Project acronym 5D Heart Patch
Project A Functional, Mature In vivo Human Ventricular Muscle Patch for Cardiomyopathy
Researcher (PI) Kenneth Randall Chien
Host Institution (HI) KAROLINSKA INSTITUTET
Call Details Advanced Grant (AdG), LS7, ERC-2016-ADG
Summary Developing new therapeutic strategies for heart regeneration is a major goal for cardiac biology and medicine. While cardiomyocytes can be generated from human pluripotent stem (hPSC) cells in vitro, it has proven difficult to use these cells to generate a large scale, mature human heart ventricular muscle graft on the injured heart in vivo. The central objective of this proposal is to optimize the generation of a large-scale pure, fully functional human ventricular muscle patch in vivo through the self-assembly of purified human ventricular progenitors and the localized expression of defined paracrine factors that drive their expansion, differentiation, vascularization, matrix formation, and maturation. Recently, we have found that purified hPSC-derived ventricular progenitors (HVPs) can self-assemble in vivo on the epicardial surface into a 3D vascularized, and functional ventricular patch with its own extracellular matrix via a cell autonomous pathway. A two-step protocol and FACS purification of HVP receptors can generate billions of pure HVPs- The current proposal will lead to the identification of defined paracrine pathways to enhance the survival, grafting/implantation, expansion, differentiation, matrix formation, vascularization and maturation of the graft in vivo. We will captalize on our unique HVP system and our novel modRNA technology to deliver therapeutic strategies by using the in vivo human ventricular muscle to model in vivo arrhythmogenic cardiomyopathy, and optimize the ability of the graft to compensate for the massive loss of functional muscle during ischemic cardiomyopathy and post-myocardial infarction. The studies will lead to new in vivo chimeric models of human cardiac disease and an experimental paradigm to optimize organ-on-organ cardiac tissue engineers of an in vivo, functional mature ventricular patch for cardiomyopathy
Summary
Developing new therapeutic strategies for heart regeneration is a major goal for cardiac biology and medicine. While cardiomyocytes can be generated from human pluripotent stem (hPSC) cells in vitro, it has proven difficult to use these cells to generate a large scale, mature human heart ventricular muscle graft on the injured heart in vivo. The central objective of this proposal is to optimize the generation of a large-scale pure, fully functional human ventricular muscle patch in vivo through the self-assembly of purified human ventricular progenitors and the localized expression of defined paracrine factors that drive their expansion, differentiation, vascularization, matrix formation, and maturation. Recently, we have found that purified hPSC-derived ventricular progenitors (HVPs) can self-assemble in vivo on the epicardial surface into a 3D vascularized, and functional ventricular patch with its own extracellular matrix via a cell autonomous pathway. A two-step protocol and FACS purification of HVP receptors can generate billions of pure HVPs- The current proposal will lead to the identification of defined paracrine pathways to enhance the survival, grafting/implantation, expansion, differentiation, matrix formation, vascularization and maturation of the graft in vivo. We will captalize on our unique HVP system and our novel modRNA technology to deliver therapeutic strategies by using the in vivo human ventricular muscle to model in vivo arrhythmogenic cardiomyopathy, and optimize the ability of the graft to compensate for the massive loss of functional muscle during ischemic cardiomyopathy and post-myocardial infarction. The studies will lead to new in vivo chimeric models of human cardiac disease and an experimental paradigm to optimize organ-on-organ cardiac tissue engineers of an in vivo, functional mature ventricular patch for cardiomyopathy
Max ERC Funding
2 149 228 €
Duration
Start date: 2017-12-01, End date: 2022-11-30
Project acronym AUTOCOMPLEMENT
Project The role of complement in the induction of autoimmunity against post-translationally modified proteins
Researcher (PI) Leendert TROUW
Host Institution (HI) ACADEMISCH ZIEKENHUIS LEIDEN
Call Details Consolidator Grant (CoG), LS7, ERC-2016-COG
Summary In many prevalent autoimmune diseases such as rheumatoid arthritis (RA) and systemic lupus erythematosus (SLE) autoantibodies are used as diagnostic and prognostic tools. Several of these autoantibodies target proteins that have been post-translationally modified (PTM). Examples of such modifications are citrullination and carbamylation. The success of B cell-targeted therapies in many auto-antibody positive diseases suggests that B cell mediated auto-immunity is playing a direct pathogenic role. Despite the wealth of information on the clinical associations of these anti-PTM protein antibodies as biomarkers we have currently no insight into why these antibodies are formed.
Immunization studies reveal that PTM proteins can induce antibody responses even in the absence of exogenous adjuvant. The reason why these PTM proteins have ‘autoadjuvant’ properties that lead to a breach of tolerance is currently unknown. In this proposal, I hypothesise that the breach of tolerance towards PTM proteins is mediated by complement factors that bind directly to these PTM. Our preliminary data indeed reveal that several complement factors bind specifically to PTM proteins. Complement could be involved in the autoadjuvant property of PTM proteins as next to killing pathogens complement can also boost adaptive immune responses. I plan to unravel the importance of the complement–PTM protein interaction by answering these questions:
1) What is the physiological function of complement binding to PTM proteins?
2) Is the breach of tolerance towards PTM proteins influenced by complement?
3) Can the adjuvant function of PTM be used to increase vaccine efficacy and/or decrease autoreactivity?
With AUTOCOMPLEMENT I will elucidate how PTM-reactive B cells receive ‘autoadjuvant’ signals. This insight will impact on patient care as we can now design strategies to either block unwanted ‘autoadjuvant’ signals to inhibit autoimmunity or to utilize ‘autoadjuvant’ signals to potentiate vaccination.
Summary
In many prevalent autoimmune diseases such as rheumatoid arthritis (RA) and systemic lupus erythematosus (SLE) autoantibodies are used as diagnostic and prognostic tools. Several of these autoantibodies target proteins that have been post-translationally modified (PTM). Examples of such modifications are citrullination and carbamylation. The success of B cell-targeted therapies in many auto-antibody positive diseases suggests that B cell mediated auto-immunity is playing a direct pathogenic role. Despite the wealth of information on the clinical associations of these anti-PTM protein antibodies as biomarkers we have currently no insight into why these antibodies are formed.
Immunization studies reveal that PTM proteins can induce antibody responses even in the absence of exogenous adjuvant. The reason why these PTM proteins have ‘autoadjuvant’ properties that lead to a breach of tolerance is currently unknown. In this proposal, I hypothesise that the breach of tolerance towards PTM proteins is mediated by complement factors that bind directly to these PTM. Our preliminary data indeed reveal that several complement factors bind specifically to PTM proteins. Complement could be involved in the autoadjuvant property of PTM proteins as next to killing pathogens complement can also boost adaptive immune responses. I plan to unravel the importance of the complement–PTM protein interaction by answering these questions:
1) What is the physiological function of complement binding to PTM proteins?
2) Is the breach of tolerance towards PTM proteins influenced by complement?
3) Can the adjuvant function of PTM be used to increase vaccine efficacy and/or decrease autoreactivity?
With AUTOCOMPLEMENT I will elucidate how PTM-reactive B cells receive ‘autoadjuvant’ signals. This insight will impact on patient care as we can now design strategies to either block unwanted ‘autoadjuvant’ signals to inhibit autoimmunity or to utilize ‘autoadjuvant’ signals to potentiate vaccination.
Max ERC Funding
1 999 803 €
Duration
Start date: 2017-09-01, End date: 2022-08-31
Project acronym BEAT
Project The functional interaction of EGFR and beta-catenin signalling in colorectal cancer: Genetics, mechanisms, and therapeutic potential.
Researcher (PI) Andrea BERTOTTI
Host Institution (HI) UNIVERSITA DEGLI STUDI DI TORINO
Call Details Consolidator Grant (CoG), LS7, ERC-2016-COG
Summary Monoclonal antibodies against the EGF receptor (EGFR) provide substantive benefit to colorectal cancer (CRC) patients. However, no genetic lesions that robustly predict ‘addiction’ to the EGFR pathway have been yet identified. Further, even in tumours that regress after EGFR blockade, subsets of drug-tolerant cells often linger and foster ‘minimal residual disease’ (MRD), which portends tumour relapse.
Our preliminary evidence suggests that reliance on EGFR activity, as opposed to MRD persistence, could be assisted by genetically-based variations in transcription factor partnerships and activities, gene expression outputs, and biological fates controlled by the WNT/beta-catenin pathway. On such premises, BEAT (Beta-catenin and EGFR Abrogation Therapy) will elucidate the mechanisms of EGFR dependency, and escape from it, with the goal to identify biomarkers for more efficient clinical management of CRC and develop new therapies for MRD eradication.
A multidisciplinary approach will be pursued spanning from integrative gene regulation analyses to functional genomics in vitro, pharmacological experiments in vivo, and clinical investigation, to address whether: (i) specific genetic alterations of the WNT pathway affect anti-EGFR sensitivity; (ii) combined neutralisation of EGFR and WNT signals fuels MRD deterioration; (iii) data from analysis of this synergy can lead to the discovery of clinically meaningful biomarkers with predictive and prognostic significance.
This proposal capitalises on a unique proprietary platform for high-content studies based on a large biobank of viable CRC samples, which ensures strong analytical power and unprecedented biological flexibility. By providing fresh insight into the mechanisms whereby WNT/beta-catenin signalling differentially sustains EGFR dependency or drug tolerance, the project is expected to put forward an innovative reinterpretation of CRC molecular bases and advance the rational application of more effective therapies.
Summary
Monoclonal antibodies against the EGF receptor (EGFR) provide substantive benefit to colorectal cancer (CRC) patients. However, no genetic lesions that robustly predict ‘addiction’ to the EGFR pathway have been yet identified. Further, even in tumours that regress after EGFR blockade, subsets of drug-tolerant cells often linger and foster ‘minimal residual disease’ (MRD), which portends tumour relapse.
Our preliminary evidence suggests that reliance on EGFR activity, as opposed to MRD persistence, could be assisted by genetically-based variations in transcription factor partnerships and activities, gene expression outputs, and biological fates controlled by the WNT/beta-catenin pathway. On such premises, BEAT (Beta-catenin and EGFR Abrogation Therapy) will elucidate the mechanisms of EGFR dependency, and escape from it, with the goal to identify biomarkers for more efficient clinical management of CRC and develop new therapies for MRD eradication.
A multidisciplinary approach will be pursued spanning from integrative gene regulation analyses to functional genomics in vitro, pharmacological experiments in vivo, and clinical investigation, to address whether: (i) specific genetic alterations of the WNT pathway affect anti-EGFR sensitivity; (ii) combined neutralisation of EGFR and WNT signals fuels MRD deterioration; (iii) data from analysis of this synergy can lead to the discovery of clinically meaningful biomarkers with predictive and prognostic significance.
This proposal capitalises on a unique proprietary platform for high-content studies based on a large biobank of viable CRC samples, which ensures strong analytical power and unprecedented biological flexibility. By providing fresh insight into the mechanisms whereby WNT/beta-catenin signalling differentially sustains EGFR dependency or drug tolerance, the project is expected to put forward an innovative reinterpretation of CRC molecular bases and advance the rational application of more effective therapies.
Max ERC Funding
1 793 421 €
Duration
Start date: 2017-10-01, End date: 2022-09-30
Project acronym Bio-ICD
Project Biological auto-detection and termination of heart rhythm disturbances
Researcher (PI) Daniël Antonie PIJNAPPELS
Host Institution (HI) ACADEMISCH ZIEKENHUIS LEIDEN
Call Details Starting Grant (StG), LS7, ERC-2016-STG
Summary Imagine a heart that could no longer suffer from life-threatening rhythm disturbances, and not because of pills or traumatizing electroshocks from an Implantable Cardioverter Defibrillator (ICD) device. Instead, this heart has become able to rapidly detect & terminate these malignant arrhythmias fully on its own, after gene transfer. In order to explore this novel concept of biological auto-detection & termination of arrhythmias, I will investigate how forced expression of particular engineered proteins could i) allow cardiac tissue to become a detector of arrhythmias through rapid sensing of acute physiological changes upon their initiation. And how after detection, ii) this cardiac tissue (now as effector), could terminate the arrhythmia by generating a painless electroshock through these proteins.
To this purpose, I will first explore the requirements for such detection & termination by studying arrhythmia initiation and termination in rat models of atrial & ventricular arrhythmias using optical probes and light-gated ion channels. These insights will guide computer-based screening of proteins to identify those properties allowing effective arrhythmia detection & termination. These data will be used for rational engineering of the proteins with the desired properties, followed by their forced expression in cardiac cells and slices to assess anti-arrhythmic potential & safety. Promising proteins will be expressed in whole hearts to study their anti-arrhythmic effects and mechanisms, after which the most effective ones will be studied in awake rats.
This unexplored concept of self-resetting an acutely disturbed physiological state by establishing a biological detector-effector system may yield unique insight into arrhythmia management. Hence, this could provide distinctively innovative therapeutic rationales in which a diseased organ begets its own remedy, e.g. a Biologically-Integrated Cardiac Defibrillator (Bio-ICD).
Summary
Imagine a heart that could no longer suffer from life-threatening rhythm disturbances, and not because of pills or traumatizing electroshocks from an Implantable Cardioverter Defibrillator (ICD) device. Instead, this heart has become able to rapidly detect & terminate these malignant arrhythmias fully on its own, after gene transfer. In order to explore this novel concept of biological auto-detection & termination of arrhythmias, I will investigate how forced expression of particular engineered proteins could i) allow cardiac tissue to become a detector of arrhythmias through rapid sensing of acute physiological changes upon their initiation. And how after detection, ii) this cardiac tissue (now as effector), could terminate the arrhythmia by generating a painless electroshock through these proteins.
To this purpose, I will first explore the requirements for such detection & termination by studying arrhythmia initiation and termination in rat models of atrial & ventricular arrhythmias using optical probes and light-gated ion channels. These insights will guide computer-based screening of proteins to identify those properties allowing effective arrhythmia detection & termination. These data will be used for rational engineering of the proteins with the desired properties, followed by their forced expression in cardiac cells and slices to assess anti-arrhythmic potential & safety. Promising proteins will be expressed in whole hearts to study their anti-arrhythmic effects and mechanisms, after which the most effective ones will be studied in awake rats.
This unexplored concept of self-resetting an acutely disturbed physiological state by establishing a biological detector-effector system may yield unique insight into arrhythmia management. Hence, this could provide distinctively innovative therapeutic rationales in which a diseased organ begets its own remedy, e.g. a Biologically-Integrated Cardiac Defibrillator (Bio-ICD).
Max ERC Funding
1 485 028 €
Duration
Start date: 2017-03-01, End date: 2022-02-28
Project acronym BrainDrain
Project Translational implications of the discovery of brain-draining lymphatics
Researcher (PI) Kari ALITALO
Host Institution (HI) HELSINGIN YLIOPISTO
Call Details Advanced Grant (AdG), LS7, ERC-2016-ADG
Summary In 2010, 800 billion Euros was spent on brain diseases in Europe and the cost is expected to increase due to the aging population. – Here I propose to exploit our new discovery for research to alleviate this disease burden. In work selected by Nature Medicine among the top 10 ”Notable Advances” and by Science as one of the 10 ”Breakthroughs of the year” 2015, we discovered a meningeal lymphatic vascular system that serves brain homeostasis. We want to reassess current concepts about cerebrovascular dynamics, fluid drainage and cellular trafficking in physiological conditions, in Alzheimer’s disease mouse models and in human postmortem tissues. First, we will study the development and properties of meningeal lymphatics and how they are sustained during aging. We then want to analyse the clearance of macromolecules and protein aggregates in Alzheimer’s disease in mice that lack the newly discovered meningeal lymphatic drainage system. We will study if growth factor-mediated expansion of lymphatic vessels alleviates the parenchymal accumulation of neurotoxic amyloid beta and pathogenesis of Alzheimer’s disease and brain damage after traumatic brain injury. We will further analyse the role of lymphangiogenic growth factors and lymphatic vessels in brain solute clearance, immune cell trafficking and in a mouse model of multiple sclerosis. The meningeal lymphatics could be involved in a number of neurodegenerative and neuroinflammatory diseases of considerable human and socioeconomic burden. Several of our previous concepts have already been translated to clinical development and we aim to develop proof-of-principle therapeutic concepts in this project. I feel that we are just now in a unique position to advance frontline European translational biomedical research in this suddenly emerging field, which has received great attention worldwide.
Summary
In 2010, 800 billion Euros was spent on brain diseases in Europe and the cost is expected to increase due to the aging population. – Here I propose to exploit our new discovery for research to alleviate this disease burden. In work selected by Nature Medicine among the top 10 ”Notable Advances” and by Science as one of the 10 ”Breakthroughs of the year” 2015, we discovered a meningeal lymphatic vascular system that serves brain homeostasis. We want to reassess current concepts about cerebrovascular dynamics, fluid drainage and cellular trafficking in physiological conditions, in Alzheimer’s disease mouse models and in human postmortem tissues. First, we will study the development and properties of meningeal lymphatics and how they are sustained during aging. We then want to analyse the clearance of macromolecules and protein aggregates in Alzheimer’s disease in mice that lack the newly discovered meningeal lymphatic drainage system. We will study if growth factor-mediated expansion of lymphatic vessels alleviates the parenchymal accumulation of neurotoxic amyloid beta and pathogenesis of Alzheimer’s disease and brain damage after traumatic brain injury. We will further analyse the role of lymphangiogenic growth factors and lymphatic vessels in brain solute clearance, immune cell trafficking and in a mouse model of multiple sclerosis. The meningeal lymphatics could be involved in a number of neurodegenerative and neuroinflammatory diseases of considerable human and socioeconomic burden. Several of our previous concepts have already been translated to clinical development and we aim to develop proof-of-principle therapeutic concepts in this project. I feel that we are just now in a unique position to advance frontline European translational biomedical research in this suddenly emerging field, which has received great attention worldwide.
Max ERC Funding
2 420 429 €
Duration
Start date: 2017-08-01, End date: 2022-07-31
Project acronym BRCA-ERC
Project Understanding cancer development in BRCA 1/2 mutation carriers for improved Early detection and Risk Control
Researcher (PI) Martin WIDSCHWENDTER
Host Institution (HI) UNIVERSITY COLLEGE LONDON
Call Details Advanced Grant (AdG), LS7, ERC-2016-ADG
Summary Recent evidence demonstrates that cancer is overtaking cardiovascular disease as the number one cause of mortality in Europe. This is largely due to the lack of preventative measures for common (e.g. breast) or highly fatal (e.g. ovarian) human cancers. Most cancers are multifactorial in origin. The core hypothesis of this research programme is that the extremely high risk of BRCA1/2 germline mutation carriers to develop breast and ovarian cancer is a net consequence of cell-autonomous (direct effect of BRCA mutation in cells at risk) and cell non-autonomous (produced in distant organs and affecting organs at risk) factors which both trigger epigenetic, cancer-initiating effects.
The project’s aims are centered around the principles of systems medicine and built on a large cohort of BRCA mutation carriers and controls who will be offered newly established cancer screening programmes. We will uncover how ‘cell non-autonomous’ factors work, provide detail on the epigenetic changes in at-risk tissues and investigate whether these changes are mechanistically linked to cancer, study whether we can neutralise this process and measure success in the organs at risk, and ideally in easy to access samples such as blood, buccal and cervical cells.
In my Department for Women’s Cancer we have assembled a powerful interdisciplinary team including computational biologists, functionalists, immunologists and clinician scientists linked to leading patient advocacy groups which is extremely well placed to lead this pioneering project to develop the fundamental understanding of cancer development in women with BRCA mutations. To reset the epigenome, re-establishing normal cell identity and consequently reducing cancer risk without the need for surgery and being able to monitor the efficacy using multicellular epigenetic outcome predictors will be a major scientific and medical breakthrough and possibly applicable to other chronic diseases.
Summary
Recent evidence demonstrates that cancer is overtaking cardiovascular disease as the number one cause of mortality in Europe. This is largely due to the lack of preventative measures for common (e.g. breast) or highly fatal (e.g. ovarian) human cancers. Most cancers are multifactorial in origin. The core hypothesis of this research programme is that the extremely high risk of BRCA1/2 germline mutation carriers to develop breast and ovarian cancer is a net consequence of cell-autonomous (direct effect of BRCA mutation in cells at risk) and cell non-autonomous (produced in distant organs and affecting organs at risk) factors which both trigger epigenetic, cancer-initiating effects.
The project’s aims are centered around the principles of systems medicine and built on a large cohort of BRCA mutation carriers and controls who will be offered newly established cancer screening programmes. We will uncover how ‘cell non-autonomous’ factors work, provide detail on the epigenetic changes in at-risk tissues and investigate whether these changes are mechanistically linked to cancer, study whether we can neutralise this process and measure success in the organs at risk, and ideally in easy to access samples such as blood, buccal and cervical cells.
In my Department for Women’s Cancer we have assembled a powerful interdisciplinary team including computational biologists, functionalists, immunologists and clinician scientists linked to leading patient advocacy groups which is extremely well placed to lead this pioneering project to develop the fundamental understanding of cancer development in women with BRCA mutations. To reset the epigenome, re-establishing normal cell identity and consequently reducing cancer risk without the need for surgery and being able to monitor the efficacy using multicellular epigenetic outcome predictors will be a major scientific and medical breakthrough and possibly applicable to other chronic diseases.
Max ERC Funding
2 497 841 €
Duration
Start date: 2017-09-01, End date: 2022-08-31
Project acronym CANCERINNOVATION
Project Using novel methodologies to target and image cancer invasion and therapeutic resistance
Researcher (PI) Margaret Frame
Host Institution (HI) THE UNIVERSITY OF EDINBURGH
Call Details Advanced Grant (AdG), LS7, ERC-2011-ADG_20110310
Summary We aim to develop and apply a suite of new technologies in a novel cancer discovery platform that will link high-definition cancer biology, via state-of-the-art disease imaging and pathway modelling, with development of novel interrogative and therapeutic interventions to test in models of cancer that closely resemble human disease. The work will lead to a new understanding of cancer invasion, how to treat advanced disease in the metastatic niche, how to monitor therapeutic responses and the compensatory mechanisms that cause acquired resistance. Platform development will be based on combined, cross-informing technologies that will enable us to predict optimal ‘maintenance therapies’ for metastatic disease by targeting cancer evolution and spread through combination therapy. A key strand of the platform is the development of quantitative multi-modal imaging in vivo by use of optical window technology to inform detailed understanding of disease and drug mechanisms and predictive capability of pathway biomarkers. Innovative methodologies are urgently needed to address declining approval rates of novel medicines and the unmet clinical needs of treating cancer patients in the advanced disease setting, where tumour spread and survival generally continues unchecked by current therapies. This work will be largely pre-clinical, but will always be mindful of the clinical problem in managing late stage human disease through rationale design of combination therapies with companion diagnostic tests. The cancer survival statistics will be changed if we can curb continuing spread of aggressive, metastatic disease and resistance to therapy by taking smarter combined approaches that make best use of emerging technologies in an innovative way, particularly where they are more predictive of clinical efficacy.
Summary
We aim to develop and apply a suite of new technologies in a novel cancer discovery platform that will link high-definition cancer biology, via state-of-the-art disease imaging and pathway modelling, with development of novel interrogative and therapeutic interventions to test in models of cancer that closely resemble human disease. The work will lead to a new understanding of cancer invasion, how to treat advanced disease in the metastatic niche, how to monitor therapeutic responses and the compensatory mechanisms that cause acquired resistance. Platform development will be based on combined, cross-informing technologies that will enable us to predict optimal ‘maintenance therapies’ for metastatic disease by targeting cancer evolution and spread through combination therapy. A key strand of the platform is the development of quantitative multi-modal imaging in vivo by use of optical window technology to inform detailed understanding of disease and drug mechanisms and predictive capability of pathway biomarkers. Innovative methodologies are urgently needed to address declining approval rates of novel medicines and the unmet clinical needs of treating cancer patients in the advanced disease setting, where tumour spread and survival generally continues unchecked by current therapies. This work will be largely pre-clinical, but will always be mindful of the clinical problem in managing late stage human disease through rationale design of combination therapies with companion diagnostic tests. The cancer survival statistics will be changed if we can curb continuing spread of aggressive, metastatic disease and resistance to therapy by taking smarter combined approaches that make best use of emerging technologies in an innovative way, particularly where they are more predictive of clinical efficacy.
Max ERC Funding
2 499 000 €
Duration
Start date: 2012-08-01, End date: 2017-07-31
Project acronym CARDIOPREVENT
Project INTEGRATION OF GENOMICS AND CARDIOMETABOLIC PLASMA BIOMARKERS FOR IMPROVED PREDICTION AND PRIMARY PREVENTION OF CARDIOVASCULAR DISEASE
Researcher (PI) Olle Sten Melander
Host Institution (HI) LUNDS UNIVERSITET
Call Details Starting Grant (StG), LS7, ERC-2011-StG_20101109
Summary "By taking advantage of great experience in genetic and cardiovascular epidemiology and some of the largest cohorts in the world including 60 000 unique individuals, the applicant aims at (1) improving CVD risk prediction and (2) identifying mechanisms causally related to CVD development in order to provide novel targets for drug discovery and targeted life style interventions for use in primary prevention.
In SUBPROJECT 1 we aim at identifying disease causing alleles of loci implicated in CVD by Genome Wide Association Studies (GWAS) and to identify rare alleles with large impact on human CVD. We thus perform whole exome and targeted sequencing in early CVD cases and healthy controls and evaluate all identified variants by relating them to incident CVD in 60.000 individuals. Further, we will create a score of all validated CVD gene variants and test whether such a score improves clinical risk assessment over and above traditional risk factors.
In SUBPROJECT 2 we test whether the plasma metabolome- a phenotype representing the product of dietary intake and inherent (e.g. genetic) metabolism- differs between incident CVD cases and controls and between individuals with high and low CVD genetic risk. We further test whether a life style intervention differentially alters the plasma metabolome between individuals with high and low CVD genetic risk. Finally, we will elucidate the mechanisms underlying CVD genetic associations by testing whether myocardial expression of such genes are affected by experimental myocardial infarction (MI) and whether heart function, MI size and the plasma metabolome are affected by adenoviral myocardial CVD gene transfer in rats.
In SUBPROJECT 3 we test whether glucose metabolism and CVD risk factors can be ameliorated by suppressing vasopressin (VP) by increased water intake in humans. Finally, we test which of the 3 VP receptors is responsible for adverse glucometabolic VP effects in rats by specific VP receptor pharmacological studies."
Summary
"By taking advantage of great experience in genetic and cardiovascular epidemiology and some of the largest cohorts in the world including 60 000 unique individuals, the applicant aims at (1) improving CVD risk prediction and (2) identifying mechanisms causally related to CVD development in order to provide novel targets for drug discovery and targeted life style interventions for use in primary prevention.
In SUBPROJECT 1 we aim at identifying disease causing alleles of loci implicated in CVD by Genome Wide Association Studies (GWAS) and to identify rare alleles with large impact on human CVD. We thus perform whole exome and targeted sequencing in early CVD cases and healthy controls and evaluate all identified variants by relating them to incident CVD in 60.000 individuals. Further, we will create a score of all validated CVD gene variants and test whether such a score improves clinical risk assessment over and above traditional risk factors.
In SUBPROJECT 2 we test whether the plasma metabolome- a phenotype representing the product of dietary intake and inherent (e.g. genetic) metabolism- differs between incident CVD cases and controls and between individuals with high and low CVD genetic risk. We further test whether a life style intervention differentially alters the plasma metabolome between individuals with high and low CVD genetic risk. Finally, we will elucidate the mechanisms underlying CVD genetic associations by testing whether myocardial expression of such genes are affected by experimental myocardial infarction (MI) and whether heart function, MI size and the plasma metabolome are affected by adenoviral myocardial CVD gene transfer in rats.
In SUBPROJECT 3 we test whether glucose metabolism and CVD risk factors can be ameliorated by suppressing vasopressin (VP) by increased water intake in humans. Finally, we test which of the 3 VP receptors is responsible for adverse glucometabolic VP effects in rats by specific VP receptor pharmacological studies."
Max ERC Funding
1 500 000 €
Duration
Start date: 2011-12-01, End date: 2016-11-30
Project acronym CELLNAIVETY
Project Deciphering the Molecular Foundations and Functional Competence of Alternative Human Naïve Pluripotent Stem Cells
Researcher (PI) Yaqub HANNA
Host Institution (HI) WEIZMANN INSTITUTE OF SCIENCE
Call Details Consolidator Grant (CoG), LS7, ERC-2016-COG
Summary An important goal of stem cell therapy is to create “customized” cells that are genetically identical to the patient, which upon transplantation can restore damaged tissues. Such cells can be obtained by in vitro direct reprogramming of somatic cells into embryonic stem (ES)-like cells, termed induced pluripotent stem cells (iPSC). This approach also opens possibilities for modelling human diseases in vitro. However, major hurdles remain that restrain fulfilling conventional human iPSC/ESC potential, as they reside in an advanced primed pluripotent state. Such hurdles include limited differentiation capacity and functional variability. Further, in vitro iPSC based research platforms are simplistic and iPSC based “humanized” chimeric mouse models may be of great benefit.
The recent isolation of distinct and new “mouse-like” naive pluripotent states in humans that correspond to earlier embryonic developmental state(s), constitutes a paradigm shift and may alleviate limitations of conventional primed iPSCs/ESCs. Thus, our proposal aims at dissecting the human naïve pluripotent state(s) and to unveil pathways that facilitate their unique identity and flexible programming.
Specific goals: 1) Transcriptional and Epigenetic Design Principles of Human Naïve Pluripotency 2) Signalling Principles Governing Human Naïve Pluripotency Maintenance and Differentiation 3) Defining Functional Competence and Safety of Human Naïve Pluripotent Stem Cells in vitro 4) Novel human naïve iPSC based cross-species chimeric mice for studying human differentiation and disease modelling in vivo. These aims will be conducted by utilizing engineered human iPSC/ESC models, CRISPR/Cas9 genome-wide screening, advanced microscopy and ex-vivo whole embryo culture methods. Our goals will synergistically lead to the design of strategies that will accelerate the safe medical application of human naive pluripotent stem cells and their use in disease specific modelling and applied stem cell research.
Summary
An important goal of stem cell therapy is to create “customized” cells that are genetically identical to the patient, which upon transplantation can restore damaged tissues. Such cells can be obtained by in vitro direct reprogramming of somatic cells into embryonic stem (ES)-like cells, termed induced pluripotent stem cells (iPSC). This approach also opens possibilities for modelling human diseases in vitro. However, major hurdles remain that restrain fulfilling conventional human iPSC/ESC potential, as they reside in an advanced primed pluripotent state. Such hurdles include limited differentiation capacity and functional variability. Further, in vitro iPSC based research platforms are simplistic and iPSC based “humanized” chimeric mouse models may be of great benefit.
The recent isolation of distinct and new “mouse-like” naive pluripotent states in humans that correspond to earlier embryonic developmental state(s), constitutes a paradigm shift and may alleviate limitations of conventional primed iPSCs/ESCs. Thus, our proposal aims at dissecting the human naïve pluripotent state(s) and to unveil pathways that facilitate their unique identity and flexible programming.
Specific goals: 1) Transcriptional and Epigenetic Design Principles of Human Naïve Pluripotency 2) Signalling Principles Governing Human Naïve Pluripotency Maintenance and Differentiation 3) Defining Functional Competence and Safety of Human Naïve Pluripotent Stem Cells in vitro 4) Novel human naïve iPSC based cross-species chimeric mice for studying human differentiation and disease modelling in vivo. These aims will be conducted by utilizing engineered human iPSC/ESC models, CRISPR/Cas9 genome-wide screening, advanced microscopy and ex-vivo whole embryo culture methods. Our goals will synergistically lead to the design of strategies that will accelerate the safe medical application of human naive pluripotent stem cells and their use in disease specific modelling and applied stem cell research.
Max ERC Funding
2 000 000 €
Duration
Start date: 2017-11-01, End date: 2022-10-31
Project acronym CellTrack
Project Cellular Position Tracking Using DNA Origami Barcodes
Researcher (PI) Björn HÖGBERG
Host Institution (HI) KAROLINSKA INSTITUTET
Call Details Consolidator Grant (CoG), LS7, ERC-2016-COG
Summary The research I propose here will provide an enabling technology; spatially resolved transcriptomics, to address important problems in cell- and developmental-biology, in particular: How are stem cells in the skin and gut proliferating without turning into cancers? How are differentiated cells related, in their transcriptome and spatial positions, to their progenitors?
To investigate these problems on a molecular level and open up paths to find completely new spatiotemporal interdependencies in complex biological systems, I propose to use our newly developed DNA-origami strategy (Benson et al, Nature, 523 p. 441 (2015) ), combined with a combinatorial cloning technique, to build a new method for deep mRNA sequencing of tissue with single-cell resolution. These new types of origami are stable in physiological salt conditions and opens up their use in in-vivo applications.
In DNA-origami we can control the exact spatial position of all nucleotides. By folding the scaffold to display sequences for hybridization of fluorophores conjugated to DNA, we can create optical nano-barcodes. By using structures made out of DNA, the patterns of the optical barcodes will be readable both by imaging and by sequencing, thus enabling the creation of a mapping between cell locations in an organ and the mRNA expression of those cells.
We will use the method to perform spatially resolved transcriptomics in small organs: the mouse hair follicle, and small intestine crypt, and also perform the procedure for multiple samples collected at different time points. This will enable a high-dimensional data analysis that most likely will expose previously unknown dependencies that would provide completely new knowledge about how these biological systems work. By studying these systems, we will uncover much more information on how stem cells contribute to regeneration, the issue of de-differentiation that is a common theme in these organs and the effect this might have on the origin of cancer.
Summary
The research I propose here will provide an enabling technology; spatially resolved transcriptomics, to address important problems in cell- and developmental-biology, in particular: How are stem cells in the skin and gut proliferating without turning into cancers? How are differentiated cells related, in their transcriptome and spatial positions, to their progenitors?
To investigate these problems on a molecular level and open up paths to find completely new spatiotemporal interdependencies in complex biological systems, I propose to use our newly developed DNA-origami strategy (Benson et al, Nature, 523 p. 441 (2015) ), combined with a combinatorial cloning technique, to build a new method for deep mRNA sequencing of tissue with single-cell resolution. These new types of origami are stable in physiological salt conditions and opens up their use in in-vivo applications.
In DNA-origami we can control the exact spatial position of all nucleotides. By folding the scaffold to display sequences for hybridization of fluorophores conjugated to DNA, we can create optical nano-barcodes. By using structures made out of DNA, the patterns of the optical barcodes will be readable both by imaging and by sequencing, thus enabling the creation of a mapping between cell locations in an organ and the mRNA expression of those cells.
We will use the method to perform spatially resolved transcriptomics in small organs: the mouse hair follicle, and small intestine crypt, and also perform the procedure for multiple samples collected at different time points. This will enable a high-dimensional data analysis that most likely will expose previously unknown dependencies that would provide completely new knowledge about how these biological systems work. By studying these systems, we will uncover much more information on how stem cells contribute to regeneration, the issue of de-differentiation that is a common theme in these organs and the effect this might have on the origin of cancer.
Max ERC Funding
1 923 263 €
Duration
Start date: 2017-08-01, End date: 2022-07-31
Project acronym ChAMPioN
Project Game-changing Precision Medicine for Curing All Myeloproliferative Neoplasms
Researcher (PI) Tessa Holyoake
Host Institution (HI) UNIVERSITY OF GLASGOW
Call Details Advanced Grant (AdG), LS7, ERC-2016-ADG
Summary Despite decades of research, developing ways to overcome drug resistance in cancer is the most challenging bottleneck for curative therapies. This is because, in some forms of cancer, the cancer stem cells from which the diseases arise are constantly evolving, particularly under the selective pressures of drug therapies, in order to survive. The events leading to drug resistance can occur within one or more individual cancer stem cell(s) – and the features of each of these cells need to be studied in detail in order to develop drugs or drug combinations that can eradicate all of them. The BCR-ABL+ and BCR-ABL- myeloproliferative neoplasms (MPN) are a group of proliferative blood diseases that can be considered both exemplars of precision medicine and of the drug resistance bottleneck. While significant advances in the management of MPN have been made using life-long and expensive tyrosine kinase inhibitors (TKI), patients are rarely cured of their disease. This is because TKI fail to eradicate the leukaemia stem cells (LSC) from which MPN arise and which persist in patients on treatment, often leading to pervasive drug resistance, loss of response to therapy and progression to fatal forms of acute leukaemia. My goal is to change the way we study the LSC that persist in MPN patients as a means of delivering more effective precision medicine in MPN that is a “game-changer” leading to therapy-free remission (TFR) and cure. Here, I will apply an innovative strategy, ChAMPioN, to study the response of the MPN LSC to TKI in innovative pre-clinical laboratory models and directly in patients with MPN - up to the resolution of individual LSC. This work will reveal, for the first time, the molecular and clonal evolution of LSC during TKI therapies, thus enabling the development of more accurate predictions of TKI efficacy and resistance and rational approaches for curative drug therapies.
Summary
Despite decades of research, developing ways to overcome drug resistance in cancer is the most challenging bottleneck for curative therapies. This is because, in some forms of cancer, the cancer stem cells from which the diseases arise are constantly evolving, particularly under the selective pressures of drug therapies, in order to survive. The events leading to drug resistance can occur within one or more individual cancer stem cell(s) – and the features of each of these cells need to be studied in detail in order to develop drugs or drug combinations that can eradicate all of them. The BCR-ABL+ and BCR-ABL- myeloproliferative neoplasms (MPN) are a group of proliferative blood diseases that can be considered both exemplars of precision medicine and of the drug resistance bottleneck. While significant advances in the management of MPN have been made using life-long and expensive tyrosine kinase inhibitors (TKI), patients are rarely cured of their disease. This is because TKI fail to eradicate the leukaemia stem cells (LSC) from which MPN arise and which persist in patients on treatment, often leading to pervasive drug resistance, loss of response to therapy and progression to fatal forms of acute leukaemia. My goal is to change the way we study the LSC that persist in MPN patients as a means of delivering more effective precision medicine in MPN that is a “game-changer” leading to therapy-free remission (TFR) and cure. Here, I will apply an innovative strategy, ChAMPioN, to study the response of the MPN LSC to TKI in innovative pre-clinical laboratory models and directly in patients with MPN - up to the resolution of individual LSC. This work will reveal, for the first time, the molecular and clonal evolution of LSC during TKI therapies, thus enabling the development of more accurate predictions of TKI efficacy and resistance and rational approaches for curative drug therapies.
Max ERC Funding
3 005 818 €
Duration
Start date: 2018-01-01, End date: 2022-12-31
Project acronym CHILDGROWTH2CANCER
Project Childhood body size, growth and pubertal timing and the risk of cancer in adulthood
Researcher (PI) Jennifer Lyn Baker
Host Institution (HI) REGION HOVEDSTADEN
Call Details Starting Grant (StG), LS7, ERC-2011-StG_20101109
Summary The goal of the proposed research is to examine how the independent and combined effects of childhood adiposity (assessed by body mass index [BMI]; kg/m2) height, change in BMI and height, and pubertal timing from the ages of 7 to 13 years are associated with the risk of cancer incidence in adulthood. Greater body size (adipose tissue and different types of lean tissue) reflecting past or ongoing growth may increase the risk of cancer in individuals as greater numbers of proliferating cells increase the risk that mutations leading to the subsequent development of cancer occur. As childhood is a period of growth, it is plausible that it is of particular relevance for the early establishment of the risk of cancer.
Data from the Copenhagen School Health Records Register, which is based on a population of schoolchildren born between 1930-1983 and contains computerised weight and height measurements on >350.000 boys and girls in the capital city of Denmark, as well as data from other cohorts will be used. Survival analysis techniques and the newly developed Dynamic Path Analysis model will be used to examine how body size (BMI and height) at each age from 7 to 13 years as well as change in body size during this period is associated with the risk of multiple forms of cancer in adulthood with a simultaneous exploration of the effects of birth weight and pubertal timing. Additionally, potential effects of childhood and adult health and social circumstances will be investigated in sub-cohorts with this information available.
Results from this research will demonstrate if childhood is a critical period for the establishment of the risk for cancer in adulthood and will lead into mechanistic explorations of the associations at the biological level, investigations into associations between childhood body size and mortality and contribute to developing improved definitions of childhood overweight and obesity that are based upon long-term health outcomes.
Summary
The goal of the proposed research is to examine how the independent and combined effects of childhood adiposity (assessed by body mass index [BMI]; kg/m2) height, change in BMI and height, and pubertal timing from the ages of 7 to 13 years are associated with the risk of cancer incidence in adulthood. Greater body size (adipose tissue and different types of lean tissue) reflecting past or ongoing growth may increase the risk of cancer in individuals as greater numbers of proliferating cells increase the risk that mutations leading to the subsequent development of cancer occur. As childhood is a period of growth, it is plausible that it is of particular relevance for the early establishment of the risk of cancer.
Data from the Copenhagen School Health Records Register, which is based on a population of schoolchildren born between 1930-1983 and contains computerised weight and height measurements on >350.000 boys and girls in the capital city of Denmark, as well as data from other cohorts will be used. Survival analysis techniques and the newly developed Dynamic Path Analysis model will be used to examine how body size (BMI and height) at each age from 7 to 13 years as well as change in body size during this period is associated with the risk of multiple forms of cancer in adulthood with a simultaneous exploration of the effects of birth weight and pubertal timing. Additionally, potential effects of childhood and adult health and social circumstances will be investigated in sub-cohorts with this information available.
Results from this research will demonstrate if childhood is a critical period for the establishment of the risk for cancer in adulthood and will lead into mechanistic explorations of the associations at the biological level, investigations into associations between childhood body size and mortality and contribute to developing improved definitions of childhood overweight and obesity that are based upon long-term health outcomes.
Max ERC Funding
1 199 998 €
Duration
Start date: 2012-02-01, End date: 2017-01-31
Project acronym ContraNPM1AML
Project Dissecting to hit the therapeutic targets in nucleophosmin (NPM1)-mutated acute myeloid leukemia
Researcher (PI) Maria Paola MARTELLI
Host Institution (HI) UNIVERSITA DEGLI STUDI DI PERUGIA
Call Details Consolidator Grant (CoG), LS7, ERC-2016-COG
Summary Acute myeloid leukemia (AML) is a group of hematologic malignancies which, due to their molecular and clinical heterogeneity, have been traditionally difficult to classify and treat. Recently, next-generation, whole-genome sequencing has uncovered several recurrent somatic mutations that better define the landscape of AML genomics. Despite these advances in deciphering AML molecular subsets, there have been no concurrent improvements in AML therapy which still relies on the ‘antracycline+cytarabine’ scheme. Hereto, only about 40-50% of adult young patients are cured whilst most of the elderly succumb to their disease. Therefore, new therapeutic approaches which would take advantage of the new discoveries are clearly needed. In the past years, we discovered and characterized nucleophosmin (NPM1) mutations as the most frequent genetic alteration (about 30%) in AML, and today NPM1-mutated AML is a new entity in the WHO classification of myeloid neoplasms. However, mechanisms of leukemogenesis and a specific therapy for this leukemia are missing. Here, I aim to unravel the complex network of molecular interactions that take place in this distinct genetic subtype, and find their vulnerabilities to identify new targets for therapy. To address this issue, I will avail of relevant pre-clinical models developed in our laboratories and propose two complementary strategies: 1) a screening-based approach, focused either on the target, by analyzing synthetic lethal interactions through CRISPR-based genome-wide interference, or on the drug, by high-throughput chemical libraries screenings; 2) a hypothesis-driven approach, based on our recent gained novel insights on the role of specific intracellular pathways/genes in NPM1-mutated AML and on pharmacological studies with ‘old’ drugs, which we have revisited in the specific AML genetic context. I expect our discoveries will lead to find novel therapeutic approaches and make clinical trials available to patients as soon as possible.
Summary
Acute myeloid leukemia (AML) is a group of hematologic malignancies which, due to their molecular and clinical heterogeneity, have been traditionally difficult to classify and treat. Recently, next-generation, whole-genome sequencing has uncovered several recurrent somatic mutations that better define the landscape of AML genomics. Despite these advances in deciphering AML molecular subsets, there have been no concurrent improvements in AML therapy which still relies on the ‘antracycline+cytarabine’ scheme. Hereto, only about 40-50% of adult young patients are cured whilst most of the elderly succumb to their disease. Therefore, new therapeutic approaches which would take advantage of the new discoveries are clearly needed. In the past years, we discovered and characterized nucleophosmin (NPM1) mutations as the most frequent genetic alteration (about 30%) in AML, and today NPM1-mutated AML is a new entity in the WHO classification of myeloid neoplasms. However, mechanisms of leukemogenesis and a specific therapy for this leukemia are missing. Here, I aim to unravel the complex network of molecular interactions that take place in this distinct genetic subtype, and find their vulnerabilities to identify new targets for therapy. To address this issue, I will avail of relevant pre-clinical models developed in our laboratories and propose two complementary strategies: 1) a screening-based approach, focused either on the target, by analyzing synthetic lethal interactions through CRISPR-based genome-wide interference, or on the drug, by high-throughput chemical libraries screenings; 2) a hypothesis-driven approach, based on our recent gained novel insights on the role of specific intracellular pathways/genes in NPM1-mutated AML and on pharmacological studies with ‘old’ drugs, which we have revisited in the specific AML genetic context. I expect our discoveries will lead to find novel therapeutic approaches and make clinical trials available to patients as soon as possible.
Max ERC Funding
1 883 750 €
Duration
Start date: 2017-04-01, End date: 2022-03-31
Project acronym CREST
Project Enrichment of macular pigment, and its impact on vision and blindness
Researcher (PI) John Michael Nolan
Host Institution (HI) WATERFORD INSTITUTE OF TECHNOLOGY
Call Details Starting Grant (StG), LS7, ERC-2011-StG_20101109
Summary Age-related macular degeneration (AMD) is the leading cause of blindness in the developed world. The macula, the central part of the retina, is responsible for optimal spatial vision. There is a growing body of evidence that a lack of a dietary pigment at the macula, known as macular pigment (MP), is associated with increased risk of AMD.
MP contains the carotenoids lutein (L), zeaxanthin (Z) and meso-zeaxanthin (meso-Z). The typical western diet contains around 60 carotenoids, and 18 have been identified in human serum. However, only three are found at the macula, indicating the unique biological selectivity for their uptake at this location. The function of MP remains undetermined. It is likely that the accumulation of MP has evolved because of its optical and antioxidant properties; for example, MP limits retinal oxidative damage passively (through filtration of blue light) and actively (by quenching free radicals). Furthermore, its optical properties suggest a key role for MP in enhancing visual performance and supporting ‘super’ vision by reducing the effects of chromatic aberration and light scatter.
Recent research has shown that MP can be augmented by dietary supplementation in most (but not all) subjects, suggesting that the macular concentrations of these carotenoids are suboptimal in many people. My laboratory has discovered that a dip in the central portion of this pigment, seen in around 12% of individuals, is an undesirable feature of its spatial profile and may be linked to an inability to generate meso-Z at the macula. However, we have identified that enrichment of MP can be achieved by inclusion of meso-Z in a dietary supplement.
We propose to uniquely enrich MP and assess its impact on visual performance in normal subjects and visual function in patients with AMD. This groundbreaking study will advance our understanding of the protective and optical hypothesis of MP, and potentially improve normal vision and prevent or delay blindness due to AMD.
Summary
Age-related macular degeneration (AMD) is the leading cause of blindness in the developed world. The macula, the central part of the retina, is responsible for optimal spatial vision. There is a growing body of evidence that a lack of a dietary pigment at the macula, known as macular pigment (MP), is associated with increased risk of AMD.
MP contains the carotenoids lutein (L), zeaxanthin (Z) and meso-zeaxanthin (meso-Z). The typical western diet contains around 60 carotenoids, and 18 have been identified in human serum. However, only three are found at the macula, indicating the unique biological selectivity for their uptake at this location. The function of MP remains undetermined. It is likely that the accumulation of MP has evolved because of its optical and antioxidant properties; for example, MP limits retinal oxidative damage passively (through filtration of blue light) and actively (by quenching free radicals). Furthermore, its optical properties suggest a key role for MP in enhancing visual performance and supporting ‘super’ vision by reducing the effects of chromatic aberration and light scatter.
Recent research has shown that MP can be augmented by dietary supplementation in most (but not all) subjects, suggesting that the macular concentrations of these carotenoids are suboptimal in many people. My laboratory has discovered that a dip in the central portion of this pigment, seen in around 12% of individuals, is an undesirable feature of its spatial profile and may be linked to an inability to generate meso-Z at the macula. However, we have identified that enrichment of MP can be achieved by inclusion of meso-Z in a dietary supplement.
We propose to uniquely enrich MP and assess its impact on visual performance in normal subjects and visual function in patients with AMD. This groundbreaking study will advance our understanding of the protective and optical hypothesis of MP, and potentially improve normal vision and prevent or delay blindness due to AMD.
Max ERC Funding
1 493 342 €
Duration
Start date: 2011-10-01, End date: 2016-09-30
Project acronym DrugComb
Project Informatics approaches for the rational selection of personalized cancer drug combinations
Researcher (PI) Jing TANG
Host Institution (HI) HELSINGIN YLIOPISTO
Call Details Starting Grant (StG), LS7, ERC-2016-STG
Summary Making cancer treatment more personalized and effective is one of the grand challenges in our health care system. However, many drugs have entered clinical trials but so far showed limited efficacy or induced rapid development of resistance. We critically need multi-targeted drug combinations, which shall selectively inhibit the cancer cells and block the emergence of drug resistance. This project will develop mathematical and computational tools to identify drug combinations that can be used to provide personalized and more effective therapeutic strategies that may prevent acquired resistance. Utilizing molecular profiling and pharmacological screening data from patient-derived leukaemia and ovarian cancer samples, I will develop model-based clustering methods for identification of patient subgroups that are differentially responsive to first-line chemotherapy. For patients resistant to chemotherapy, I will develop network modelling approaches to predict the most potential drug combinations by understanding the underlying drug target interactions. The drug combination prediction will be made for each patient and will be validated using a preclinical drug testing platform on patient samples. I will explore the drug combination screen data to identify significant synergy at the therapeutically relevant doses. The drug combination hits will be mapped into signalling networks to infer their mechanisms. Drug combinations with selective efficacy in individual patient samples or in sample subgroups will be further translated into in treatment options by clinical collaborators. This will lead to novel and personalized strategies to treat cancer patients.
Summary
Making cancer treatment more personalized and effective is one of the grand challenges in our health care system. However, many drugs have entered clinical trials but so far showed limited efficacy or induced rapid development of resistance. We critically need multi-targeted drug combinations, which shall selectively inhibit the cancer cells and block the emergence of drug resistance. This project will develop mathematical and computational tools to identify drug combinations that can be used to provide personalized and more effective therapeutic strategies that may prevent acquired resistance. Utilizing molecular profiling and pharmacological screening data from patient-derived leukaemia and ovarian cancer samples, I will develop model-based clustering methods for identification of patient subgroups that are differentially responsive to first-line chemotherapy. For patients resistant to chemotherapy, I will develop network modelling approaches to predict the most potential drug combinations by understanding the underlying drug target interactions. The drug combination prediction will be made for each patient and will be validated using a preclinical drug testing platform on patient samples. I will explore the drug combination screen data to identify significant synergy at the therapeutically relevant doses. The drug combination hits will be mapped into signalling networks to infer their mechanisms. Drug combinations with selective efficacy in individual patient samples or in sample subgroups will be further translated into in treatment options by clinical collaborators. This will lead to novel and personalized strategies to treat cancer patients.
Max ERC Funding
1 500 000 €
Duration
Start date: 2017-06-01, End date: 2022-05-31
Project acronym ECSTATIC
Project Electrostructural Tomography – Towards Multiparametric Imaging of Cardiac Electrical Disorders
Researcher (PI) Hubert, Yann, Marie COCHET
Host Institution (HI) UNIVERSITE DE BORDEAUX
Call Details Starting Grant (StG), LS7, ERC-2016-STG
Summary Cardiac electrical diseases are directly responsible for sudden cardiac death, heart failure and stroke. They result from a complex interplay between myocardial electrical activation and structural heterogeneity. Current diagnostic strategy based on separate electrocardiographic and imaging assessment is unable to grasp both these aspects. Improvements in personalised diagnostics are urgently needed as existing curative or preventive therapies (catheter ablation, multisite pacing, and implantable defibrillators) cannot be offered until patients are correctly recognised.
My aim is to achieve a major advance in the way cardiac electrical diseases are characterised and thus diagnosed and treated, through the development of a novel non-invasive modality (Electrostructural Tomography), combining magnetic resonance imaging (MRI) and non-invasive cardiac mapping (NIM) technologies.
The approach will consist of: (1) hybridising NIM and MRI technologies to enable the joint acquisition of magnetic resonance images of the heart and torso and of a large array of body surface potentials within a single environment; (2) personalising the inverse problem of electrocardiography based on MRI characteristics within the heart and torso, to enable accurate reconstruction of cardiac electrophysiological maps from body surface potentials within the 3D cardiac tissue; and (3) developing a novel disease characterisation framework based on registered non-invasive imaging and electrophysiological data, and propose novel diagnostic and prognostic markers.
This project will dramatically impact the tailored management of cardiac electrical disorders, with applications for diagnosis, risk stratification/patient selection and guidance of pacing and catheter ablation therapies. It will bridge two medical fields (cardiac electrophysiology and imaging), thereby creating a new research area and a novel semiology with the potential to modify the existing classification of cardiac electrical diseases.
Summary
Cardiac electrical diseases are directly responsible for sudden cardiac death, heart failure and stroke. They result from a complex interplay between myocardial electrical activation and structural heterogeneity. Current diagnostic strategy based on separate electrocardiographic and imaging assessment is unable to grasp both these aspects. Improvements in personalised diagnostics are urgently needed as existing curative or preventive therapies (catheter ablation, multisite pacing, and implantable defibrillators) cannot be offered until patients are correctly recognised.
My aim is to achieve a major advance in the way cardiac electrical diseases are characterised and thus diagnosed and treated, through the development of a novel non-invasive modality (Electrostructural Tomography), combining magnetic resonance imaging (MRI) and non-invasive cardiac mapping (NIM) technologies.
The approach will consist of: (1) hybridising NIM and MRI technologies to enable the joint acquisition of magnetic resonance images of the heart and torso and of a large array of body surface potentials within a single environment; (2) personalising the inverse problem of electrocardiography based on MRI characteristics within the heart and torso, to enable accurate reconstruction of cardiac electrophysiological maps from body surface potentials within the 3D cardiac tissue; and (3) developing a novel disease characterisation framework based on registered non-invasive imaging and electrophysiological data, and propose novel diagnostic and prognostic markers.
This project will dramatically impact the tailored management of cardiac electrical disorders, with applications for diagnosis, risk stratification/patient selection and guidance of pacing and catheter ablation therapies. It will bridge two medical fields (cardiac electrophysiology and imaging), thereby creating a new research area and a novel semiology with the potential to modify the existing classification of cardiac electrical diseases.
Max ERC Funding
1 475 000 €
Duration
Start date: 2017-02-01, End date: 2022-01-31
Project acronym ENGVASC
Project Engineering Vascularized Tissues
Researcher (PI) Shulamit Levenberg
Host Institution (HI) TECHNION - ISRAEL INSTITUTE OF TECHNOLOGY
Call Details Starting Grant (StG), LS7, ERC-2011-StG_20101109
Summary Vascularization, the process in which new blood vessels assemble, is fundamental to tissue vitality. Vessel network assembly within 3D tissues can be induced in-vitro by means of multicellular culturing of endothelial cells (EC), fibroblasts and cells specific to the tissue of interest. This approach supports formation of endothelial vessels and promotes EC and tissue-specific cell interactions. Such EC-dependent tube-like openings may also form the basis for improved media penetration to the inner regions of thick 3D constructs, allowing for enhanced construct survival and for effective engineering of large complex tissues in the lab. Moreover, our own breakthrough results describe the beneficial impact of in vitro prevascularization of engineered muscle tissue on its survival and vascularization upon implantation. These studies have also demonstrated that implanted vascular networks of in vitro engineered constructs, can anastomose with host vasculature and form functional blood vessels in vivo. However, the mechanisms underlying enhanced vascularization of endothelialized engineered constructs and implant-host vessel integration remain unclear. In this proposal, our research objectives are (1) to uncover the mechanisms governing in vitro vessel network formation in engineered 3D tissues and (2) to elucidate the process of graft-host vessel network integration and implant vessel-stimulated promotion of neovascularization in vivo. In addition, the impact of construct prevascularization on implant survival and function will be explored in animal disease models. While there are still many challenges ahead, should we succeed, our research could lay the foundation for significantly enhanced tissue construct vascularization procedures and for their application in regenerative medicine. In addition, it may provide alternative models for studying the vascularization processes in embryogenesis and disease.
Summary
Vascularization, the process in which new blood vessels assemble, is fundamental to tissue vitality. Vessel network assembly within 3D tissues can be induced in-vitro by means of multicellular culturing of endothelial cells (EC), fibroblasts and cells specific to the tissue of interest. This approach supports formation of endothelial vessels and promotes EC and tissue-specific cell interactions. Such EC-dependent tube-like openings may also form the basis for improved media penetration to the inner regions of thick 3D constructs, allowing for enhanced construct survival and for effective engineering of large complex tissues in the lab. Moreover, our own breakthrough results describe the beneficial impact of in vitro prevascularization of engineered muscle tissue on its survival and vascularization upon implantation. These studies have also demonstrated that implanted vascular networks of in vitro engineered constructs, can anastomose with host vasculature and form functional blood vessels in vivo. However, the mechanisms underlying enhanced vascularization of endothelialized engineered constructs and implant-host vessel integration remain unclear. In this proposal, our research objectives are (1) to uncover the mechanisms governing in vitro vessel network formation in engineered 3D tissues and (2) to elucidate the process of graft-host vessel network integration and implant vessel-stimulated promotion of neovascularization in vivo. In addition, the impact of construct prevascularization on implant survival and function will be explored in animal disease models. While there are still many challenges ahead, should we succeed, our research could lay the foundation for significantly enhanced tissue construct vascularization procedures and for their application in regenerative medicine. In addition, it may provide alternative models for studying the vascularization processes in embryogenesis and disease.
Max ERC Funding
1 500 000 €
Duration
Start date: 2012-10-01, End date: 2017-09-30
Project acronym EPI-Centrd
Project Epilepsy Controlled with Electronic Neurotransmitter Delivery
Researcher (PI) Adam WILLIAMSON
Host Institution (HI) UNIVERSITE D'AIX MARSEILLE
Call Details Starting Grant (StG), LS7, ERC-2016-STG
Summary Many efficient drugs have been designed to treat neurological disorders, but have failed in the clinic because they were toxic, could not cross the blood-brain barrier, and/or had deleterious side effects in healthy regions. I propose a conceptual breakthrough to solve these three issues, with minimally-invasive organic electronic ion pumps (OEIPs) to provide targeted treatment where and when it is needed. I will use epilepsy as the disease model because of its high rate of drug-resistance (30%) and will offer concrete opportunities for clinical transfer of such state-of-the-art technology.
The clinical problem: Resective surgery is frequently the last option available to a patient with drug-resistant epilepsy (> 1 million persons in the EU). However, surgery fails in 30% of the cases and can have deleterious consequences with severe postoperative neurological deficits (impaired motor function, speech and memory). Furthermore, some cases of epilepsy are simply untreatable surgically because resective surgery would leave unacceptable damage to core functions. Clearly, a new therapeutic approach is needed when neurosurgery is not possible or deemed too risky.
The OEIP solution: As I have demonstrated, OEIPs combine state-of-the-art organic electronics and pharmacology to control epileptiform activity in vitro by directly delivering inhibitory neurotransmitters on-demand. I additionally demonstrated that thin-film flexible organic electronics can be used to create minimally-invasive depth probes for implantation which significantly reduced tissue damage compared to standard rigid implants in vivo. I will integrate OEIPs on such probes creating devices which will have both the high-quality recordings provided by the organic electrodes for electrophysiological seizure detection and the molecular delivery capability of the OEIP for seizure intervention. The devices will be a closed-loop system to detect seizure onset and intervene in the affected brain region.
Summary
Many efficient drugs have been designed to treat neurological disorders, but have failed in the clinic because they were toxic, could not cross the blood-brain barrier, and/or had deleterious side effects in healthy regions. I propose a conceptual breakthrough to solve these three issues, with minimally-invasive organic electronic ion pumps (OEIPs) to provide targeted treatment where and when it is needed. I will use epilepsy as the disease model because of its high rate of drug-resistance (30%) and will offer concrete opportunities for clinical transfer of such state-of-the-art technology.
The clinical problem: Resective surgery is frequently the last option available to a patient with drug-resistant epilepsy (> 1 million persons in the EU). However, surgery fails in 30% of the cases and can have deleterious consequences with severe postoperative neurological deficits (impaired motor function, speech and memory). Furthermore, some cases of epilepsy are simply untreatable surgically because resective surgery would leave unacceptable damage to core functions. Clearly, a new therapeutic approach is needed when neurosurgery is not possible or deemed too risky.
The OEIP solution: As I have demonstrated, OEIPs combine state-of-the-art organic electronics and pharmacology to control epileptiform activity in vitro by directly delivering inhibitory neurotransmitters on-demand. I additionally demonstrated that thin-film flexible organic electronics can be used to create minimally-invasive depth probes for implantation which significantly reduced tissue damage compared to standard rigid implants in vivo. I will integrate OEIPs on such probes creating devices which will have both the high-quality recordings provided by the organic electrodes for electrophysiological seizure detection and the molecular delivery capability of the OEIP for seizure intervention. The devices will be a closed-loop system to detect seizure onset and intervene in the affected brain region.
Max ERC Funding
1 636 250 €
Duration
Start date: 2017-03-01, End date: 2022-02-28
Project acronym EPLORE
Project EPidemiological Left ventriclar Outcomes Research in Europe
Researcher (PI) Jan Albert Hendrik Staessen
Host Institution (HI) KATHOLIEKE UNIVERSITEIT LEUVEN
Call Details Advanced Grant (AdG), LS7, ERC-2011-ADG_20110310
Summary Heart failure (HF) affects 15 million Europeans and entails higher mortality and health care costs than cancer. EPLORE addresses this issue by prospective epidemiological research in 4 European countries and by a proof-of-concept clinical trial. WP1 will for the first time at the population level document the incidence and progression of subclinical LV dysfunction and clarify whether asymptomatic LV dysfunction, as picked up by the newest echocardiographic techniques, predicts cardiovascular (CV) outcomes, including HF. WP2 will investigate the contribution of ventricular-arterial coupling disease and mechanical LV dyssynchrony to subclinical LV dysfunction. WP3 will identify a set of urinary polypeptides that signify early LV dysfunction and validate these biomarkers by showing that they predict deterioration of LV function, progression to HF and the incidence of CV complications over and beyond established risk factors. WP3 will also search for novel panels of circulating biomarkers, of which combined measurement will add information (accuracy, sensitivity and specificity) to established biomarkers (e.g., NT-proBNP) and identify genetic variants involved in the progression of LV dysfunction, either causally or as biomarker. WP4 consists of a randomised clinical trial to translate in a high-risk high-gain setting the results of WPs 1-3 into clinical practice and to identify a new treatment modality that potentially slows progression of diastolic LV dysfunction. Dissemination in WP5 will contribute to new guidelines for the prevention and treatment of HF. WP6 includes governance, monitoring research strategies and output, and protection of IPR. In conclusion, EPLORE will advance risk stratification and the early diagnosis of subclinical HF. The project will potentially result into specific treatments for diastolic LV dysfunction and inform guidelines for prevention and treatment of HF. It will benefit 20% of Europeans who currently have subclinical LV dysfunction.
Summary
Heart failure (HF) affects 15 million Europeans and entails higher mortality and health care costs than cancer. EPLORE addresses this issue by prospective epidemiological research in 4 European countries and by a proof-of-concept clinical trial. WP1 will for the first time at the population level document the incidence and progression of subclinical LV dysfunction and clarify whether asymptomatic LV dysfunction, as picked up by the newest echocardiographic techniques, predicts cardiovascular (CV) outcomes, including HF. WP2 will investigate the contribution of ventricular-arterial coupling disease and mechanical LV dyssynchrony to subclinical LV dysfunction. WP3 will identify a set of urinary polypeptides that signify early LV dysfunction and validate these biomarkers by showing that they predict deterioration of LV function, progression to HF and the incidence of CV complications over and beyond established risk factors. WP3 will also search for novel panels of circulating biomarkers, of which combined measurement will add information (accuracy, sensitivity and specificity) to established biomarkers (e.g., NT-proBNP) and identify genetic variants involved in the progression of LV dysfunction, either causally or as biomarker. WP4 consists of a randomised clinical trial to translate in a high-risk high-gain setting the results of WPs 1-3 into clinical practice and to identify a new treatment modality that potentially slows progression of diastolic LV dysfunction. Dissemination in WP5 will contribute to new guidelines for the prevention and treatment of HF. WP6 includes governance, monitoring research strategies and output, and protection of IPR. In conclusion, EPLORE will advance risk stratification and the early diagnosis of subclinical HF. The project will potentially result into specific treatments for diastolic LV dysfunction and inform guidelines for prevention and treatment of HF. It will benefit 20% of Europeans who currently have subclinical LV dysfunction.
Max ERC Funding
2 391 440 €
Duration
Start date: 2012-07-01, End date: 2017-06-30
Project acronym EVICARE
Project Extracellular Vesicle-Inspired CArdiac Repair
Researcher (PI) Joseph Petrus Gerardus SLUIJTER
Host Institution (HI) UNIVERSITAIR MEDISCH CENTRUM UTRECHT
Call Details Consolidator Grant (CoG), LS7, ERC-2016-COG
Summary More than 3.5 million people are newly diagnosed with heart failure every year in Europe with a long-term prognosis of 50% mortality within 4 years. There is a major need for more innovative, regenerative therapies that have the potential to change the course of disease. My hypothesis is that we can recondition heart failure by stimulating cardiac repair with extracellular vesicles that are derived from progenitor cells. In my laboratory, extracellular released vesicles containing a cocktail of stimulating factors, are amongst the most potent vectors for cardiac repair.
To achieve a sustainable and long-term therapeutic effect of these vesicles and enhance cardiac function by stimulating myocardial repair, we will 1) improve local cardiac delivery of progenitor cell-derived extracellular vesicles, 2) understand the mechanism of action of extracellular vesicles, and 3) stimulate extracellular vesicles release and/or production by progenitor cells.
These questions form the rationale for the current proposal in which we will co-inject extracellular vesicles and slow-release biomaterials into the damaged myocardium. By subsequent genetic tracing, we will determine fate mapping of injected vesicles in vivo, and perform further mechanistic understanding in in vitro culture models of targeted and identified myocardial cell types. Moreover, we will upscale the vesicles production by progenitor cells further via bioreactor culturing and medium-throughput screening on factors that stimulate vesicles release.
The use of stem cell-derived extracellular vesicles to stimulate cardiac repair will potentially allow for an off-the shelf approach, including mechanistic understanding and future clinical use. Additionally, since these vesicles act as a natural carrier system outperforming current artificial drug delivery, we might understand and mimic their characteristics to enhance local (RNA-based) drug delivery systems for cardiovascular application.
Summary
More than 3.5 million people are newly diagnosed with heart failure every year in Europe with a long-term prognosis of 50% mortality within 4 years. There is a major need for more innovative, regenerative therapies that have the potential to change the course of disease. My hypothesis is that we can recondition heart failure by stimulating cardiac repair with extracellular vesicles that are derived from progenitor cells. In my laboratory, extracellular released vesicles containing a cocktail of stimulating factors, are amongst the most potent vectors for cardiac repair.
To achieve a sustainable and long-term therapeutic effect of these vesicles and enhance cardiac function by stimulating myocardial repair, we will 1) improve local cardiac delivery of progenitor cell-derived extracellular vesicles, 2) understand the mechanism of action of extracellular vesicles, and 3) stimulate extracellular vesicles release and/or production by progenitor cells.
These questions form the rationale for the current proposal in which we will co-inject extracellular vesicles and slow-release biomaterials into the damaged myocardium. By subsequent genetic tracing, we will determine fate mapping of injected vesicles in vivo, and perform further mechanistic understanding in in vitro culture models of targeted and identified myocardial cell types. Moreover, we will upscale the vesicles production by progenitor cells further via bioreactor culturing and medium-throughput screening on factors that stimulate vesicles release.
The use of stem cell-derived extracellular vesicles to stimulate cardiac repair will potentially allow for an off-the shelf approach, including mechanistic understanding and future clinical use. Additionally, since these vesicles act as a natural carrier system outperforming current artificial drug delivery, we might understand and mimic their characteristics to enhance local (RNA-based) drug delivery systems for cardiovascular application.
Max ERC Funding
1 997 298 €
Duration
Start date: 2017-09-01, End date: 2022-08-31
Project acronym EVOLVE
Project Extracellular Vesicle-Internalizing Receptors (EVIRs) for Cancer ImmunoGeneTherapy
Researcher (PI) Michele DE PALMA
Host Institution (HI) ECOLE POLYTECHNIQUE FEDERALE DE LAUSANNE
Call Details Consolidator Grant (CoG), LS7, ERC-2016-COG
Summary We are witnessing transformative results in the clinical application of both cancer immunotherapies and gene transfer
technologies. Tumor vaccines are a specific modality of cancer immunotherapy. Similar to vaccination against pathogens, tumor vaccines are designed to elicit a specific immune response against cancer. They are based on the administration of inactivated cancer cells or tumor antigens, or the inoculation of antigen-presenting cells (APCs) previously exposed to tumor antigens. In spite of significant development and testing, tumor vaccines have largely delivered unsatisfactory clinical results. Indeed, while some patients show dramatic and durable cancer regressions, many do not respond, highlighting both the potential and the shortcomings of current vaccination strategies. Hence, identifying and abating the barriers to effective cancer vaccines is key to broadening their therapeutic reach. The goal of EVOLVE (EVirs to Optimize and Leverage Vaccines for cancer Eradication) is to propel the development of effective APC-based tumor vaccines using an innovative strategy that overcomes several key hurdles associated with available treatments. EVOLVE puts forward a novel APC engineering platform whereby chimeric receptors are used to both enable the specific and efficient uptake of cancer-derived extracellular vesicles (EVs) into APCs, and to promote the cross-presentation of EV-associated tumor antigens for stimulating anti-tumor immunity. EVOLVE also envisions a combination of ancillary ‘outside of the box’ interventions, primarily based on further APC engineering combined with innovative pre-conditioning of the tumor microenvironment, to facilitate the deployment of effective APC-driven, T-cellmediated anti-tumor immunity. Further to preclinical trials in mouse models of breast cancer and melanoma, our APC platform will be used to prospectively identify novel human melanoma antigens and reactive T cell clones for broader immunotherapy applications.
Summary
We are witnessing transformative results in the clinical application of both cancer immunotherapies and gene transfer
technologies. Tumor vaccines are a specific modality of cancer immunotherapy. Similar to vaccination against pathogens, tumor vaccines are designed to elicit a specific immune response against cancer. They are based on the administration of inactivated cancer cells or tumor antigens, or the inoculation of antigen-presenting cells (APCs) previously exposed to tumor antigens. In spite of significant development and testing, tumor vaccines have largely delivered unsatisfactory clinical results. Indeed, while some patients show dramatic and durable cancer regressions, many do not respond, highlighting both the potential and the shortcomings of current vaccination strategies. Hence, identifying and abating the barriers to effective cancer vaccines is key to broadening their therapeutic reach. The goal of EVOLVE (EVirs to Optimize and Leverage Vaccines for cancer Eradication) is to propel the development of effective APC-based tumor vaccines using an innovative strategy that overcomes several key hurdles associated with available treatments. EVOLVE puts forward a novel APC engineering platform whereby chimeric receptors are used to both enable the specific and efficient uptake of cancer-derived extracellular vesicles (EVs) into APCs, and to promote the cross-presentation of EV-associated tumor antigens for stimulating anti-tumor immunity. EVOLVE also envisions a combination of ancillary ‘outside of the box’ interventions, primarily based on further APC engineering combined with innovative pre-conditioning of the tumor microenvironment, to facilitate the deployment of effective APC-driven, T-cellmediated anti-tumor immunity. Further to preclinical trials in mouse models of breast cancer and melanoma, our APC platform will be used to prospectively identify novel human melanoma antigens and reactive T cell clones for broader immunotherapy applications.
Max ERC Funding
1 958 919 €
Duration
Start date: 2017-07-01, End date: 2022-06-30
Project acronym FAST
Project Investigating new therapeutic approaches to Friedreich's Ataxia
Researcher (PI) Roberto Testi
Host Institution (HI) UNIVERSITA DEGLI STUDI DI ROMA TOR VERGATA
Call Details Advanced Grant (AdG), LS7, ERC-2011-ADG_20110310
Summary Friedreich’s Ataxia (FRDA) is a devastating degenerative disease with no specific therapy. It is passed by autosomal recessive inheritance and affects 1:30,000 individuals in Caucasian populations. Symptoms appear in the first decade of life and include progressive and unremitting lack of movement coordination, leading to complete inability, and dilated cardiomyopathy leading to congestive heart failure, the most common cause of premature death. FRDA is due to the insufficient transcription of the gene coding for the mitochondrial protein frataxin. Reduced cellular levels of frataxin cause impaired mitochondrial function and increased sensitivity to oxidative stress, leading to accelerated cell death in critical tissues.
Severity of the disease critically depends on residual frataxin levels. Therapeutic efforts are mostly focused on increasing cellular frataxin . We found that frataxin is normally degraded by the ubiquitin-proteasome system. We identified the lysine responsible for the ubiquitination of frataxin and, by computational screening followed by experimental validation, we identified and validated a series of small molecules, called ubiquitin-competing molecules (UCM), that prevent frataxin ubiquitination and induce frataxin accumulation in cells derived from FRDA patients. Moreover, treatment with UCM partially rescues aconitase and ATP production defects in cells derived from FRDA patients.
Our goal is two fold: 1) submit a set of leads we already identified, as well as their new and more complex derivatives, to preclinical testing in FRDA mice 2) identify the E3 ligase that is responsible for frataxin ubiquitination, and investigate the possibility to use it as a druggable target for small molecules to prevent frataxin degradation.
Summary
Friedreich’s Ataxia (FRDA) is a devastating degenerative disease with no specific therapy. It is passed by autosomal recessive inheritance and affects 1:30,000 individuals in Caucasian populations. Symptoms appear in the first decade of life and include progressive and unremitting lack of movement coordination, leading to complete inability, and dilated cardiomyopathy leading to congestive heart failure, the most common cause of premature death. FRDA is due to the insufficient transcription of the gene coding for the mitochondrial protein frataxin. Reduced cellular levels of frataxin cause impaired mitochondrial function and increased sensitivity to oxidative stress, leading to accelerated cell death in critical tissues.
Severity of the disease critically depends on residual frataxin levels. Therapeutic efforts are mostly focused on increasing cellular frataxin . We found that frataxin is normally degraded by the ubiquitin-proteasome system. We identified the lysine responsible for the ubiquitination of frataxin and, by computational screening followed by experimental validation, we identified and validated a series of small molecules, called ubiquitin-competing molecules (UCM), that prevent frataxin ubiquitination and induce frataxin accumulation in cells derived from FRDA patients. Moreover, treatment with UCM partially rescues aconitase and ATP production defects in cells derived from FRDA patients.
Our goal is two fold: 1) submit a set of leads we already identified, as well as their new and more complex derivatives, to preclinical testing in FRDA mice 2) identify the E3 ligase that is responsible for frataxin ubiquitination, and investigate the possibility to use it as a druggable target for small molecules to prevent frataxin degradation.
Max ERC Funding
1 496 200 €
Duration
Start date: 2012-03-01, End date: 2015-02-28
Project acronym FRAGMENT2DRUG
Project Jigsaw puzzles at atomic resolution: Computational design of GPCR drugs from fragments
Researcher (PI) Jens CARLSSON
Host Institution (HI) UPPSALA UNIVERSITET
Call Details Starting Grant (StG), LS7, ERC-2016-STG
Summary Despite technological advances, industry struggles to develop new pharmaceuticals and therefore novel strategies for drug discovery are urgently needed. G protein-coupled receptors (GPCRs) play important roles in numerous physiological processes and are important drug targets for neurological diseases. My research focuses on modelling of GPCR-ligand interactions at the atomic level, with the goal to increase knowledge of receptor function and develop new methods for drug discovery. Breakthroughs in GPCR structural biology and access to sensitive screening assays provide opportunities to utilize fragment-based lead discovery (FBLD), a powerful approach for drug design. The objective of the project is to create a computational platform for FBLD, with a vision to transform the early drug discovery process for GPCRs. As structural information for these targets is limited, predictive models of receptor-fragment complexes will be crucial for the successful use of FBLD. In this project, computational structure-based methods for discovery of fragment ligands and further optimization of these to potent leads will be developed. These techniques will be applied to address two difficult problems in drug discovery. The first of these is to design ligands of peptide-binding GPCRs that have been challenging for existing methods. One of the promises of FBLD is to provide access to difficult targets, which will be explored by combining molecular docking and biophysical screening against peptide-GPCRs to identify novel lead candidates. A second challenge is that efficient treatment of neurological disorders often requires modulation of multiple targets, which also will be the focus of the project.
Summary
Despite technological advances, industry struggles to develop new pharmaceuticals and therefore novel strategies for drug discovery are urgently needed. G protein-coupled receptors (GPCRs) play important roles in numerous physiological processes and are important drug targets for neurological diseases. My research focuses on modelling of GPCR-ligand interactions at the atomic level, with the goal to increase knowledge of receptor function and develop new methods for drug discovery. Breakthroughs in GPCR structural biology and access to sensitive screening assays provide opportunities to utilize fragment-based lead discovery (FBLD), a powerful approach for drug design. The objective of the project is to create a computational platform for FBLD, with a vision to transform the early drug discovery process for GPCRs. As structural information for these targets is limited, predictive models of receptor-fragment complexes will be crucial for the successful use of FBLD. In this project, computational structure-based methods for discovery of fragment ligands and further optimization of these to potent leads will be developed. These techniques will be applied to address two difficult problems in drug discovery. The first of these is to design ligands of peptide-binding GPCRs that have been challenging for existing methods. One of the promises of FBLD is to provide access to difficult targets, which will be explored by combining molecular docking and biophysical screening against peptide-GPCRs to identify novel lead candidates. A second challenge is that efficient treatment of neurological disorders often requires modulation of multiple targets, which also will be the focus of the project.
Max ERC Funding
1 467 500 €
Duration
Start date: 2017-11-01, End date: 2022-10-31
Project acronym FUNMETA
Project Metabolomics of fungal diseases: a systems biology approach for biomarkers discovery and therapy
Researcher (PI) Luigina Romani
Host Institution (HI) UNIVERSITA DEGLI STUDI DI PERUGIA
Call Details Advanced Grant (AdG), LS7, ERC-2011-ADG_20110310
Summary Humans have evolved intimate symbiotic relationships with a consortium of gut microbes (including fungi) and individual variations in the microbiome influence host health and disease. The fact that fungi are capable of colonizing almost every niche within the human body suggests that they must possess particular immune adaptation mechanisms, the breakdown of which may result in fatal fungal infections and severe fungal diseases. Traditional reductionist approaches of the past have not been sufficient to address these new challenges in the pathogenesis of fungal diseases. Here, I propose an integrated, systems biology approach to understand the role of L-tryptophan (trp) metabolic pathways in multilevel host−fungus interactions. Present in mammals as well as in fungi, pathways of trp metabolic pathways are exploited by the host and the fungal biota for survival and immune adaptation. A variety of indole derivatives, generated through conversion from dietary trp by symbiotic bacteria, activate the aryl hydrocarbon receptor/IL-22 pathway that provides antifungal resistance and tissue repair. Harmful inflammatory responses to fungi are instead tamed by kynurenines generated via the enzyme indoleamine 2,3–dioxygenase (IDO) of the trp pathway. Through high-throughput wet-lab ‘omics’ techniques combined with computational techniques, the project aims at defining the molecular basis of mammalian and fungal IDO activity and a metabolic network linking the metabolic phenotype (metabotype) to immune adaptations and its possible breakdown in experimental and human fungal infections. The project will provide ideal post-graduate training focussed on the development of metabolomics for diagnosis of fungal diseases and optimization of current antifungal therapy and diet that are of relevance to public health care solutions.
Summary
Humans have evolved intimate symbiotic relationships with a consortium of gut microbes (including fungi) and individual variations in the microbiome influence host health and disease. The fact that fungi are capable of colonizing almost every niche within the human body suggests that they must possess particular immune adaptation mechanisms, the breakdown of which may result in fatal fungal infections and severe fungal diseases. Traditional reductionist approaches of the past have not been sufficient to address these new challenges in the pathogenesis of fungal diseases. Here, I propose an integrated, systems biology approach to understand the role of L-tryptophan (trp) metabolic pathways in multilevel host−fungus interactions. Present in mammals as well as in fungi, pathways of trp metabolic pathways are exploited by the host and the fungal biota for survival and immune adaptation. A variety of indole derivatives, generated through conversion from dietary trp by symbiotic bacteria, activate the aryl hydrocarbon receptor/IL-22 pathway that provides antifungal resistance and tissue repair. Harmful inflammatory responses to fungi are instead tamed by kynurenines generated via the enzyme indoleamine 2,3–dioxygenase (IDO) of the trp pathway. Through high-throughput wet-lab ‘omics’ techniques combined with computational techniques, the project aims at defining the molecular basis of mammalian and fungal IDO activity and a metabolic network linking the metabolic phenotype (metabotype) to immune adaptations and its possible breakdown in experimental and human fungal infections. The project will provide ideal post-graduate training focussed on the development of metabolomics for diagnosis of fungal diseases and optimization of current antifungal therapy and diet that are of relevance to public health care solutions.
Max ERC Funding
2 299 200 €
Duration
Start date: 2012-04-01, End date: 2018-03-31
Project acronym GLIOMADDS
Project Development of tumor penetrating peptides for glioma targeting
Researcher (PI) Tambet Teesalu
Host Institution (HI) TARTU ULIKOOL
Call Details Starting Grant (StG), LS7, ERC-2011-StG_20101109
Summary This application addresses a major problem in therapy of solid tumors: poor penetration of anti-cancer drugs into tumor tissue and to infiltrating tumor cells. Recently, we have identified tumor penetrating peptides (TPP) that trigger specific penetration of co-administered un-conjugated drugs deep into tumor and increase their therapeutic index. Current TPP target angiogenic tumor vessels and may not be suitable for targeting slow-growing tumors and invasive tumor cells. TPP are composed of functional modules (tumor recruitment motif, cryptic tissue penetrating C-end Rule element, and a protease cleavage site), which can be rearranged to yield peptides of novel specificities.
Our goal is to develop TPP platform for delivery of co-administered drugs to the deadliest brain tumor – glioblastoma (GBM). High-grade glioma is a target that is particularly evasive and well-suited for tissue penetrative drug delivery. We will develop glioma-specific TPP (gTPP) by combination of in vivo and ex vivo phage display of constrained peptide libraries on state-of-the-art glioma animal models. These gTPP will be able to penetrate gliomas (and potentially other tumors) independent of their angiogenic status, and to deliver drugs to infiltrating malignant cells far from the bulk glioma lesion. We will characterize, validate, and optimize the gTPP platform for enhanced glioma delivery of co-injected drugs. These studies will provide the preclinical data needed to advance the gTPP combination therapy of glioma to GLP toxicology and subsequent IND filing.
Summary
This application addresses a major problem in therapy of solid tumors: poor penetration of anti-cancer drugs into tumor tissue and to infiltrating tumor cells. Recently, we have identified tumor penetrating peptides (TPP) that trigger specific penetration of co-administered un-conjugated drugs deep into tumor and increase their therapeutic index. Current TPP target angiogenic tumor vessels and may not be suitable for targeting slow-growing tumors and invasive tumor cells. TPP are composed of functional modules (tumor recruitment motif, cryptic tissue penetrating C-end Rule element, and a protease cleavage site), which can be rearranged to yield peptides of novel specificities.
Our goal is to develop TPP platform for delivery of co-administered drugs to the deadliest brain tumor – glioblastoma (GBM). High-grade glioma is a target that is particularly evasive and well-suited for tissue penetrative drug delivery. We will develop glioma-specific TPP (gTPP) by combination of in vivo and ex vivo phage display of constrained peptide libraries on state-of-the-art glioma animal models. These gTPP will be able to penetrate gliomas (and potentially other tumors) independent of their angiogenic status, and to deliver drugs to infiltrating malignant cells far from the bulk glioma lesion. We will characterize, validate, and optimize the gTPP platform for enhanced glioma delivery of co-injected drugs. These studies will provide the preclinical data needed to advance the gTPP combination therapy of glioma to GLP toxicology and subsequent IND filing.
Max ERC Funding
1 499 931 €
Duration
Start date: 2012-01-01, End date: 2016-12-31
Project acronym GRIP-HEALTH
Project Getting Research Into Policy in Public Health : transforming the evidence-to-policy response through utilisation of political insights
Researcher (PI) Justin Oliver Parkhurst
Host Institution (HI) LONDON SCHOOL OF HYGIENE AND TROPICAL MEDICINE ROYAL CHARTER
Call Details Starting Grant (StG), LS7, ERC-2011-StG_20101109
Summary There has been increasing frustration in the field of public health with the lack of uptake of research evidence, and continued difficulties in getting evidence of effective health interventions into policy and practice. Calls have often been made to ‘bridge the gap’ between researchers and policy makers, yet little work has been done to analyse the political factors influencing research-to-policy process in public health, and the ways national governments may respond to these challenges. We airm to take the next step in improving the use of evidence in health policy by integrating political data and analysis into efforts to build the government institutions and responses which take up research evidence. Specifically we will apply political science theories and methods from the sub-fields of policy analysis, and public administration to achieve this end. These disciplines have not yet been applied to the challenge of getting evidence into policy in health. A programme of work drawing on these fields can take the next step in the evidence-based health policy movement. The work will map out the multiple political challenges to evidence use, while providing recommendations on how to build government structures supportive of evidence informed policy in particular policy contexts. We will review existing literature, construct a conceptual framework linking political insights with national responses, conduct empirical case studies across a range of countries, and develop a tool to improve government responses to the use of evidence in public health. Finally this programme will work to create a center of excellence on the study of evidence and policy in health.
Summary
There has been increasing frustration in the field of public health with the lack of uptake of research evidence, and continued difficulties in getting evidence of effective health interventions into policy and practice. Calls have often been made to ‘bridge the gap’ between researchers and policy makers, yet little work has been done to analyse the political factors influencing research-to-policy process in public health, and the ways national governments may respond to these challenges. We airm to take the next step in improving the use of evidence in health policy by integrating political data and analysis into efforts to build the government institutions and responses which take up research evidence. Specifically we will apply political science theories and methods from the sub-fields of policy analysis, and public administration to achieve this end. These disciplines have not yet been applied to the challenge of getting evidence into policy in health. A programme of work drawing on these fields can take the next step in the evidence-based health policy movement. The work will map out the multiple political challenges to evidence use, while providing recommendations on how to build government structures supportive of evidence informed policy in particular policy contexts. We will review existing literature, construct a conceptual framework linking political insights with national responses, conduct empirical case studies across a range of countries, and develop a tool to improve government responses to the use of evidence in public health. Finally this programme will work to create a center of excellence on the study of evidence and policy in health.
Max ERC Funding
1 496 403 €
Duration
Start date: 2012-01-01, End date: 2016-12-31
Project acronym GTNCTV
Project Gene therapy and nanotechnology based CNS targeted vectors
Researcher (PI) Mimoun Azzouz
Host Institution (HI) THE UNIVERSITY OF SHEFFIELD
Call Details Advanced Grant (AdG), LS7, ERC-2011-ADG_20110310
Summary Targeting therapeutic genes selectively into the central nervous system (CNS) is a crucial precondition for translation of gene therapy strategies into human trials. The current multidisciplinary proposal integrates expertise identified as essential in the effective acceleration of research to overcome bottlenecks in the field including: 1) Inefficiency of therapy delivery to the CNS because of factors like the blood-brain barrier (BBB); 2) Poor understanding of disease mechanisms at the molecular and cellular levels. These problems must be overcome to develop fully effective treatments for neurological disorders. Currently the adeno-associated (AAV)-based system is one of the most refined and effective gene delivery systems for neuronal cells. In contrast to all other systems, it has been possible to engineer AAV9 to deliver genes through the BBB to the CNS by intravascular (IV) administration. However, following IV delivery, these vectors also target liver and other tissues, with significant potential for untoward effects. This has prompted us to adopt two major strategies: i) targeting of AAV9 vectors at the level of transcription by insertion of hybrid motor neuron specific promoters into the vector genome; ii) development of a CNS-targeted delivery approach based on state-of-the art nanoparticle-mediated encapsulation of AAV9 vectors. We anticipate that engineering strategies with the ability to restrict transgene expression to CNS tissue will significantly overcome various existing hurdles in CNS gene therapy development. Our objectives are to: 1) explore mechanisms leading to penetration of scAAV9 vectors through BBB since the exact mechanism of AAV9 diffusion through BBB is unknown; 2) design novel targeted strategies with enhanced tropism to CNS; 3) use CNS targeted vectors to investigate mechanisms of motor neuron death linked to mutations in RNA processing genes; 4) utilise CNS-targeted systems to test therapeutic strategies for motor neuron diseases.
Summary
Targeting therapeutic genes selectively into the central nervous system (CNS) is a crucial precondition for translation of gene therapy strategies into human trials. The current multidisciplinary proposal integrates expertise identified as essential in the effective acceleration of research to overcome bottlenecks in the field including: 1) Inefficiency of therapy delivery to the CNS because of factors like the blood-brain barrier (BBB); 2) Poor understanding of disease mechanisms at the molecular and cellular levels. These problems must be overcome to develop fully effective treatments for neurological disorders. Currently the adeno-associated (AAV)-based system is one of the most refined and effective gene delivery systems for neuronal cells. In contrast to all other systems, it has been possible to engineer AAV9 to deliver genes through the BBB to the CNS by intravascular (IV) administration. However, following IV delivery, these vectors also target liver and other tissues, with significant potential for untoward effects. This has prompted us to adopt two major strategies: i) targeting of AAV9 vectors at the level of transcription by insertion of hybrid motor neuron specific promoters into the vector genome; ii) development of a CNS-targeted delivery approach based on state-of-the art nanoparticle-mediated encapsulation of AAV9 vectors. We anticipate that engineering strategies with the ability to restrict transgene expression to CNS tissue will significantly overcome various existing hurdles in CNS gene therapy development. Our objectives are to: 1) explore mechanisms leading to penetration of scAAV9 vectors through BBB since the exact mechanism of AAV9 diffusion through BBB is unknown; 2) design novel targeted strategies with enhanced tropism to CNS; 3) use CNS targeted vectors to investigate mechanisms of motor neuron death linked to mutations in RNA processing genes; 4) utilise CNS-targeted systems to test therapeutic strategies for motor neuron diseases.
Max ERC Funding
2 499 959 €
Duration
Start date: 2012-03-01, End date: 2017-10-31
Project acronym HEARTMAPAS
Project Single Heart beat MApping of myocardial Performance, Activation, and Scar by ultrasound
Researcher (PI) Jan Robert Michel D'hooge
Host Institution (HI) KATHOLIEKE UNIVERSITEIT LEUVEN
Call Details Starting Grant (StG), LS7, ERC-2011-StG_20101109
Summary Heart failure affects about 2% of the European population with an annual mortality rate of 10%. Cardiac Resynchronization Therapy (CRT) has been proven to reduce morbidity and mortality and has become a recommended treatment. Optimal lead placement for CRT not only requires exact anatomically mapped information on the mechanical activation sequence to be corrected but also tissue viability and performance maps. Unfortunately, to date, no imaging technique allows building such maps non-invasively in a single examination in CRT patients.
In this project, a new ultrasound imaging approach is proposed that will not only allow mapping all of these characteristics in a single examination but that will actually do this in a single heart beat.
Hereto, real-time segmentation of the left ventricle will be used to limit the data acquisition to the spatial regions that contain myocardium. In this way unnecessary spatial sampling can be avoided which combined with new beam forming strategies will allow imaging the entire left ventricle at frame rates above 1000Hz. This very high temporal resolution will be used to accurately measure the onset of local deformation of the left ventricle in order to construct a mechanical activation map. Subsequently, cardiac motion estimates will be used to track anatomical regions throughout the cardiac cycle in order to construct a temporally averaged backscatter intensity map. As scar tissue is known to be more reflective than normal myocardium, this should allow mapping of scar. Finally, the automatic segmentation process will allow to locally measure wall thickness and curvature from which the mechanical load distribution within the ventricle can be derived. This, combined with estimates of regional myocardial deformation will produce a map of myocardial performance.
The proposed system will thus provide important new diagnostic and therapeutic information and will therefore allow better CRT planning for the individual heart failure patient.
Summary
Heart failure affects about 2% of the European population with an annual mortality rate of 10%. Cardiac Resynchronization Therapy (CRT) has been proven to reduce morbidity and mortality and has become a recommended treatment. Optimal lead placement for CRT not only requires exact anatomically mapped information on the mechanical activation sequence to be corrected but also tissue viability and performance maps. Unfortunately, to date, no imaging technique allows building such maps non-invasively in a single examination in CRT patients.
In this project, a new ultrasound imaging approach is proposed that will not only allow mapping all of these characteristics in a single examination but that will actually do this in a single heart beat.
Hereto, real-time segmentation of the left ventricle will be used to limit the data acquisition to the spatial regions that contain myocardium. In this way unnecessary spatial sampling can be avoided which combined with new beam forming strategies will allow imaging the entire left ventricle at frame rates above 1000Hz. This very high temporal resolution will be used to accurately measure the onset of local deformation of the left ventricle in order to construct a mechanical activation map. Subsequently, cardiac motion estimates will be used to track anatomical regions throughout the cardiac cycle in order to construct a temporally averaged backscatter intensity map. As scar tissue is known to be more reflective than normal myocardium, this should allow mapping of scar. Finally, the automatic segmentation process will allow to locally measure wall thickness and curvature from which the mechanical load distribution within the ventricle can be derived. This, combined with estimates of regional myocardial deformation will produce a map of myocardial performance.
The proposed system will thus provide important new diagnostic and therapeutic information and will therefore allow better CRT planning for the individual heart failure patient.
Max ERC Funding
1 602 401 €
Duration
Start date: 2012-02-01, End date: 2018-01-31
Project acronym HEPASPHER
Project Mimicking liver disease and regeneration in vitro for drug development and liver transplantation
Researcher (PI) Magnus INGELMAN-SUNDBERG
Host Institution (HI) KAROLINSKA INSTITUTET
Call Details Advanced Grant (AdG), LS7, ERC-2016-ADG
Summary The liver is a vital organ for synthesis and detoxification. The most significant liver diseases are hepatitis, non alcoholic fatty liver disease (NAFLD), non-alcoholic fatty liver steatohepatitis (NASH), carcinoma and cirrhosis. An additional and important cause of liver injury is adverse drug reactions (ADRs). In particular NAFLD is the most common liver disease affecting between 20% and 44% of European adults and 43-70% of patients with type 2 diabetes, and is one prime cause for chronic and end-stage liver disease, such as cirrhosis and primary hepatocellular carcinoma.
This proposal is based on recent findings in the laboratory: The development of novel 3D spheroid system with chemically defined media allowing studies of chronic drug toxicity, relevant liver disease and liver function for 5 weeks in vitro, the finding of the role of miRNA in hepatocyte dedifferentiation and that hepatocytes during spheroid formation first de-differentiate but later in spheroids re-differentiate to an in vivo relevant phenotype. This forms the basis for the main objectives: i) to study diseased liver in vitro with identification of mechanisms, biomarkers and novel drug candidates for treatment of NAFLD and fibrosis, ii) evaluate drug toxicity sensitivity and mechanisms in diseased liver systems and iii) further develop methods for hepatocyte proliferation and regeneration in vitro for transplantation purposes, including genetic editing in cases of hepatocytes obtained from patients with genetically inherited liver diseases.
This work is carried out in close contact with the Hepatology unit at the Karolinska Hospital partly using resources at the Science for Life Laboratory at Karolinska. It is anticipated that the project can provide with novel mechanisms, biomarkers and new targets for treatment of liver disease as well as novel methods for clinically applicable liver regeneration without the use of stem cells or transformed cells.
Summary
The liver is a vital organ for synthesis and detoxification. The most significant liver diseases are hepatitis, non alcoholic fatty liver disease (NAFLD), non-alcoholic fatty liver steatohepatitis (NASH), carcinoma and cirrhosis. An additional and important cause of liver injury is adverse drug reactions (ADRs). In particular NAFLD is the most common liver disease affecting between 20% and 44% of European adults and 43-70% of patients with type 2 diabetes, and is one prime cause for chronic and end-stage liver disease, such as cirrhosis and primary hepatocellular carcinoma.
This proposal is based on recent findings in the laboratory: The development of novel 3D spheroid system with chemically defined media allowing studies of chronic drug toxicity, relevant liver disease and liver function for 5 weeks in vitro, the finding of the role of miRNA in hepatocyte dedifferentiation and that hepatocytes during spheroid formation first de-differentiate but later in spheroids re-differentiate to an in vivo relevant phenotype. This forms the basis for the main objectives: i) to study diseased liver in vitro with identification of mechanisms, biomarkers and novel drug candidates for treatment of NAFLD and fibrosis, ii) evaluate drug toxicity sensitivity and mechanisms in diseased liver systems and iii) further develop methods for hepatocyte proliferation and regeneration in vitro for transplantation purposes, including genetic editing in cases of hepatocytes obtained from patients with genetically inherited liver diseases.
This work is carried out in close contact with the Hepatology unit at the Karolinska Hospital partly using resources at the Science for Life Laboratory at Karolinska. It is anticipated that the project can provide with novel mechanisms, biomarkers and new targets for treatment of liver disease as well as novel methods for clinically applicable liver regeneration without the use of stem cells or transformed cells.
Max ERC Funding
2 413 449 €
Duration
Start date: 2017-09-01, End date: 2022-08-31
Project acronym HIVBIOCHIP
Project A POINT-OF-CARE BIOCHIP FOR HIV MONITORING IN THE DEVELOPING WORLD
Researcher (PI) Nikolaos Chronis
Host Institution (HI) "NATIONAL CENTER FOR SCIENTIFIC RESEARCH ""DEMOKRITOS"""
Call Details Starting Grant (StG), LS7, ERC-2011-StG_20101109
Summary HIV/AIDS is one of the most destructive pandemics in human history, responsible for more than 25 million deaths. More than 30 million people live with limited or no access to therapeutic treatments, mainly due to the high cost of highly active antiretroviral therapies (HAART) and current diagnostic tests as well as due to the lack of basic infrastructure (e.g. lack of electricity, no trained personnel) that can support these tests. The need for innovative, inexpensive diagnostic instrumentation technology that can be used in resource-limited settings is immediate.
While programs that offer free HAART are being implemented in resource-limited settings, no diagnostic tests are available for evaluating the efficacy of HAART provided for the reasons mentioned above. Efficient management of HAART requires monitoring the course of HIV infection over time. The World Health Organization (WHO) recommends the CD4 T-cell count test for monitoring the clinical status of HIV individuals in resource-limited settings.
We propose to develop a portable, inexpensive, MEMS (MicroElectroMechanical Systems)-based, imaging system for counting the absolute number of CD4 cells from 1 l of whole blood. We use the term ‘imaging system’ to denote the different approach we follow for counting CD4 cells: rather the reading one by one singles cells (as it is done with flow cytometry), our system can image simultaneously thousands of individual cells, pre-assembled on the surface of a biochip. Although the proposed imaging system can replace current expensive cell counting instrumentation, our goal is to develop a system that can reach the end-user wherever limited infrastructure is present and no access to a hospital or clinic is possible. Such technology will not only enable to monitor the efficacy of an individual’s HAART in the developing world, but it will make more medicines available by identifying patients who need a treatment from patients who do not need it.
Summary
HIV/AIDS is one of the most destructive pandemics in human history, responsible for more than 25 million deaths. More than 30 million people live with limited or no access to therapeutic treatments, mainly due to the high cost of highly active antiretroviral therapies (HAART) and current diagnostic tests as well as due to the lack of basic infrastructure (e.g. lack of electricity, no trained personnel) that can support these tests. The need for innovative, inexpensive diagnostic instrumentation technology that can be used in resource-limited settings is immediate.
While programs that offer free HAART are being implemented in resource-limited settings, no diagnostic tests are available for evaluating the efficacy of HAART provided for the reasons mentioned above. Efficient management of HAART requires monitoring the course of HIV infection over time. The World Health Organization (WHO) recommends the CD4 T-cell count test for monitoring the clinical status of HIV individuals in resource-limited settings.
We propose to develop a portable, inexpensive, MEMS (MicroElectroMechanical Systems)-based, imaging system for counting the absolute number of CD4 cells from 1 l of whole blood. We use the term ‘imaging system’ to denote the different approach we follow for counting CD4 cells: rather the reading one by one singles cells (as it is done with flow cytometry), our system can image simultaneously thousands of individual cells, pre-assembled on the surface of a biochip. Although the proposed imaging system can replace current expensive cell counting instrumentation, our goal is to develop a system that can reach the end-user wherever limited infrastructure is present and no access to a hospital or clinic is possible. Such technology will not only enable to monitor the efficacy of an individual’s HAART in the developing world, but it will make more medicines available by identifying patients who need a treatment from patients who do not need it.
Max ERC Funding
1 986 000 €
Duration
Start date: 2012-06-01, End date: 2017-05-31
Project acronym HULSCTARGETING
Project Human leukemic stem cells: from identification towards targeting and eradication
Researcher (PI) Jan Jacob Schuringa
Host Institution (HI) ACADEMISCH ZIEKENHUIS GRONINGEN
Call Details Starting Grant (StG), LS7, ERC-2011-StG_20101109
Summary Human myeloid leukemias are life-threatening disorders. Although with current therapies the bulk of the tumor is readily eradicated, it is particularly the rare population of leukemic stem cells (LSCs) that is relatively quiescent and is very difficult to target. As a consequence, relapse of the disease occurs frequently resulting in poor survival rates. Nevertheless, remarkable differences do exist between subsets of patients. It is important to realize that leukemias are rather heterogeneous disorders that can be caused by a multiplicity of genetic and epigenetic changes. One of the most important challenges in the field today lies in establishing which of the differences between LSCs and normal hematopoietic stem cells (HSCs) provide attractive targets for the identification, targeting and ultimately the selective eradication of LSCs. Given the heterogeneity of the disorder, it will be important to address these aspects in detail in a leukemia subtype-specific manner.
Central to this proposal is the establishment of a human xenograft mouse leukemia “clinic” in which all important human leukemia subtypes will be represented. LSCs from patients as well as from cord blood and bone marrow model systems that we have generated will be equipped with luciferase-GFP tracers in order to allow detection of leukemic development in alive immunodeficient mice, as well as with unique barcodes that will allow clonal tracking. Within this mouse clinic, in a leukemia subtype-specific manner, we will be able to: 1) identify (novel) leukemic stem cell markers and evaluate their targetability; 2) evaluate whether stem cell intrinsic versus extrinsic signaling can be used as targets in alternative approaches to eradicate LSCs, and 3) study clonal evolution from de novo to relapsed leukemia. Our studies should provide insight into the biology of leukemia and novel rational approaches to treat leukemia patients more successfully.
Summary
Human myeloid leukemias are life-threatening disorders. Although with current therapies the bulk of the tumor is readily eradicated, it is particularly the rare population of leukemic stem cells (LSCs) that is relatively quiescent and is very difficult to target. As a consequence, relapse of the disease occurs frequently resulting in poor survival rates. Nevertheless, remarkable differences do exist between subsets of patients. It is important to realize that leukemias are rather heterogeneous disorders that can be caused by a multiplicity of genetic and epigenetic changes. One of the most important challenges in the field today lies in establishing which of the differences between LSCs and normal hematopoietic stem cells (HSCs) provide attractive targets for the identification, targeting and ultimately the selective eradication of LSCs. Given the heterogeneity of the disorder, it will be important to address these aspects in detail in a leukemia subtype-specific manner.
Central to this proposal is the establishment of a human xenograft mouse leukemia “clinic” in which all important human leukemia subtypes will be represented. LSCs from patients as well as from cord blood and bone marrow model systems that we have generated will be equipped with luciferase-GFP tracers in order to allow detection of leukemic development in alive immunodeficient mice, as well as with unique barcodes that will allow clonal tracking. Within this mouse clinic, in a leukemia subtype-specific manner, we will be able to: 1) identify (novel) leukemic stem cell markers and evaluate their targetability; 2) evaluate whether stem cell intrinsic versus extrinsic signaling can be used as targets in alternative approaches to eradicate LSCs, and 3) study clonal evolution from de novo to relapsed leukemia. Our studies should provide insight into the biology of leukemia and novel rational approaches to treat leukemia patients more successfully.
Max ERC Funding
1 499 820 €
Duration
Start date: 2012-01-01, End date: 2016-12-31
Project acronym iAML-lncTARGET
Project Targeting the transcriptional landscape in infant AML
Researcher (PI) Jan-Henning Cornelius KLUSMANN
Host Institution (HI) MARTIN-LUTHER-UNIVERSITAET HALLE-WITTENBERG
Call Details Starting Grant (StG), LS7, ERC-2016-STG
Summary Infant acute myeloid leukemia (AML) has a dismal prognosis, with a high prevalence of unfavorable features and increased susceptibility to therapy-related toxicities, highlighting the need for innovative treatment approaches. Despite the discovery of an enormous number and diversity of transcriptional products arising from the previously presumed wastelands of the non-protein-coding genome, our knowledge of non-coding RNAs is far from being incorporated into standards of AML diagnosis and treatment. I hypothesize that the highly developmental stage- and cell-specific expression of long non-coding RNAs shapes a chromatin and transcriptional landscape in fetal hematopoietic stem cells that renders them permissive towards transformation. I predict this landscape to synergize with particular oncogenes that are otherwise not oncogenic in adult cells, by providing a fertile transcriptional background for establishing and maintaining oncogenic programs. Therefore, the non-coding transcriptome, inherited from the fetal cell of origin, may reflect a previously unrecognized Achilles heel of infant AML, which I will identify with my expertise to understand and edit the AML genome and transcriptome.
I will apply recent breakthroughs from various research areas to i) create a comprehensive transcriptomic atlas of infant AML and fetal stem cells, ii) define aberrant or fetal stage-specific non-coding RNAs that drive leukemia progression, and iii) resolve their features to probe the oncogenic interactome. After iv) establishing a biobank of patient-derived xenografts, I will v) evaluate preclinical RNA-centered therapeutic interventions to overcome current obstacles in the treatment of infant AML. Targeting the vulnerable fetal stage-specific background of infant AML inherited from the cell of origin may set a paradigm shift for cancer treatment, by focusing on the permissive basis required by the oncogene for inducing and sustaining cancer, rather than on the oncogene itself.
Summary
Infant acute myeloid leukemia (AML) has a dismal prognosis, with a high prevalence of unfavorable features and increased susceptibility to therapy-related toxicities, highlighting the need for innovative treatment approaches. Despite the discovery of an enormous number and diversity of transcriptional products arising from the previously presumed wastelands of the non-protein-coding genome, our knowledge of non-coding RNAs is far from being incorporated into standards of AML diagnosis and treatment. I hypothesize that the highly developmental stage- and cell-specific expression of long non-coding RNAs shapes a chromatin and transcriptional landscape in fetal hematopoietic stem cells that renders them permissive towards transformation. I predict this landscape to synergize with particular oncogenes that are otherwise not oncogenic in adult cells, by providing a fertile transcriptional background for establishing and maintaining oncogenic programs. Therefore, the non-coding transcriptome, inherited from the fetal cell of origin, may reflect a previously unrecognized Achilles heel of infant AML, which I will identify with my expertise to understand and edit the AML genome and transcriptome.
I will apply recent breakthroughs from various research areas to i) create a comprehensive transcriptomic atlas of infant AML and fetal stem cells, ii) define aberrant or fetal stage-specific non-coding RNAs that drive leukemia progression, and iii) resolve their features to probe the oncogenic interactome. After iv) establishing a biobank of patient-derived xenografts, I will v) evaluate preclinical RNA-centered therapeutic interventions to overcome current obstacles in the treatment of infant AML. Targeting the vulnerable fetal stage-specific background of infant AML inherited from the cell of origin may set a paradigm shift for cancer treatment, by focusing on the permissive basis required by the oncogene for inducing and sustaining cancer, rather than on the oncogene itself.
Max ERC Funding
1 499 750 €
Duration
Start date: 2017-06-01, End date: 2022-05-31
Project acronym iHEAR
Project Investigating the meanings and mechanisms of psychotic experiences in young people: a novel, mixed-methods approach
Researcher (PI) Mary CANNON
Host Institution (HI) ROYAL COLLEGE OF SURGEONS IN IRELAND
Call Details Consolidator Grant (CoG), LS7, ERC-2016-COG
Summary Up to one fifth of young people have had the experience of psychotic symptoms, such as hearing voices when there is no-one around, or seeing visions. We now know that young people who experience these symptoms are at increased risk of developing psychotic disorders in adulthood. We also know that these young people are at higher risk of a range of co-morbid disorders such as depression and anxiety, and particularly suicidal behaviours. On the other hand, many of these young people will remain well and, for them, the psychotic experiences were merely a transitory phenomenon.
Childhood trauma is known to be associated with increased risk for psychotic symptoms and is a promising target for intervention. However we do not yet know enough about what types or timing of stressors are involved in the pathogenesis of psychotic symptoms, nor the mechanism by which early life stress may lead to changes in brain structure and function resulting in symptoms such as hallucinations. We also need to be able to identify those young people who will benefit most from intervention.
This ground-breaking, multi-disciplinary programme of work sets out to address these issues by drawing together epidemiology, social science, anthropology and neuroscience to devise a comprehensive programme of work examining the relationship between early life stress and psychotic symptoms among young people.
Designed as three inter-related work packages, this iHEAR programme will exploit a large population-based cohort and will capitalise on my existing unique cohort of young people, who were known to have experienced psychotic symptoms in childhood, as they enter young adulthood. This iHEAR programme will result in new information which will allow the development of innovative interventions to prevent or pre-empt severe mental illness in later life.
Summary
Up to one fifth of young people have had the experience of psychotic symptoms, such as hearing voices when there is no-one around, or seeing visions. We now know that young people who experience these symptoms are at increased risk of developing psychotic disorders in adulthood. We also know that these young people are at higher risk of a range of co-morbid disorders such as depression and anxiety, and particularly suicidal behaviours. On the other hand, many of these young people will remain well and, for them, the psychotic experiences were merely a transitory phenomenon.
Childhood trauma is known to be associated with increased risk for psychotic symptoms and is a promising target for intervention. However we do not yet know enough about what types or timing of stressors are involved in the pathogenesis of psychotic symptoms, nor the mechanism by which early life stress may lead to changes in brain structure and function resulting in symptoms such as hallucinations. We also need to be able to identify those young people who will benefit most from intervention.
This ground-breaking, multi-disciplinary programme of work sets out to address these issues by drawing together epidemiology, social science, anthropology and neuroscience to devise a comprehensive programme of work examining the relationship between early life stress and psychotic symptoms among young people.
Designed as three inter-related work packages, this iHEAR programme will exploit a large population-based cohort and will capitalise on my existing unique cohort of young people, who were known to have experienced psychotic symptoms in childhood, as they enter young adulthood. This iHEAR programme will result in new information which will allow the development of innovative interventions to prevent or pre-empt severe mental illness in later life.
Max ERC Funding
1 781 623 €
Duration
Start date: 2017-06-01, End date: 2022-05-31
Project acronym iMDx
Project Microfiber Electrofluidics for Integrated Molecular Diagnostics
Researcher (PI) Mahiar Max HAMEDI
Host Institution (HI) KUNGLIGA TEKNISKA HOEGSKOLAN
Call Details Starting Grant (StG), LS7, ERC-2016-STG
Summary "Diagnosis of an increasing number of diseases (such as HIV, Ebola and antibiotic resistant bacteria) rely on centralized laboratories with specialized instruments and skilled personnel. This type of clinical analysis with long lead times and high cost is inappropriate for point of care diagnostics. Portable, automated, and disposable devices with integrated molecular diagnostics (iMDx), could solve this problem. Such devices, however, basically do not exist today.
This research program will develop materials, methods, and diagnostic techniques to enable portable integrated molecular diagnostics (iMDx) devices, that are disposable (almost zero-cost), and that can detect a broad range of diseases using DNA amplification. To achieve this, we will develop ""3D microfiber electrofluidics (MEF)"". MEFs rely on simple and readily available porous sheets including paper, and textiles. Their fabrication is simple, and yet they are very powerful because they can monolithically integrate 3D microfluidics, 3D microelectronics, electrochemical analysis, storage of reagents / biomolecules, as well as handling and manipulation of cells.
We will develop techniques, using the combined capabilities of MEFs, for real-time DNA amplification and electrochemical DNA detection directly from samples. We will use three amplifications techniques for realizing a comprehensive iMDx platform: i) DNA detection, using established and state of the art amplification methods (e.g. PCR, LAMP, RPA). ii) RNA detection, using ""nucleic acid sequence-based amplification"" (NASBA). iii) Protein detection, using antibody to DNA translation through ""proximity ligation assay"" LPA, and using the detection of the amplified DNA is the indirect measure of proteins.
The outcome of this program has the potential to be ground breaking, and change the structure of measuring, processing, and pricing of biomedical information, and dramatically broaden its accessibility and applicability.
"
Summary
"Diagnosis of an increasing number of diseases (such as HIV, Ebola and antibiotic resistant bacteria) rely on centralized laboratories with specialized instruments and skilled personnel. This type of clinical analysis with long lead times and high cost is inappropriate for point of care diagnostics. Portable, automated, and disposable devices with integrated molecular diagnostics (iMDx), could solve this problem. Such devices, however, basically do not exist today.
This research program will develop materials, methods, and diagnostic techniques to enable portable integrated molecular diagnostics (iMDx) devices, that are disposable (almost zero-cost), and that can detect a broad range of diseases using DNA amplification. To achieve this, we will develop ""3D microfiber electrofluidics (MEF)"". MEFs rely on simple and readily available porous sheets including paper, and textiles. Their fabrication is simple, and yet they are very powerful because they can monolithically integrate 3D microfluidics, 3D microelectronics, electrochemical analysis, storage of reagents / biomolecules, as well as handling and manipulation of cells.
We will develop techniques, using the combined capabilities of MEFs, for real-time DNA amplification and electrochemical DNA detection directly from samples. We will use three amplifications techniques for realizing a comprehensive iMDx platform: i) DNA detection, using established and state of the art amplification methods (e.g. PCR, LAMP, RPA). ii) RNA detection, using ""nucleic acid sequence-based amplification"" (NASBA). iii) Protein detection, using antibody to DNA translation through ""proximity ligation assay"" LPA, and using the detection of the amplified DNA is the indirect measure of proteins.
The outcome of this program has the potential to be ground breaking, and change the structure of measuring, processing, and pricing of biomedical information, and dramatically broaden its accessibility and applicability.
"
Max ERC Funding
1 499 180 €
Duration
Start date: 2017-08-01, End date: 2022-07-31
Project acronym IndiviStat
Project Individualizing statin therapy by using a systems pharmacology decision support algorithm
Researcher (PI) Mikko Olavi NIEMI
Host Institution (HI) HELSINGIN YLIOPISTO
Call Details Consolidator Grant (CoG), LS7, ERC-2016-COG
Summary Background: Statins are essential drugs in the treatment of hypercholesterolaemia and are among the most prescribed drugs worldwide. The response to statin therapy varies widely between individuals. While most patients show good efficacy, a significant proportion of individuals show poor or even a lack of cholesterol-lowering efficacy. Moreover, a number of patients experience adverse drug reactions. These together with the lack of immediate effect on well-being likely explain the relatively poor adherence to statin therapy. Poor adherence to statins in turn increases the incidence of cardiovascular events and mortality.
Aims: The objectives of this project are 1) to develop a systems pharmacology model for predicting statin efficacy and tolerability at the level of an individual patient and 2) to investigate whether selecting the statin based on the model improves treatment adherence.
Methods: A systems pharmacology approach will be used to integrate data from in vitro and clinical studies. Semi-physiological pharmacokinetic-dynamic-toxicologic models will be built for each statin allowing the prediction of the pharmacokinetic and clinical outcomes for patients with different characteristics, genotypes, and concomitant medications. The ability of the systems pharmacology algorithm to enhance adherence will be investigated in a randomized clinical trial.
Significance: Systems pharmacology models have been increasingly applied in drug development, for example to predict the effect of organ dysfunction on pharmacokinetics. The proposed project is the first to use systems pharmacology predictions to guide clinical drug therapy, thus going beyond the state of the art. If successful, the project will not only improve the prevention and treatment of cardiovascular disease, but it will open new horizons to individualizing drug therapies.
Summary
Background: Statins are essential drugs in the treatment of hypercholesterolaemia and are among the most prescribed drugs worldwide. The response to statin therapy varies widely between individuals. While most patients show good efficacy, a significant proportion of individuals show poor or even a lack of cholesterol-lowering efficacy. Moreover, a number of patients experience adverse drug reactions. These together with the lack of immediate effect on well-being likely explain the relatively poor adherence to statin therapy. Poor adherence to statins in turn increases the incidence of cardiovascular events and mortality.
Aims: The objectives of this project are 1) to develop a systems pharmacology model for predicting statin efficacy and tolerability at the level of an individual patient and 2) to investigate whether selecting the statin based on the model improves treatment adherence.
Methods: A systems pharmacology approach will be used to integrate data from in vitro and clinical studies. Semi-physiological pharmacokinetic-dynamic-toxicologic models will be built for each statin allowing the prediction of the pharmacokinetic and clinical outcomes for patients with different characteristics, genotypes, and concomitant medications. The ability of the systems pharmacology algorithm to enhance adherence will be investigated in a randomized clinical trial.
Significance: Systems pharmacology models have been increasingly applied in drug development, for example to predict the effect of organ dysfunction on pharmacokinetics. The proposed project is the first to use systems pharmacology predictions to guide clinical drug therapy, thus going beyond the state of the art. If successful, the project will not only improve the prevention and treatment of cardiovascular disease, but it will open new horizons to individualizing drug therapies.
Max ERC Funding
2 211 565 €
Duration
Start date: 2017-08-01, End date: 2022-07-31
Project acronym InflammaTENSION
Project A study of the roles of the immune and inflammatory systems in hypertension.
Researcher (PI) Tomasz Jan GUZIK
Host Institution (HI) UNIVERSITY OF GLASGOW
Call Details Consolidator Grant (CoG), LS7, ERC-2016-COG
Summary Hypertension is a common disease impacting 1 billion people worldwide, which leads to catastrophic cardiovascular complications. The cause of primary hypertension is unknown and the disease remains uncontrolled in many patients. By interrogating the key hypothesis that inflammatory dysregulation fundamentally controls development of hypertension and vascular remodelling, InflammaTENSION provides a new paradigm for the management of the disease, with the potential to lead to identification of novel therapeutic targets to control hypertension. InflammaTENSION will result in the discovery of novel biomarkers, capable of identifying patients who could benefit from such immune targeted therapies. Importantly, we already made the seminal observation that the immune system not only mediates target organ damage, but is essential for the development of hypertension. This finding has initiated numerous studies, that defined the roles of pro-inflammatory T cells, monocytes and anti-inflammatory T regulatory cells. However, our current knowledge remains very fragmented and so far has not been applied to human pathology. InflammaTENSION will for the first time advance the knowledge procured in rodent models into human studies. By combining clinical translational and model mechanistic studies it will identify novel inflammatory factors that can control immune mechanisms of hypertension. We will: (1) characterize the immunophenotypic signature of human hypertension; (2) define key concepts in cytokine biology of hypertension with TNF-α and IL-6 as key exemplars; (3) understand how chronic cytokines regulate the T cell dependent mechanisms of hypertension. InflammaTENSION will go beyond current state-of-the-art through comprehensive combination of immunology and cardiovascular medicine to create a new understanding of how the immune system may lead to human hypertension and will have major impact on the field, enabling translation of these exciting findings to clinical practice.
Summary
Hypertension is a common disease impacting 1 billion people worldwide, which leads to catastrophic cardiovascular complications. The cause of primary hypertension is unknown and the disease remains uncontrolled in many patients. By interrogating the key hypothesis that inflammatory dysregulation fundamentally controls development of hypertension and vascular remodelling, InflammaTENSION provides a new paradigm for the management of the disease, with the potential to lead to identification of novel therapeutic targets to control hypertension. InflammaTENSION will result in the discovery of novel biomarkers, capable of identifying patients who could benefit from such immune targeted therapies. Importantly, we already made the seminal observation that the immune system not only mediates target organ damage, but is essential for the development of hypertension. This finding has initiated numerous studies, that defined the roles of pro-inflammatory T cells, monocytes and anti-inflammatory T regulatory cells. However, our current knowledge remains very fragmented and so far has not been applied to human pathology. InflammaTENSION will for the first time advance the knowledge procured in rodent models into human studies. By combining clinical translational and model mechanistic studies it will identify novel inflammatory factors that can control immune mechanisms of hypertension. We will: (1) characterize the immunophenotypic signature of human hypertension; (2) define key concepts in cytokine biology of hypertension with TNF-α and IL-6 as key exemplars; (3) understand how chronic cytokines regulate the T cell dependent mechanisms of hypertension. InflammaTENSION will go beyond current state-of-the-art through comprehensive combination of immunology and cardiovascular medicine to create a new understanding of how the immune system may lead to human hypertension and will have major impact on the field, enabling translation of these exciting findings to clinical practice.
Max ERC Funding
1 957 574 €
Duration
Start date: 2017-05-01, End date: 2022-04-30
Project acronym INSTANTCOUNT
Project Printed microfluidic counting chambers for low-cost point-of-care diagnostics
Researcher (PI) Markus Beck
Host Institution (HI) UNIVERSITEIT TWENTE
Call Details Starting Grant (StG), LS7, ERC-2011-StG_20101109
Summary "The goal of ""InstantCount"" is to develop low-cost point-of-care blood tests based on microfluidic counting chambers and image cytometry (InstantCount tests) for use in challenging environments, e.g. remote areas in developing countries.
Preliminary work on CD4 counting (measuring the concentration of helper T-cells in blood in order to diagnose the progression of an HIV infection) demonstrates the feasibility of the concept.
I have assembled a prototype instrument for large area fluorescence imaging, developed the image analysis software and microfluidic counting chambers containing dried reagents. The fabrication of these chambers is simple and the used materials cheap, making affordable diagnostics even in the poorest regions appear feasible.
However, a clinical evaluation of the tests outside the laboratory environment in rural areas requires improved reproducibility in the fabrication of the chambers, which would also enable the development of new assays, once the properties of the reagent layers can be tailored according to the requirements of different tests.
I propose the development of printing techniques for the fabrication of InstantCount tests. Counting chambers with precisely defined thickness will be realised by printing of microbeads embedded in glue. Printing of different types of hydrogel will offer solutions like self-sealing chambers or timed release of reagents with a wide range of molecular sizes without the need for mixing other than by diffusion.
With these techniques, we will realise rapid low-cost point-of-care diagnostics for primary health care in remote areas and small medical practices. Complete blood counts for general health screening, CD4 counting and the detection of parasites in red blood cells such as in malaria will be provided.
The project will neither require expensive fabrication methods in clean room facilities nor high-resolution optics in the instruments.
Printing can bridge the gap between prototyping and production."
Summary
"The goal of ""InstantCount"" is to develop low-cost point-of-care blood tests based on microfluidic counting chambers and image cytometry (InstantCount tests) for use in challenging environments, e.g. remote areas in developing countries.
Preliminary work on CD4 counting (measuring the concentration of helper T-cells in blood in order to diagnose the progression of an HIV infection) demonstrates the feasibility of the concept.
I have assembled a prototype instrument for large area fluorescence imaging, developed the image analysis software and microfluidic counting chambers containing dried reagents. The fabrication of these chambers is simple and the used materials cheap, making affordable diagnostics even in the poorest regions appear feasible.
However, a clinical evaluation of the tests outside the laboratory environment in rural areas requires improved reproducibility in the fabrication of the chambers, which would also enable the development of new assays, once the properties of the reagent layers can be tailored according to the requirements of different tests.
I propose the development of printing techniques for the fabrication of InstantCount tests. Counting chambers with precisely defined thickness will be realised by printing of microbeads embedded in glue. Printing of different types of hydrogel will offer solutions like self-sealing chambers or timed release of reagents with a wide range of molecular sizes without the need for mixing other than by diffusion.
With these techniques, we will realise rapid low-cost point-of-care diagnostics for primary health care in remote areas and small medical practices. Complete blood counts for general health screening, CD4 counting and the detection of parasites in red blood cells such as in malaria will be provided.
The project will neither require expensive fabrication methods in clean room facilities nor high-resolution optics in the instruments.
Printing can bridge the gap between prototyping and production."
Max ERC Funding
1 496 400 €
Duration
Start date: 2011-09-01, End date: 2017-04-30
Project acronym LightTouch
Project Feeling with Light – Development of a multimodal optofluidic platform for high-content blood cell analysis
Researcher (PI) Jochen Guck
Host Institution (HI) TECHNISCHE UNIVERSITAET DRESDEN
Call Details Starting Grant (StG), LS7, ERC-2011-StG_20101109
Summary The reliable characterization of heterogeneous cell populations is a central prerequisite in many areas of medicine, biology, and biotechnology. Conventional techniques used for this purpose are either high-throughput, such as fluorescence-activated cell sorting (FACS), but limited to a small number of parameters or high-content, such as slide-based imaging approaches, which require surface attachment and preclude cell sorting. It is the overall objective of this proposal to develop a multimodal, microfluidic, laser trap-assisted cell screening platform technology – µFLAX – for the contact-free manipulation and high-content screening of suspended cells with high-throughput. Our approach is especially designed for blood cells, which will be serially trapped from microfluidic flow with a dual-beam trap. In addition to the extraction of molecular information (similar to FACS), we also incorporate mechanical phenotyping as a powerful new functional cell marker. In addition to looking, we can feel for functional changes using optically induced forces. This will be further augmented by structural analysis using digital holographic microscopy, cell size analysis using an optical cell rotator, and biological stimulation by microfluidic delivery of biochemical agents. This combination will offer much higher sensitivity and specificity in determining functional states compared to currently available techniques. And since the cells are suspended they can be sorted and analyzed further, which will aid potential molecular target identification. Once developed, we will demonstrate the applicability of µFLAX for the investigation and diagnosis of inflammation and infection. Based on our track record in pioneering innovative physical solutions to biomedical problems we anticipate that through this project we will provide novel insight into system biological aspects of these disorders on the single-cell level, as well as novel diagnostic and therapeutic options.
Summary
The reliable characterization of heterogeneous cell populations is a central prerequisite in many areas of medicine, biology, and biotechnology. Conventional techniques used for this purpose are either high-throughput, such as fluorescence-activated cell sorting (FACS), but limited to a small number of parameters or high-content, such as slide-based imaging approaches, which require surface attachment and preclude cell sorting. It is the overall objective of this proposal to develop a multimodal, microfluidic, laser trap-assisted cell screening platform technology – µFLAX – for the contact-free manipulation and high-content screening of suspended cells with high-throughput. Our approach is especially designed for blood cells, which will be serially trapped from microfluidic flow with a dual-beam trap. In addition to the extraction of molecular information (similar to FACS), we also incorporate mechanical phenotyping as a powerful new functional cell marker. In addition to looking, we can feel for functional changes using optically induced forces. This will be further augmented by structural analysis using digital holographic microscopy, cell size analysis using an optical cell rotator, and biological stimulation by microfluidic delivery of biochemical agents. This combination will offer much higher sensitivity and specificity in determining functional states compared to currently available techniques. And since the cells are suspended they can be sorted and analyzed further, which will aid potential molecular target identification. Once developed, we will demonstrate the applicability of µFLAX for the investigation and diagnosis of inflammation and infection. Based on our track record in pioneering innovative physical solutions to biomedical problems we anticipate that through this project we will provide novel insight into system biological aspects of these disorders on the single-cell level, as well as novel diagnostic and therapeutic options.
Max ERC Funding
1 385 683 €
Duration
Start date: 2012-02-01, End date: 2017-01-31
Project acronym LiverFibrosisImaging
Project Quantitative Imaging of Liver Fibrosis and Fibrogenesis
Researcher (PI) Detlef Schuppan
Host Institution (HI) UNIVERSITAETSMEDIZIN DER JOHANNES GUTENBERG-UNIVERSITAET MAINZ
Call Details Advanced Grant (AdG), LS7, ERC-2011-ADG_20110310
Summary Chronic liver disease can progress to cirrhosis, with death due to liver failure and cancer. Cirrhosis prevalence in the EU is 0.5%-1%. However, development of therapies that prevent progression to cirrhosis is hampered by the lack of a sensitive, non-invasive method to quantify fibrosis or fibrosis progression (fibrogenesis). Liver biopsy 1) is risky, 2) shows high sampling variability, and 3) is too insensitive to assess fibrosis progression in clinical studies. Conventional radiological imaging, serum markers, and ultrasound- or MR-elastography do neither permit exact fibrosis nor any fibrogenesis measurement.
We plan to develop a clinically applicable methodology to quantitate fibrosis and fibrogenesis over the whole liver using imaging agents that target and thus quantify abundant fibrillar collagen or key cells that drive fibrogenesis (activated myofibroblasts and cholangiocytes). We demonstrated the feasibility of this approach using radiolabeled conjugates of high affinity that target integrin alphaVbeta6 and PDGFbeta receptor that are cell surface molecules of activated cholangiocytes and myofibroblasts. i.v. injection of the integrin conjugate allowed quantitative imaging of alphaVbeta6 expression and correlated with whole liver fibrogenesis. We intend to optimize nonpeptide and peptide ligands for integrin alphaVbeta6, PDGF beta receptor and fibrillar collagens using novel linkers and oligomerization, using PET-radioimaging with Ga-68, Sc-44 and F-18. The targeted imaging constructs will be validated in vivo using established rodent models with defined liver fibrosis and fibrogenesis, with and without antifibrotic drug therapy. Translation to phase I and II clinical studies is planned in years 4-5 of the project.
The technology will for the first time allow for 1. individual risk assessment of fibrosis progression, and 2. rapid testing of antifibrotic drugs and their combinations in small groups of individual patients.
Summary
Chronic liver disease can progress to cirrhosis, with death due to liver failure and cancer. Cirrhosis prevalence in the EU is 0.5%-1%. However, development of therapies that prevent progression to cirrhosis is hampered by the lack of a sensitive, non-invasive method to quantify fibrosis or fibrosis progression (fibrogenesis). Liver biopsy 1) is risky, 2) shows high sampling variability, and 3) is too insensitive to assess fibrosis progression in clinical studies. Conventional radiological imaging, serum markers, and ultrasound- or MR-elastography do neither permit exact fibrosis nor any fibrogenesis measurement.
We plan to develop a clinically applicable methodology to quantitate fibrosis and fibrogenesis over the whole liver using imaging agents that target and thus quantify abundant fibrillar collagen or key cells that drive fibrogenesis (activated myofibroblasts and cholangiocytes). We demonstrated the feasibility of this approach using radiolabeled conjugates of high affinity that target integrin alphaVbeta6 and PDGFbeta receptor that are cell surface molecules of activated cholangiocytes and myofibroblasts. i.v. injection of the integrin conjugate allowed quantitative imaging of alphaVbeta6 expression and correlated with whole liver fibrogenesis. We intend to optimize nonpeptide and peptide ligands for integrin alphaVbeta6, PDGF beta receptor and fibrillar collagens using novel linkers and oligomerization, using PET-radioimaging with Ga-68, Sc-44 and F-18. The targeted imaging constructs will be validated in vivo using established rodent models with defined liver fibrosis and fibrogenesis, with and without antifibrotic drug therapy. Translation to phase I and II clinical studies is planned in years 4-5 of the project.
The technology will for the first time allow for 1. individual risk assessment of fibrosis progression, and 2. rapid testing of antifibrotic drugs and their combinations in small groups of individual patients.
Max ERC Funding
2 454 604 €
Duration
Start date: 2012-08-01, End date: 2017-07-31
Project acronym LUPUSCARE
Project PRECISION CARE IN SYSTEMIC AUTOIMMUNITY: AN INTEGRATED MULTI-TISSUE/LEVEL APPROACH FOR SYSTEMIC LUPUS ERYTHEMATOSUS (SLE)
Researcher (PI) DIMITRIOS BOUMPAS
Host Institution (HI) IDRYMA IATROVIOLOGIKON EREUNON AKADEMIAS ATHINON
Call Details Advanced Grant (AdG), LS7, ERC-2016-ADG
Summary Systemic lupus erythematosus (SLE) is a heterogeneous disease whereby an interplay of environmental, genetic and epigenetic factors lead to perturbation of complex biological networks culminating into diverse clinical phenotypes of varying severity. High throughput methods have allowed an “initial glimpse” into pathogenesis and have laid the foundations for a molecular-based taxonomy for personalized therapy. Based on our experience with the molecular characterization of SLE, a recently completed RNA sequencing analysis of 150 patients, and our track- record of “paradigm shift” trials in SLE, we will integrate data from multi-tissue analyses with novel technologies to improve its diagnosis, monitoring and therapy, and ask fundamental pathogenetic questions in systemic autoimmunity. More specifically, we will design gene expression panels and “expression profile”/”clinical trait” correlation matrices for diagnostics, personalized immunotherapy and improved clinical trial design. In a systematic multi-tissue approach, we will examine the role of somatic mutations in enhancing immune hyperactivity and the risk for lymphoma. The staggering (7-9:1) female predominance will be elucidated through elaborate genomic, epigenomic and microbiota analyses of family trios. Finally, we will be pursuing the innovative hypothesis that the fundamental abnormalities of SLE lie within the bone marrow hematopoietic stem cells (HSCs) - from which all cells that participate in the pathogenesis of SLE originate - and establish it as a unifying pathogenetic mechanism. By a combination of novel experimental analyses with single cell genomics, multi–omics, humanized animal models, genome editing and an “organ on-a-chip” device, we will validate HSCs as a therapeutic target. The utility of SLE research extends beyond its boundaries, by providing unique insights as to how the immune system recognizes self-constituents and maintains its homeostasis, and how gender impacts on disease biology.
Summary
Systemic lupus erythematosus (SLE) is a heterogeneous disease whereby an interplay of environmental, genetic and epigenetic factors lead to perturbation of complex biological networks culminating into diverse clinical phenotypes of varying severity. High throughput methods have allowed an “initial glimpse” into pathogenesis and have laid the foundations for a molecular-based taxonomy for personalized therapy. Based on our experience with the molecular characterization of SLE, a recently completed RNA sequencing analysis of 150 patients, and our track- record of “paradigm shift” trials in SLE, we will integrate data from multi-tissue analyses with novel technologies to improve its diagnosis, monitoring and therapy, and ask fundamental pathogenetic questions in systemic autoimmunity. More specifically, we will design gene expression panels and “expression profile”/”clinical trait” correlation matrices for diagnostics, personalized immunotherapy and improved clinical trial design. In a systematic multi-tissue approach, we will examine the role of somatic mutations in enhancing immune hyperactivity and the risk for lymphoma. The staggering (7-9:1) female predominance will be elucidated through elaborate genomic, epigenomic and microbiota analyses of family trios. Finally, we will be pursuing the innovative hypothesis that the fundamental abnormalities of SLE lie within the bone marrow hematopoietic stem cells (HSCs) - from which all cells that participate in the pathogenesis of SLE originate - and establish it as a unifying pathogenetic mechanism. By a combination of novel experimental analyses with single cell genomics, multi–omics, humanized animal models, genome editing and an “organ on-a-chip” device, we will validate HSCs as a therapeutic target. The utility of SLE research extends beyond its boundaries, by providing unique insights as to how the immune system recognizes self-constituents and maintains its homeostasis, and how gender impacts on disease biology.
Max ERC Funding
2 355 000 €
Duration
Start date: 2017-09-01, End date: 2022-08-31
Project acronym MENINGENE
Project Nationwide prospective study on community-acquired bacterial meningitis: from genetics to therapy
Researcher (PI) Diederik Van De Beek
Host Institution (HI) ACADEMISCH MEDISCH CENTRUM BIJ DE UNIVERSITEIT VAN AMSTERDAM
Call Details Starting Grant (StG), LS7, ERC-2011-StG_20101109
Summary Meningitis is an inflammation of the membranes covering the brain and spinal cord (the meninges). Each year 35,000 European patients suffer from bacterial meningitis, resulting in 7000 deaths and leaving 7000 disabled. Streptococcus pneumoniae and Neisseria meningitidis are the most common causative bacteria of bacterial meningitis. These bacteria are common colonizers, but in some individuals, they are able to spread to the bloodstream, slip through the blood-brain barrier and cause meningitis, with devastating consequences. Genetics of host and pathogen are considered crucial in this host-pathogen interaction. Our objectives are to identify and characterize host genetic traits and bacterial virulence factors controlling occurrence and outcome of bacterial meningitis. In a nationwide prospective cohort study, using pathway and total exome sequencing, we will search for genetic variants in 2000 patients with community-acquired bacterial meningitis and 2000 controls. On the pathogen side, using whole genome sequencing, we will systematically search for new bacterial virulence factors in the 2000 causative bacteria. Subsequently, by analyzing clinical data, serum and cerebrospinal fluid, we will examine the potential impact and functionality of these host and pathogen factors. Next, we will validate and explore our findings in an animal model of meningitis and investigate whether treatment against these specific components can improve outcome. Finally, we will initiate an open-access European database on the genetics in bacterial meningitis. In addressing our objectives, I will combine my expertise in bacterial meningitis with groundbreaking, translational approaches using clinical data, human samples, next generation sequencing, in vitro techniques, and a mouse model. The results of this project will be translated into novel therapeutic strategies to advance patient care and will also be important for the development of disease prevention and vaccines.
Summary
Meningitis is an inflammation of the membranes covering the brain and spinal cord (the meninges). Each year 35,000 European patients suffer from bacterial meningitis, resulting in 7000 deaths and leaving 7000 disabled. Streptococcus pneumoniae and Neisseria meningitidis are the most common causative bacteria of bacterial meningitis. These bacteria are common colonizers, but in some individuals, they are able to spread to the bloodstream, slip through the blood-brain barrier and cause meningitis, with devastating consequences. Genetics of host and pathogen are considered crucial in this host-pathogen interaction. Our objectives are to identify and characterize host genetic traits and bacterial virulence factors controlling occurrence and outcome of bacterial meningitis. In a nationwide prospective cohort study, using pathway and total exome sequencing, we will search for genetic variants in 2000 patients with community-acquired bacterial meningitis and 2000 controls. On the pathogen side, using whole genome sequencing, we will systematically search for new bacterial virulence factors in the 2000 causative bacteria. Subsequently, by analyzing clinical data, serum and cerebrospinal fluid, we will examine the potential impact and functionality of these host and pathogen factors. Next, we will validate and explore our findings in an animal model of meningitis and investigate whether treatment against these specific components can improve outcome. Finally, we will initiate an open-access European database on the genetics in bacterial meningitis. In addressing our objectives, I will combine my expertise in bacterial meningitis with groundbreaking, translational approaches using clinical data, human samples, next generation sequencing, in vitro techniques, and a mouse model. The results of this project will be translated into novel therapeutic strategies to advance patient care and will also be important for the development of disease prevention and vaccines.
Max ERC Funding
1 400 000 €
Duration
Start date: 2012-02-01, End date: 2017-01-31
Project acronym MGUS screening RCT
Project Screening for monoclonal gammopathy of undetermined significance: A population-based randomized clinical trial
Researcher (PI) Sigurdur Yngvi KRISTINSSON
Host Institution (HI) HASKOLI ISLANDS
Call Details Starting Grant (StG), LS7, ERC-2016-STG
Summary Monoclonal gammopathy of undetermined significance (MGUS) is a very common precursor condition to multiple myeloma (MM), and related diseases, and can be found in approximately 4-5% of individuals over the age of 50 years. MM is always preceded by MGUS. Current risk stratification schemes, designed to predict those that will progress, are based on retrospective data and rely almost solely on serum protein markers. While they can differentiate high and low-risk patients, they cannot predict outcome for individual patients, are not integrated with one another, and have limited biological correlation. Based on retrospective data, it is recommended that individuals with MGUS are followed indefinitely; however no prospective study has ever been performed to evaluate this or identify optimal monitoring in MGUS individuals. We recently showed that MM patients with a prior knowledge of MGUS had superior survival compared to MM patients without, which raises the question whether routine screening for MGUS in the population might improve survival. To evaluate the impact of screening for MGUS on overall survival, to provide evidence for the optimal MGUS follow-up, and to integrate biological, imaging, and germline genetic markers in evaluating individual risk of progression, we propose to invite all individuals >50 years in Iceland (N=104,000) to participate in a screening study for MGUS. This will be done by utilizing already present infrastructure for screening in Iceland and the fact that most individuals >50 years have their blood drawn for various reasons during 3 years. We plan to perform electrophoresis and free light chain analyses in these individuals to diagnose MGUS. Individuals with MGUS will be invited to be included in a randomized clinical trial with 3 different arms to identify the optimal work-up and follow-up strategy and to build a new risk model for progression. Our large, unique, population-based study has major clinical and scientific implications.
Summary
Monoclonal gammopathy of undetermined significance (MGUS) is a very common precursor condition to multiple myeloma (MM), and related diseases, and can be found in approximately 4-5% of individuals over the age of 50 years. MM is always preceded by MGUS. Current risk stratification schemes, designed to predict those that will progress, are based on retrospective data and rely almost solely on serum protein markers. While they can differentiate high and low-risk patients, they cannot predict outcome for individual patients, are not integrated with one another, and have limited biological correlation. Based on retrospective data, it is recommended that individuals with MGUS are followed indefinitely; however no prospective study has ever been performed to evaluate this or identify optimal monitoring in MGUS individuals. We recently showed that MM patients with a prior knowledge of MGUS had superior survival compared to MM patients without, which raises the question whether routine screening for MGUS in the population might improve survival. To evaluate the impact of screening for MGUS on overall survival, to provide evidence for the optimal MGUS follow-up, and to integrate biological, imaging, and germline genetic markers in evaluating individual risk of progression, we propose to invite all individuals >50 years in Iceland (N=104,000) to participate in a screening study for MGUS. This will be done by utilizing already present infrastructure for screening in Iceland and the fact that most individuals >50 years have their blood drawn for various reasons during 3 years. We plan to perform electrophoresis and free light chain analyses in these individuals to diagnose MGUS. Individuals with MGUS will be invited to be included in a randomized clinical trial with 3 different arms to identify the optimal work-up and follow-up strategy and to build a new risk model for progression. Our large, unique, population-based study has major clinical and scientific implications.
Max ERC Funding
1 474 304 €
Duration
Start date: 2017-02-01, End date: 2022-01-31
Project acronym MOPIT
Project Molecular photoacoustic imaging of stem-cell driven tissue regeneration
Researcher (PI) Gottfried Jan Laufer
Host Institution (HI) TECHNISCHE UNIVERSITAT BERLIN
Call Details Starting Grant (StG), LS7, ERC-2011-StG_20101109
Summary "I propose to develop a new generation of molecular photoacoustic imaging technologies and methods capable of detecting single deep tissue cells in preclinical studies of tissue regeneration. In order to achieve this goal, an interdisciplinary research programme involving physicists, engineers and life scientists is required to address the following objectives: 1) the development of novel photoacoustic imaging technology that provides high sensitivity and acquisition speed, 2) the development of the theoretical framework and experimental methods for quantitative imaging, 3) the development of novel genetically expressed reporters, and 4) the preclinical application in small animal models of tissue regeneration. This will result in a preclinical imaging modality has the potential to combine single cell sensitivity and microscale spatial resolution in deep (centimetre range) tissue regions with molecular, physiological, and anatomical imaging capabilities. The instrumentation and methodologies developed in this project will be applied to noninvasive, longitudinal, and quantitative studies of stem cell driven tissue regeneration, such as angiogenesis in bone fractures and muscle trauma. It will allow the detection and tracking of single stem cells and the probing of stem cell function. This will provide unprecedented opportunities for correlating cellular localization, migration, and function and with anatomical changes - knowledge that can be exploited to develop novel drugs and cell-based clinical therapies. Crucially, the technologies and methodologies developed in this project will be directly applicable to a wide range of other fields of the life sciences, such as cancer research and neurology."
Summary
"I propose to develop a new generation of molecular photoacoustic imaging technologies and methods capable of detecting single deep tissue cells in preclinical studies of tissue regeneration. In order to achieve this goal, an interdisciplinary research programme involving physicists, engineers and life scientists is required to address the following objectives: 1) the development of novel photoacoustic imaging technology that provides high sensitivity and acquisition speed, 2) the development of the theoretical framework and experimental methods for quantitative imaging, 3) the development of novel genetically expressed reporters, and 4) the preclinical application in small animal models of tissue regeneration. This will result in a preclinical imaging modality has the potential to combine single cell sensitivity and microscale spatial resolution in deep (centimetre range) tissue regions with molecular, physiological, and anatomical imaging capabilities. The instrumentation and methodologies developed in this project will be applied to noninvasive, longitudinal, and quantitative studies of stem cell driven tissue regeneration, such as angiogenesis in bone fractures and muscle trauma. It will allow the detection and tracking of single stem cells and the probing of stem cell function. This will provide unprecedented opportunities for correlating cellular localization, migration, and function and with anatomical changes - knowledge that can be exploited to develop novel drugs and cell-based clinical therapies. Crucially, the technologies and methodologies developed in this project will be directly applicable to a wide range of other fields of the life sciences, such as cancer research and neurology."
Max ERC Funding
1 622 736 €
Duration
Start date: 2012-01-01, End date: 2016-12-31
Project acronym MRexcite
Project Unlocking the potential of ultra-high-field MRI through manipulation of radiofrequency excitation fields in human tissue
Researcher (PI) Mark Edward Ladd
Host Institution (HI) DEUTSCHES KREBSFORSCHUNGSZENTRUM HEIDELBERG
Call Details Advanced Grant (AdG), LS7, ERC-2011-ADG_20110310
Summary In the past three decades, magnetic resonance imaging (MRI) has become a vital tool for clinical diagnosis and research. A major current trend is the introduction of magnets with much more powerful static magnetic fields, including magnets at 7 Tesla (7T) and higher. Advantages of higher magnetic fields include higher signal-to-noise ratios enabling improved spatial and temporal resolution, and new, unique tissue contrasts due to enhanced sensitivity to tissue susceptibility differences.
Unfortunately, the radiofrequency (RF) fields used to excite tissue at higher magnetic fields are subject to interference and penetration effects, leading to signal dropouts which vary from subject to subject depending on body habitus. These effects imply that the inherent advantages of 7T often cannot be leveraged to realise practical imaging benefits. A fair evaluation of the diagnostic potential of 7T cannot be achieved, as image quality improvements are handicapped and often counteracted by these unresolved technical hurdles. 7T MRI cannot be considered for routine clinical use or even effectively evaluated for such use until these hurdles have been overcome.
Preliminary research indicates that these effects can be addressed by use of parallel transmission strategies. The goal of the proposed project is to develop a highly optimized multi-channel transmit/receive RF coil for body MRI at 7T. This coil should then be used to exploit and manipulate the complex RF field patterns at 7T using parallel transmission approaches. In contrast to previous approaches, a hybrid method including both static and dynamic shimming of the RF field will be investigated. We hypothesise that such an approach would greatly enhance the flexibility of RF manipulation while limiting overall system complexity. It can be conjectured based on the known properties of ultra-high-field MRI that success would have ground-breaking impact on the diagnosis and characterisation of manifold disease processes.
Summary
In the past three decades, magnetic resonance imaging (MRI) has become a vital tool for clinical diagnosis and research. A major current trend is the introduction of magnets with much more powerful static magnetic fields, including magnets at 7 Tesla (7T) and higher. Advantages of higher magnetic fields include higher signal-to-noise ratios enabling improved spatial and temporal resolution, and new, unique tissue contrasts due to enhanced sensitivity to tissue susceptibility differences.
Unfortunately, the radiofrequency (RF) fields used to excite tissue at higher magnetic fields are subject to interference and penetration effects, leading to signal dropouts which vary from subject to subject depending on body habitus. These effects imply that the inherent advantages of 7T often cannot be leveraged to realise practical imaging benefits. A fair evaluation of the diagnostic potential of 7T cannot be achieved, as image quality improvements are handicapped and often counteracted by these unresolved technical hurdles. 7T MRI cannot be considered for routine clinical use or even effectively evaluated for such use until these hurdles have been overcome.
Preliminary research indicates that these effects can be addressed by use of parallel transmission strategies. The goal of the proposed project is to develop a highly optimized multi-channel transmit/receive RF coil for body MRI at 7T. This coil should then be used to exploit and manipulate the complex RF field patterns at 7T using parallel transmission approaches. In contrast to previous approaches, a hybrid method including both static and dynamic shimming of the RF field will be investigated. We hypothesise that such an approach would greatly enhance the flexibility of RF manipulation while limiting overall system complexity. It can be conjectured based on the known properties of ultra-high-field MRI that success would have ground-breaking impact on the diagnosis and characterisation of manifold disease processes.
Max ERC Funding
2 099 996 €
Duration
Start date: 2012-05-01, End date: 2017-04-30
Project acronym MULTIEPIGEN
Project Ancestral environmental exposures and offspring health – a multigenerational epidemiologic cohort study across 3 generations
Researcher (PI) Olli RAITAKARI
Host Institution (HI) TURUN YLIOPISTO
Call Details Advanced Grant (AdG), LS7, ERC-2016-ADG
Summary MULTIEPIGEN seeks to solve does ancestral exposure to various stressors transmit to offspring via epigenetic mechanisms. Thus far animal models have indicated that exposure to certain stressors can lead to phenotypic changes not only in the predisposed individuals, but also in the future generations, such that individuals can acquire phenotypes caused by exposures of their ancestors. Such effects do not involve new DNA mutations, but are transmitted to offspring via epigenetic mechanisms such as the transfer of non-coding RNA molecules in the semen. In humans, intergenerational transmission has been examined extremely little because a priori designed population-based studies across several generations are lacking. To close this gap MULTIEPIGEN will expand the well-characterized Cardiovascular Risk in Young Finns Study (YFS) to the parents and offspring of the original YFS participants. During the ERC funding period, we will perform field studies involving N~9000 individuals across 3 generations and test 3 key ancestral exposures with very high plausibility causing intergenerational effects on obesity-related phenotypes, cognitive function and psychological well-being. The studied exposures are 1) tobacco smoke, 2) persistent organic pollutants, and 3) accumulation of psychosocial adversities. We will collect serum, blood and semen samples for epigenetic marker analysis to provide understanding of the mechanisms of intergenerational transmission in humans. Specifically, we will seek proof for the hypothesis that paternal stressors can lead to phenotypic changes in the offspring via non-coding RNA molecules in the semen. Multigenerational epidemiologic data showing robust links between ancestral exposures and offspring phenotypes that operate with biologically plausible epigenetic mechanism would provide a conceptual change in the developmental biology in humans and have substantial ramifications on public health.
Summary
MULTIEPIGEN seeks to solve does ancestral exposure to various stressors transmit to offspring via epigenetic mechanisms. Thus far animal models have indicated that exposure to certain stressors can lead to phenotypic changes not only in the predisposed individuals, but also in the future generations, such that individuals can acquire phenotypes caused by exposures of their ancestors. Such effects do not involve new DNA mutations, but are transmitted to offspring via epigenetic mechanisms such as the transfer of non-coding RNA molecules in the semen. In humans, intergenerational transmission has been examined extremely little because a priori designed population-based studies across several generations are lacking. To close this gap MULTIEPIGEN will expand the well-characterized Cardiovascular Risk in Young Finns Study (YFS) to the parents and offspring of the original YFS participants. During the ERC funding period, we will perform field studies involving N~9000 individuals across 3 generations and test 3 key ancestral exposures with very high plausibility causing intergenerational effects on obesity-related phenotypes, cognitive function and psychological well-being. The studied exposures are 1) tobacco smoke, 2) persistent organic pollutants, and 3) accumulation of psychosocial adversities. We will collect serum, blood and semen samples for epigenetic marker analysis to provide understanding of the mechanisms of intergenerational transmission in humans. Specifically, we will seek proof for the hypothesis that paternal stressors can lead to phenotypic changes in the offspring via non-coding RNA molecules in the semen. Multigenerational epidemiologic data showing robust links between ancestral exposures and offspring phenotypes that operate with biologically plausible epigenetic mechanism would provide a conceptual change in the developmental biology in humans and have substantial ramifications on public health.
Max ERC Funding
2 498 606 €
Duration
Start date: 2017-11-01, End date: 2022-10-31
Project acronym MUMI
Project Multimodal Molecular Imaging
Researcher (PI) Carl Markus Maximilian Schwaiger
Host Institution (HI) KLINIKUM RECHTS DER ISAR DER TECHNISCHEN UNIVERSITAT MUNCHEN
Call Details Advanced Grant (AdG), LS7, ERC-2011-ADG_20110310
Summary "Imaging has become an integral part of diagnosis and staging of disease. This proposal aims at the development if innovative imaging methods for the visualization and quantitative assessment of biologic processes in-vivo. The simultaneous MR/PET data acquisition will be employed o integrate structural, physiologic and molecular information for the phenotyping of human disease and therapeutic decision making process. Such multimodality imaging is thought to serve as unique clinical tool to support the realization of personalized medicine."
Summary
"Imaging has become an integral part of diagnosis and staging of disease. This proposal aims at the development if innovative imaging methods for the visualization and quantitative assessment of biologic processes in-vivo. The simultaneous MR/PET data acquisition will be employed o integrate structural, physiologic and molecular information for the phenotyping of human disease and therapeutic decision making process. Such multimodality imaging is thought to serve as unique clinical tool to support the realization of personalized medicine."
Max ERC Funding
2 238 616 €
Duration
Start date: 2012-06-01, End date: 2017-05-31
Project acronym NeuroTRACK
Project Tracking and predicting neurodegeneration spreading across the brain connectome
Researcher (PI) Federica Agosta
Host Institution (HI) OSPEDALE SAN RAFFAELE SRL
Call Details Starting Grant (StG), LS7, ERC-2016-STG
Summary Current knowledge of neurodegenerative diseases is limited by poor understanding of how they progress through the central nervous system (CNS). It has recently been hypothesized that clinical progression in these conditions involves the systematic spreading of protein misfolding along neuronal pathways. Protein aggregates would trigger misfolding of adjacent homologue proteins in newly-affected regions, and this would propagate in a “prion-like” fashion across anatomical connections. This proposal seeks to decipher the mechanisms of network-based neurodegeneration by understanding how the complex architecture of brain networks (the connectome) shapes the evolving pathology of neurodegenerative diseases, and to develop tools for monitoring disease progression from presymptomatic to later stages of the disease.
NeuroTRACK will apply emerging network science tools to longitudinal, structural and functional brain connectivity 3T magnetic resonance imaging data from patients with frontotemporal lobar degeneration (FTLD) – a devastating, relentlessly progressive, young onset, neurodegenerative disorder. The study will involve both sporadic and familial cases, including presymptomatic gene mutation carriers. The proposal addresses the following fundamental questions: i) How and where does pathological protein propagation occur in the FTLD phenotypes? ii) Can pathological spreading be predicted from brain connectome fingerprinting? iii) How do different protein abnormalities translate into large-scale network degeneration? iv) How early are brain network changes detectable in the (even presymptomatic) course of the disease?
The ground-breaking nature of the experiments planned in this proposal will pave the way to the development of novel tools for understanding the biological underpinnings of other CNS proteinopathies such as Alzheimer’s disease and Parkinson’s disease, and to identifying individualized, early interventions to modify disease progression.
Summary
Current knowledge of neurodegenerative diseases is limited by poor understanding of how they progress through the central nervous system (CNS). It has recently been hypothesized that clinical progression in these conditions involves the systematic spreading of protein misfolding along neuronal pathways. Protein aggregates would trigger misfolding of adjacent homologue proteins in newly-affected regions, and this would propagate in a “prion-like” fashion across anatomical connections. This proposal seeks to decipher the mechanisms of network-based neurodegeneration by understanding how the complex architecture of brain networks (the connectome) shapes the evolving pathology of neurodegenerative diseases, and to develop tools for monitoring disease progression from presymptomatic to later stages of the disease.
NeuroTRACK will apply emerging network science tools to longitudinal, structural and functional brain connectivity 3T magnetic resonance imaging data from patients with frontotemporal lobar degeneration (FTLD) – a devastating, relentlessly progressive, young onset, neurodegenerative disorder. The study will involve both sporadic and familial cases, including presymptomatic gene mutation carriers. The proposal addresses the following fundamental questions: i) How and where does pathological protein propagation occur in the FTLD phenotypes? ii) Can pathological spreading be predicted from brain connectome fingerprinting? iii) How do different protein abnormalities translate into large-scale network degeneration? iv) How early are brain network changes detectable in the (even presymptomatic) course of the disease?
The ground-breaking nature of the experiments planned in this proposal will pave the way to the development of novel tools for understanding the biological underpinnings of other CNS proteinopathies such as Alzheimer’s disease and Parkinson’s disease, and to identifying individualized, early interventions to modify disease progression.
Max ERC Funding
1 496 994 €
Duration
Start date: 2017-04-01, End date: 2022-03-31
Project acronym New Chol
Project Development of New therapies against cholangiopathies.
Researcher (PI) ludovic VALLIER
Host Institution (HI) THE CHANCELLOR MASTERS AND SCHOLARS OF THE UNIVERSITY OF CAMBRIDGE
Call Details Advanced Grant (AdG), LS7, ERC-2016-ADG
Summary Cholangiopathies represent a diverse group of diseases affecting cholangiocytes which are the main cell type of the biliary tract. These disorders range from inherited (Cystic Fibrosis) and developmental (Alagille Syndrome, Biliary Atresia) to autoimmune (Primary Biliary Cirrhosis), idiopathic (Primary Sclerosing Cholangitis) and drug or toxin induced diseases. Cholangiopathies result in toxic bile accumulation in the liver inducing cell death and ultimately cirrhosis. They carry high morbidity and mortality, accounting for up to a third of chronic liver disorders. Whole liver transplantation remains the main treatment. However, organ transplant requires immunosuppression with significant side effects and an increasing number of patients die while on the transplant list due to the shortage of suitable donors. Finally, the absence of physiologically relevant in vitro systems to model and to study cholangiopathies prevents the development of new therapeutics while cell based therapy approach have been unexplored. Here, we propose to systematically address these challenges by developing a novel and innovative program of translational research focusing on cholangiopathies. We will first investigate the cellular and functional diversity of the biliary tract and its impact on disease by taking advantage of recent developments in single cell transcriptomic analyses. We will then use this basic knowledge to generate and to characterize for the first time a renewable source of cholangiocytes from human induced pluripotent stem cells and from biliary tissue. The resulting cells will be used to model cholangiopathies in vitro and to create a new platform for drug target identification. Finally, we will explore the potential for in vitro generated cholangiocytes to be used in regenerative medicine applications including cell based therapy. Overall this comprehensive program will uniquely path the way for the development of a whole range of new therapies for cholangiopathies.
Summary
Cholangiopathies represent a diverse group of diseases affecting cholangiocytes which are the main cell type of the biliary tract. These disorders range from inherited (Cystic Fibrosis) and developmental (Alagille Syndrome, Biliary Atresia) to autoimmune (Primary Biliary Cirrhosis), idiopathic (Primary Sclerosing Cholangitis) and drug or toxin induced diseases. Cholangiopathies result in toxic bile accumulation in the liver inducing cell death and ultimately cirrhosis. They carry high morbidity and mortality, accounting for up to a third of chronic liver disorders. Whole liver transplantation remains the main treatment. However, organ transplant requires immunosuppression with significant side effects and an increasing number of patients die while on the transplant list due to the shortage of suitable donors. Finally, the absence of physiologically relevant in vitro systems to model and to study cholangiopathies prevents the development of new therapeutics while cell based therapy approach have been unexplored. Here, we propose to systematically address these challenges by developing a novel and innovative program of translational research focusing on cholangiopathies. We will first investigate the cellular and functional diversity of the biliary tract and its impact on disease by taking advantage of recent developments in single cell transcriptomic analyses. We will then use this basic knowledge to generate and to characterize for the first time a renewable source of cholangiocytes from human induced pluripotent stem cells and from biliary tissue. The resulting cells will be used to model cholangiopathies in vitro and to create a new platform for drug target identification. Finally, we will explore the potential for in vitro generated cholangiocytes to be used in regenerative medicine applications including cell based therapy. Overall this comprehensive program will uniquely path the way for the development of a whole range of new therapies for cholangiopathies.
Max ERC Funding
2 499 111 €
Duration
Start date: 2017-12-01, End date: 2022-11-30
Project acronym OAPROGRESS
Project Evaluation of Osteoarthritis Progression in a Patient-Specific Manner using Magnetic Resonance Imaging and Computational Modeling
Researcher (PI) Rami Kristian Korhonen
Host Institution (HI) ITA-SUOMEN YLIOPISTO
Call Details Starting Grant (StG), LS7, ERC-2011-StG_20101109
Summary Background
Osteoarthritis (OA) is one of the most prevalent disorders of the musculoskeletal system. In OA, articular cartilage degenerates and its structure and mechanical properties change, but monitoring or predicting the progression of OA has not been possible. Magnetic resonance imaging (MRI) is a potential tool for the imaging of joint tissues, estimating cartilage structure and diagnostics of OA, whereas joint loading and estimation of stresses/strains within joint tissues necessitates computational modeling. It would be a major breakthrough if one could develop a technique where, based on MRI and computational modeling, prediction and evaluation of OA progression of a patient under a certain loading condition would be possible.
Objectives
1) to combine MRI with computational modeling for the estimation of stresses and possible failure points within human knee joints, and 2) to develop second generation adaptive models of articular cartilage for the prediction of altered tissue structure and composition during OA progression. For the model validation, cartilage structure, composition and biomechanical properties as well as cell responses in situ are characterized. At the end of the project these main aims will be merged 3) to estimate the effect of loading on cartilage degeneration during the progression of OA in a patient-specific manner.
Significance
Combining MRI information of joint tissues with computational modeling, we develop a tool to evaluate the effect of different interventions on stresses in human joints. By combining this tool with an adaptive model that can estimate the effect of loading on cartilage composition and structure, we hope to be able to predict changes in cartilage properties during OA progression in a patient-specific manner several years ahead. This would help in decision making of clinical treatments and interventions (conservative or surgical) for the prevention or further progression of OA.
Summary
Background
Osteoarthritis (OA) is one of the most prevalent disorders of the musculoskeletal system. In OA, articular cartilage degenerates and its structure and mechanical properties change, but monitoring or predicting the progression of OA has not been possible. Magnetic resonance imaging (MRI) is a potential tool for the imaging of joint tissues, estimating cartilage structure and diagnostics of OA, whereas joint loading and estimation of stresses/strains within joint tissues necessitates computational modeling. It would be a major breakthrough if one could develop a technique where, based on MRI and computational modeling, prediction and evaluation of OA progression of a patient under a certain loading condition would be possible.
Objectives
1) to combine MRI with computational modeling for the estimation of stresses and possible failure points within human knee joints, and 2) to develop second generation adaptive models of articular cartilage for the prediction of altered tissue structure and composition during OA progression. For the model validation, cartilage structure, composition and biomechanical properties as well as cell responses in situ are characterized. At the end of the project these main aims will be merged 3) to estimate the effect of loading on cartilage degeneration during the progression of OA in a patient-specific manner.
Significance
Combining MRI information of joint tissues with computational modeling, we develop a tool to evaluate the effect of different interventions on stresses in human joints. By combining this tool with an adaptive model that can estimate the effect of loading on cartilage composition and structure, we hope to be able to predict changes in cartilage properties during OA progression in a patient-specific manner several years ahead. This would help in decision making of clinical treatments and interventions (conservative or surgical) for the prevention or further progression of OA.
Max ERC Funding
1 303 056 €
Duration
Start date: 2012-02-01, End date: 2017-01-31
Project acronym onCOMBINE
Project Towards evidence-based combinations of approved and novel cancer drugs
Researcher (PI) Yosef YARDEN
Host Institution (HI) WEIZMANN INSTITUTE OF SCIENCE
Call Details Advanced Grant (AdG), LS7, ERC-2016-ADG
Summary Background: Molecular targeted therapy (TT; e.g., monoclonal antibodies, mAbs, and protein kinase inhibitors, PKIs) intercepts oncogene and other addictions of tumours. However, unlike chemotherapy, which employs cocktails of drugs, only rarely does TT harness poly-pharmacology. Because lung cancer is the major cause of oncology related fatalities and many driver mutations are known, this disease offers opportunities for establishing and generalizing novel TT combinations and their interface with the immune system.
Working hypothesis: High granularity maps of compensatory loops evoked by TT, along with deeper understanding of mechanisms underlying drug action, resistance and interactions with lymphoid/myeloid cells, will conceptualize drug combinations able to persistently inhibit tumours, while inducing only limited toxicities.
Goal and specific aims: Addressing resistance to TT, potential synergies and the immune system, we will employ lung cancer models driven by mutant EGFR, HER2, MET or AXL. Phosphoproteomics, transcriptomics and RNA interference, will enable mapping adaptations evoked by specific drugs. Once identified, we will test combinations of interceptors able to inhibit the primary target as well as the emerging, resistance-conferring route(s). Next, we will determine the mechanisms of action of selected interceptors (e.g., apoptosis, immunological cytotoxicity and senescence) as bases for optimising effective combinations. Homo-combinations of antibodies (i.e., antibodies recognising distinct epitopes of a receptor), hetero-combinations targeting distinct signalling and immune receptors, and combinations with PKIs will be examined in animal models.
Significance: More than 30 PKIs and >25 mAbs are approved in oncology, but most are used as monotherapies. Detailed knowledge of adaptation-driven resistance, mechanisms of drug action and immune effectors, will guide the long awaited application of TT combinations in oncology, including lung cancer.
Summary
Background: Molecular targeted therapy (TT; e.g., monoclonal antibodies, mAbs, and protein kinase inhibitors, PKIs) intercepts oncogene and other addictions of tumours. However, unlike chemotherapy, which employs cocktails of drugs, only rarely does TT harness poly-pharmacology. Because lung cancer is the major cause of oncology related fatalities and many driver mutations are known, this disease offers opportunities for establishing and generalizing novel TT combinations and their interface with the immune system.
Working hypothesis: High granularity maps of compensatory loops evoked by TT, along with deeper understanding of mechanisms underlying drug action, resistance and interactions with lymphoid/myeloid cells, will conceptualize drug combinations able to persistently inhibit tumours, while inducing only limited toxicities.
Goal and specific aims: Addressing resistance to TT, potential synergies and the immune system, we will employ lung cancer models driven by mutant EGFR, HER2, MET or AXL. Phosphoproteomics, transcriptomics and RNA interference, will enable mapping adaptations evoked by specific drugs. Once identified, we will test combinations of interceptors able to inhibit the primary target as well as the emerging, resistance-conferring route(s). Next, we will determine the mechanisms of action of selected interceptors (e.g., apoptosis, immunological cytotoxicity and senescence) as bases for optimising effective combinations. Homo-combinations of antibodies (i.e., antibodies recognising distinct epitopes of a receptor), hetero-combinations targeting distinct signalling and immune receptors, and combinations with PKIs will be examined in animal models.
Significance: More than 30 PKIs and >25 mAbs are approved in oncology, but most are used as monotherapies. Detailed knowledge of adaptation-driven resistance, mechanisms of drug action and immune effectors, will guide the long awaited application of TT combinations in oncology, including lung cancer.
Max ERC Funding
2 488 306 €
Duration
Start date: 2017-10-01, End date: 2022-09-30