Although multiple protocols have been described for generating 3D brain organoids starting from pluripotent stem cells, current models display several limitations, including the lack of extracellular matrix (ECM) and the absence of glial and immune cells. With this project we aim to develop and optimize a new method for generating 3D neuro-immune cell culture models to study and modulate neuro-inflammatory responses. For this, 3D cell cultures comprising induced pluripotent stem cell (iPSC)-derived neurons, astrocytes and microglia will be established on decellularized mouse brain sections in order to provide growth and organizational support by the original brain ECM. Further inclusion of genetic engineering strategies, to allow for real time bioluminescence imaging and live cell confocal microscopy, will be applied to ensure high throughput screening applications. Once established, we will use this technology to further extend our research efforts to optimize therapeutic strategies based on interleukin (IL)13-mediated immunomodulation, following hypoxic and hypoglycemic stress. Once validated, we believe that implementation of this technology by pharmaceutical industry will have great impact on the reliability of pre-clinical drug screening, and consequently on the medical and social investments associated with patient care.

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

ASCID: Antwerp Study Centre for Infectious Diseases. 01/01/2015 - 31/12/2019

Abstract

This project represents a research contract awarded by the University of Antwerp. The supervisor provides the Antwerp University research mentioned in the title of the project under the conditions stipulated by the university.

Researcher(s)

Research team(s)

Project type(s)

  • Research Project

CSL Behring Chair in von Willebrand disease. 01/10/2010 - 31/05/2023

Abstract

The purpose of the Chair is a full characferization of Von Willebrand Disease ( VWD) in Belgium, including all up-fo-date laboratory techniques, multimeric analysis and molecular investigation, and the setting up of a plasma and DNA bank for future research in VWD.

Researcher(s)

Research team(s)

Project type(s)

  • Research Project

Multiple Sclerosis, a multidisciplinary approach. 01/01/2010 - 31/12/2019

Abstract

This is a fundamental research project financed by the Research Foundation - Flanders (FWO). The project was subsidized after selection by the FWO-expert panel.

Researcher(s)

Research team(s)

Project type(s)

  • Research Project