Project acronym Antibodyomics
Project Vaccine profiling and immunodiagnostic discovery by high-throughput antibody repertoire analysis
Researcher (PI) Sai Tota Reddy
Host Institution (HI) EIDGENOESSISCHE TECHNISCHE HOCHSCHULE ZUERICH
Call Details Starting Grant (StG), LS7, ERC-2015-STG
Summary Vaccines and immunodiagnostics have been vital for public health and medicine, however a quantitative molecular understanding of vaccine-induced antibody responses is lacking. Antibody research is currently going through a big-data driven revolution, largely due to progress in next-generation sequencing (NGS) and bioinformatic analysis of antibody repertoires. A main advantage of high-throughput antibody repertoire analysis is that it provides a wealth of quantitative information not possible with other classical methods of antibody analysis (i.e., serum titers); this information includes: clonal distribution and diversity, somatic hypermutation patterns, and lineage tracing. In preliminary work my group has established standardized methods for antibody repertoire NGS, including an experimental-bioinformatic pipeline for error and bias correction that enables highly accurate repertoire sequencing and analysis. The overall goal of this proposal will be to apply high-throughput antibody repertoire analysis for quantitative vaccine profiling and discovery of next-generation immunodiagnostics. Using mouse subunit vaccination as our model system, we will answer for the first time, a fundamental biological question within the context of antibody responses - what is the link between genotype (antibody repertoire) and phenotype (serum antibodies)? We will expand upon this approach for improved rational vaccine design by quantitatively determining the impact of a comprehensive set of subunit vaccination parameters on complete antibody landscapes. Finally, we will develop advanced bioinformatic methods to discover immunodiagnostics based on antibody repertoire sequences. In summary, this proposal lays the foundation for fundamentally new approaches in the quantitative analysis of antibody responses, which long-term will promote the development of next-generation vaccines and immunodiagnostics.
Summary
Vaccines and immunodiagnostics have been vital for public health and medicine, however a quantitative molecular understanding of vaccine-induced antibody responses is lacking. Antibody research is currently going through a big-data driven revolution, largely due to progress in next-generation sequencing (NGS) and bioinformatic analysis of antibody repertoires. A main advantage of high-throughput antibody repertoire analysis is that it provides a wealth of quantitative information not possible with other classical methods of antibody analysis (i.e., serum titers); this information includes: clonal distribution and diversity, somatic hypermutation patterns, and lineage tracing. In preliminary work my group has established standardized methods for antibody repertoire NGS, including an experimental-bioinformatic pipeline for error and bias correction that enables highly accurate repertoire sequencing and analysis. The overall goal of this proposal will be to apply high-throughput antibody repertoire analysis for quantitative vaccine profiling and discovery of next-generation immunodiagnostics. Using mouse subunit vaccination as our model system, we will answer for the first time, a fundamental biological question within the context of antibody responses - what is the link between genotype (antibody repertoire) and phenotype (serum antibodies)? We will expand upon this approach for improved rational vaccine design by quantitatively determining the impact of a comprehensive set of subunit vaccination parameters on complete antibody landscapes. Finally, we will develop advanced bioinformatic methods to discover immunodiagnostics based on antibody repertoire sequences. In summary, this proposal lays the foundation for fundamentally new approaches in the quantitative analysis of antibody responses, which long-term will promote the development of next-generation vaccines and immunodiagnostics.
Max ERC Funding
1 492 586 €
Duration
Start date: 2016-06-01, End date: 2021-05-31
Project acronym BioProbe
Project "VERTICAL MICROFLUIDIC PROBE: A nanoliter ""Swiss army knife"" for chemistry and physics at biological interfaces"
Researcher (PI) Govindkrishna Govind Kaigala
Host Institution (HI) IBM RESEARCH GMBH
Call Details Starting Grant (StG), LS7, ERC-2012-StG_20111109
Summary Life is fundamentally characterised by order, compartmentalisation and biochemical reactions, which occurs at the right place right time – within, on the surface and between cells. Only a proportion of life processes can be addressed with contemporary approaches like liquid encapsulations (e.g. droplets) or engineering compartments (e.g. scaffolds). I believe these approaches are severely limited. I am convinced that a technique to study, work and locally probe adherent cells & tissues at micrometer distances from cell surfaces in “open space” would represent a major advance for the biology of biointerfaces. I therefore propose a non-contact, scanning technology, which spatially confines nanoliter volumes of chemicals for interacting with cells at the µm-length scale. This technology called the vertical microfluidic probe (vMFP) – that I developed at IBM-Zurich – shapes liquid on surfaces hydrodynamically and is compatible with samples on Petri dishes & microtiter plates. The project is organized in 4 themes:
(1) Advancing the vMFP by understanding the interaction of liquid flows with biointerfaces, integrating functional elements (e.g. heaters/electrodes, cell traps) & precision control.
(2) Developing a higher resolution method to stain tissue sections for multiple markers & better quality information.
(3) Retrieving rare elements such as circulating tumor cells from biologically diverse libraries.
(4) Patterning cells for applications in regenerative medicine.
Since cells & tissues will no longer be limited by closed systems, the vMFP will enable a completely new range of experiments to be performed in a highly interactive, versatile & precise manner – this approach departs from classical “closed” microfluidics. It is very likely that such a tool by providing multifunctional capabilities akin to the proverbial ‘Swiss army knife’ will be a unique facilitator for investigations of previously unapproachable problems in cell biology & the life science.
Summary
Life is fundamentally characterised by order, compartmentalisation and biochemical reactions, which occurs at the right place right time – within, on the surface and between cells. Only a proportion of life processes can be addressed with contemporary approaches like liquid encapsulations (e.g. droplets) or engineering compartments (e.g. scaffolds). I believe these approaches are severely limited. I am convinced that a technique to study, work and locally probe adherent cells & tissues at micrometer distances from cell surfaces in “open space” would represent a major advance for the biology of biointerfaces. I therefore propose a non-contact, scanning technology, which spatially confines nanoliter volumes of chemicals for interacting with cells at the µm-length scale. This technology called the vertical microfluidic probe (vMFP) – that I developed at IBM-Zurich – shapes liquid on surfaces hydrodynamically and is compatible with samples on Petri dishes & microtiter plates. The project is organized in 4 themes:
(1) Advancing the vMFP by understanding the interaction of liquid flows with biointerfaces, integrating functional elements (e.g. heaters/electrodes, cell traps) & precision control.
(2) Developing a higher resolution method to stain tissue sections for multiple markers & better quality information.
(3) Retrieving rare elements such as circulating tumor cells from biologically diverse libraries.
(4) Patterning cells for applications in regenerative medicine.
Since cells & tissues will no longer be limited by closed systems, the vMFP will enable a completely new range of experiments to be performed in a highly interactive, versatile & precise manner – this approach departs from classical “closed” microfluidics. It is very likely that such a tool by providing multifunctional capabilities akin to the proverbial ‘Swiss army knife’ will be a unique facilitator for investigations of previously unapproachable problems in cell biology & the life science.
Max ERC Funding
1 488 600 €
Duration
Start date: 2013-01-01, End date: 2017-12-31
Project acronym BRuSH
Project Oral bacteria as determinants for respiratory health
Researcher (PI) Randi BERTELSEN
Host Institution (HI) UNIVERSITETET I BERGEN
Call Details Starting Grant (StG), LS7, ERC-2018-STG
Summary The oral cavity is the gateway to the lower respiratory tract, and oral bacteria are likely to play a role in lung health. This may be the case for pathogens as well as commensal bacteria and the balance between species. The oral bacterial community of patients with periodontitis is dominated by gram-negative bacteria and a higher lipopolysaccharide (LPS) activity than in healthy microbiota. Furthermore, bacteria with especially potent pro-inflammatory LPS have been shown to be more common in the lungs of asthmatic than in healthy individuals. The working hypothesis of BRuSH is that microbiome communities dominated by LPS-producing bacteria which induce a particularly strong pro-inflammatory immune response in the host, will have a negative effect on respiratory health. I will test this hypothesis in two longitudinally designed population-based lung health studies. I aim to identify whether specific bacterial composition and types of LPS producing bacteria in oral and dust samples predict lung function and respiratory health over time; and if the different types of LPS-producing bacteria affect LPS in saliva saliva and dust. BRuSH will apply functional genome annotation that can assign biological significance to raw bacterial DNA sequences. With this bioinformatics tool I will cluster microbiome data into various LPS-producers: bacteria with LPS with strong inflammatory effects and others with weak- or antagonistic effects. The epidemiological studies will be supported by mice-models of asthma and cell assays of human bronchial epithelial cells, by exposing mice and bronchial cells to chemically synthesized Lipid A (the component that drive the LPS-induced immune responses) of various potency. The goal of BRuSH is to prove a causal relationship between oral microbiome and lung health, and gain knowledge that will enable us to make oral health a feasible target for intervention programs aimed at optimizing lung health and preventing respiratory disease.
Summary
The oral cavity is the gateway to the lower respiratory tract, and oral bacteria are likely to play a role in lung health. This may be the case for pathogens as well as commensal bacteria and the balance between species. The oral bacterial community of patients with periodontitis is dominated by gram-negative bacteria and a higher lipopolysaccharide (LPS) activity than in healthy microbiota. Furthermore, bacteria with especially potent pro-inflammatory LPS have been shown to be more common in the lungs of asthmatic than in healthy individuals. The working hypothesis of BRuSH is that microbiome communities dominated by LPS-producing bacteria which induce a particularly strong pro-inflammatory immune response in the host, will have a negative effect on respiratory health. I will test this hypothesis in two longitudinally designed population-based lung health studies. I aim to identify whether specific bacterial composition and types of LPS producing bacteria in oral and dust samples predict lung function and respiratory health over time; and if the different types of LPS-producing bacteria affect LPS in saliva saliva and dust. BRuSH will apply functional genome annotation that can assign biological significance to raw bacterial DNA sequences. With this bioinformatics tool I will cluster microbiome data into various LPS-producers: bacteria with LPS with strong inflammatory effects and others with weak- or antagonistic effects. The epidemiological studies will be supported by mice-models of asthma and cell assays of human bronchial epithelial cells, by exposing mice and bronchial cells to chemically synthesized Lipid A (the component that drive the LPS-induced immune responses) of various potency. The goal of BRuSH is to prove a causal relationship between oral microbiome and lung health, and gain knowledge that will enable us to make oral health a feasible target for intervention programs aimed at optimizing lung health and preventing respiratory disease.
Max ERC Funding
1 499 938 €
Duration
Start date: 2019-01-01, End date: 2023-12-31
Project acronym C-MORPH
Project Noninvasive cell specific morphometry in neuroinflammation and degeneration
Researcher (PI) Henrik LUNDELL
Host Institution (HI) REGION HOVEDSTADEN
Call Details Starting Grant (StG), LS7, ERC-2018-STG
Summary Brain structure determines function. Disentangling regional microstructural properties and understanding how these properties constitute brain function is a central goal of neuroimaging of the human brain and a key prerequisite for a mechanistic understanding of brain diseases and their treatment. Using magnetic resonance (MR) imaging, previous research has established links between regional brain microstructure and inter-individual variation in brain function, but this line of research has been limited by the non-specificity of MR-derived markers. This hampers the application of MR imaging as a tool to identify specific fingerprints of the underlying disease process.
Exploiting state-of-the-art ultra-high field MR imaging techniques, I have recently developed two independent spectroscopic MR methods that have the potential to tackle this challenge: Powder averaged diffusion weighted spectroscopy (PADWS) can provide an unbiased marker for cell specific structural degeneration, and Spectrally tuned gradient trajectories (STGT) can isolate cell shape and size. In this project, I will harness these innovations for MR-based precision medicine. I will advance PADWS and STGT methodology on state-of-the-art MR hardware and harvest the synergy of these methods to realize Cell-specific in-vivo MORPHOMETRY (C-MORPH) of the intact human brain. I will establish novel MR read-outs and analyses to derive cell-type specific tissue properties in the healthy and diseased brain and validate them with the help of a strong translational experimental framework, including histological validation. Once validated, the experimental methods and analyses will be simplified and adapted to provide clinically applicable tools. This will push the frontiers of MR-based personalized medicine, guiding therapeutic decisions by providing sensitive probes of cell-specific microstructural changes caused by inflammation, neurodegeneration or treatment response.
Summary
Brain structure determines function. Disentangling regional microstructural properties and understanding how these properties constitute brain function is a central goal of neuroimaging of the human brain and a key prerequisite for a mechanistic understanding of brain diseases and their treatment. Using magnetic resonance (MR) imaging, previous research has established links between regional brain microstructure and inter-individual variation in brain function, but this line of research has been limited by the non-specificity of MR-derived markers. This hampers the application of MR imaging as a tool to identify specific fingerprints of the underlying disease process.
Exploiting state-of-the-art ultra-high field MR imaging techniques, I have recently developed two independent spectroscopic MR methods that have the potential to tackle this challenge: Powder averaged diffusion weighted spectroscopy (PADWS) can provide an unbiased marker for cell specific structural degeneration, and Spectrally tuned gradient trajectories (STGT) can isolate cell shape and size. In this project, I will harness these innovations for MR-based precision medicine. I will advance PADWS and STGT methodology on state-of-the-art MR hardware and harvest the synergy of these methods to realize Cell-specific in-vivo MORPHOMETRY (C-MORPH) of the intact human brain. I will establish novel MR read-outs and analyses to derive cell-type specific tissue properties in the healthy and diseased brain and validate them with the help of a strong translational experimental framework, including histological validation. Once validated, the experimental methods and analyses will be simplified and adapted to provide clinically applicable tools. This will push the frontiers of MR-based personalized medicine, guiding therapeutic decisions by providing sensitive probes of cell-specific microstructural changes caused by inflammation, neurodegeneration or treatment response.
Max ERC Funding
1 498 811 €
Duration
Start date: 2018-12-01, End date: 2023-11-30
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 CHILIC
Project Child health intervention interactions in low-income countries
Researcher (PI) Christine Benn
Host Institution (HI) STATENS SERUM INSTITUT
Call Details Starting Grant (StG), LS7, ERC-2009-StG
Summary Vitamin A supplementation (VAS) and vaccines are the most powerful tools to reduce child mortality in low-income countries. However, we may not use these interventions optimally because we disregard that the interventions may have immunomodulatory effects which differ for boys and girls and which may interact with the effects of other interventions. I have proposed the hypothesis that VAS and vaccines interact. This hypothesis is supported by randomised and observational studies showing that the combination of VAS and DTP may be harmful. I have furthermore proposed that VAS has sex-differential effects. VAS seems beneficial for boys but may not carry any benefits for girls. These findings challenge the current understanding that VAS and vaccines have only targeted effects and can be given together without considering interactions. This is of outmost importance for policy makers. The global trend is to combine health interventions for logistic reasons. My research suggests that this may not always be a good idea. Furthermore, the concept of sex-differential response to our common health interventions opens up for a completely new understanding of the immunology of the two sexes and may imply that we need to treat the two sexes differently in order to treat them optimally possibly also in high-income countries. In the present proposal I outline a series of inter-disciplinary epidemiological and immunological studies, which will serve to determine the overall and sex-differential effects of VAS and vaccines, the mechanisms behind these effects, and the basis for the immunological difference between boys and girls. If my hypotheses are true we can use the existing tools in a more optimal way to reduce child mortality without increasing costs. Thus, the results could lead to shifts in policy as well as paradigms.
Summary
Vitamin A supplementation (VAS) and vaccines are the most powerful tools to reduce child mortality in low-income countries. However, we may not use these interventions optimally because we disregard that the interventions may have immunomodulatory effects which differ for boys and girls and which may interact with the effects of other interventions. I have proposed the hypothesis that VAS and vaccines interact. This hypothesis is supported by randomised and observational studies showing that the combination of VAS and DTP may be harmful. I have furthermore proposed that VAS has sex-differential effects. VAS seems beneficial for boys but may not carry any benefits for girls. These findings challenge the current understanding that VAS and vaccines have only targeted effects and can be given together without considering interactions. This is of outmost importance for policy makers. The global trend is to combine health interventions for logistic reasons. My research suggests that this may not always be a good idea. Furthermore, the concept of sex-differential response to our common health interventions opens up for a completely new understanding of the immunology of the two sexes and may imply that we need to treat the two sexes differently in order to treat them optimally possibly also in high-income countries. In the present proposal I outline a series of inter-disciplinary epidemiological and immunological studies, which will serve to determine the overall and sex-differential effects of VAS and vaccines, the mechanisms behind these effects, and the basis for the immunological difference between boys and girls. If my hypotheses are true we can use the existing tools in a more optimal way to reduce child mortality without increasing costs. Thus, the results could lead to shifts in policy as well as paradigms.
Max ERC Funding
1 686 043 €
Duration
Start date: 2010-01-01, End date: 2014-12-31
Project acronym CHIPS
Project Effects of Prenatal Exposure to Acrylamide on Health: Prospective Biomarker-Based Studies
Researcher (PI) Marie Pedersen
Host Institution (HI) KOBENHAVNS UNIVERSITET
Call Details Starting Grant (StG), LS7, ERC-2017-STG
Summary Background: Acrylamide is a chemical formed in many commonly consumed foods and beverages. It is neurotoxic, crosses the placenta and has been associated with restriction of fetal growth in humans. In animals, acrylamide causes heritable mutations, tumors, developmental toxicity, reduced fertility and impaired growth. Therefore, the discovery of acrylamide in food in 2002 raised concern about human health effects worldwide. Still, epidemiological studies are limited and effects on health of prenatal exposure have never been evaluated.
Research gaps: Epidemiological studies have mostly addressed exposure during adulthood, focused on cancer risk in adults, and relied on questionnaires entailing a high degree of exposure misclassification. Biomarker studies on prenatal exposure to acrylamide from diet are critically needed to improve exposure assessment and to determine whether acrylamide leads to major diseases later in life.
Own results: I have first authored a prospective European study showing that prenatal exposure to acrylamide, estimated by measuring hemoglobin adducts in cord blood, was associated with fetal growth restriction, for the first time.
Objectives: To determine the effects of prenatal exposure to acrylamide alone and in combination with other potentially toxic adduct-forming exposures on the health of children and young adults.
Methods: Both well-established and innovative biomarker methods will be used for characterization of prenatal exposure to acrylamide and related toxicants in blood from pregnant women and their offspring in prospective cohort studies with long-term follow-up. Risk of neurological disorders, impaired cognition, disturbed reproductive function and metabolic outcomes such as obesity and diabetes will be evaluated.
Perspectives: CHIPS project will provide a better understanding of the impact of prenatal exposure to acrylamide from diet on human health urgently needed for targeted strategies for the protection of the health.
Summary
Background: Acrylamide is a chemical formed in many commonly consumed foods and beverages. It is neurotoxic, crosses the placenta and has been associated with restriction of fetal growth in humans. In animals, acrylamide causes heritable mutations, tumors, developmental toxicity, reduced fertility and impaired growth. Therefore, the discovery of acrylamide in food in 2002 raised concern about human health effects worldwide. Still, epidemiological studies are limited and effects on health of prenatal exposure have never been evaluated.
Research gaps: Epidemiological studies have mostly addressed exposure during adulthood, focused on cancer risk in adults, and relied on questionnaires entailing a high degree of exposure misclassification. Biomarker studies on prenatal exposure to acrylamide from diet are critically needed to improve exposure assessment and to determine whether acrylamide leads to major diseases later in life.
Own results: I have first authored a prospective European study showing that prenatal exposure to acrylamide, estimated by measuring hemoglobin adducts in cord blood, was associated with fetal growth restriction, for the first time.
Objectives: To determine the effects of prenatal exposure to acrylamide alone and in combination with other potentially toxic adduct-forming exposures on the health of children and young adults.
Methods: Both well-established and innovative biomarker methods will be used for characterization of prenatal exposure to acrylamide and related toxicants in blood from pregnant women and their offspring in prospective cohort studies with long-term follow-up. Risk of neurological disorders, impaired cognition, disturbed reproductive function and metabolic outcomes such as obesity and diabetes will be evaluated.
Perspectives: CHIPS project will provide a better understanding of the impact of prenatal exposure to acrylamide from diet on human health urgently needed for targeted strategies for the protection of the health.
Max ERC Funding
1 499 531 €
Duration
Start date: 2018-07-01, End date: 2023-06-30
Project acronym DE-ORPHAN
Project DEtermination of Orphan Receptor PHysiological Agonists and sigNals
Researcher (PI) David Erik Immanuel Gloriam
Host Institution (HI) KOBENHAVNS UNIVERSITET
Call Details Starting Grant (StG), LS7, ERC-2014-STG
Summary G protein-coupled receptors make up both the largest membrane protein and drug target families. DE-ORPHAN aims to determine the close functional context; specifically physiological agonists and signaling pathways; and provide the first research tool compounds, of orphan peptide receptors.
Determination of physiological agonists (aka de-orphanization), by high-throughput screening has largely failed. We will introduce a new research strategy: 1) developing highly innovative bioinformatics methods for handpicking of all orphan receptor targets and candidate ligand screening libraries; and 2) employing a screening technique that can measure all signaling pathways simultaneously.
The first potent and selective pharmacological tool compounds will be identified by chemoinformatic design of focused screening libraries. We will establish the ligands’ structure-activity relationships important for biological activity and further optimization towards drugs.
The first potent and selective Gs- and G12/13 protein inhibitors will be designed by structure-based re-optimization from a recent crystal structure of a Gq-inhibitor complex, and applied to determine orphan receptor signaling pathways and ligand pathway-bias. They will open up for efficient dissection of important signaling networks and development of drugs with fewer side effects.
DE-ORPHANs design hypotheses are based on unique computational methods to analyze protein and ligand similarities and are founded on genomic and protein sequences, structural data and ligands. The interdisciplinary research strategy applies multiple ligands acting independently but in concert to provide complementary receptor characterization. The results will allow the research field to advance into studies of receptor functions and exploitation of druggable targets, ligands and mechanisms. Which physiological insights and therapeutic breakthroughs will we witness when these receptors find their place in human pharmacology and medicine?
Summary
G protein-coupled receptors make up both the largest membrane protein and drug target families. DE-ORPHAN aims to determine the close functional context; specifically physiological agonists and signaling pathways; and provide the first research tool compounds, of orphan peptide receptors.
Determination of physiological agonists (aka de-orphanization), by high-throughput screening has largely failed. We will introduce a new research strategy: 1) developing highly innovative bioinformatics methods for handpicking of all orphan receptor targets and candidate ligand screening libraries; and 2) employing a screening technique that can measure all signaling pathways simultaneously.
The first potent and selective pharmacological tool compounds will be identified by chemoinformatic design of focused screening libraries. We will establish the ligands’ structure-activity relationships important for biological activity and further optimization towards drugs.
The first potent and selective Gs- and G12/13 protein inhibitors will be designed by structure-based re-optimization from a recent crystal structure of a Gq-inhibitor complex, and applied to determine orphan receptor signaling pathways and ligand pathway-bias. They will open up for efficient dissection of important signaling networks and development of drugs with fewer side effects.
DE-ORPHANs design hypotheses are based on unique computational methods to analyze protein and ligand similarities and are founded on genomic and protein sequences, structural data and ligands. The interdisciplinary research strategy applies multiple ligands acting independently but in concert to provide complementary receptor characterization. The results will allow the research field to advance into studies of receptor functions and exploitation of druggable targets, ligands and mechanisms. Which physiological insights and therapeutic breakthroughs will we witness when these receptors find their place in human pharmacology and medicine?
Max ERC Funding
1 499 926 €
Duration
Start date: 2015-05-01, End date: 2020-04-30
Project acronym DNA-AMP
Project DNA Adduct Molecular Probes: Elucidating the Diet-Cancer Connection at Chemical Resolution
Researcher (PI) Shana Jocette Sturla
Host Institution (HI) EIDGENOESSISCHE TECHNISCHE HOCHSCHULE ZUERICH
Call Details Starting Grant (StG), LS7, ERC-2010-StG_20091118
Summary Bulky DNA adducts formed from chemical carcinogens dictate structure, reactivity, and mechanism of chemical-biological reactions; therefore, their identification is central to evaluating and mitigating cancer risk. Natural food components, or others associated with certain food preparations or metabolic conversions, initiate potentially damaging genetic mutations after forming DNA adducts, which contribute critically to carcinogenesis, despite the fact that they are typically repaired biochemically and they are formed at extremely low levels. This situation places significant limitations on our ability to understand the role of formation, repair, and mutagenesis on the basis of the complex DNA reactivity profiles of food components. The long-term goals of this research are to contribute basic knowledge and advanced experimental tools required to understand, on the basis of chemical structure, the contributions of chronic, potentially adverse, dietary chemical carcinogen exposure to cancer development. It is proposed that a new class of synthetic nucleosides, devised on the basis of preliminary discoveries made in the independent laboratory of the applicant, will serve as molecular probes for bulky DNA adducts and can be effectively used to study and AMPlify, i.e. as a sensitive diagnostic tool, low levels of chemically-specific modes of DNA damage. The proposed research is a chemical biology-based approach to the study of carcinogenesis. Experiments involve chemical synthesis, thermodynamic and kinetic characterization DNA-DNA and enzyme-DNA interactions, and nanoparticle-based molecular probes. The proposal describes a potentially ground-breaking approach for profiling the biological reactivities of chemical carcinogens, and we expect to gain fundamental knowledge and chemical tools that can contribute to the prevention of diseases influenced by gene-environment interactions.
Summary
Bulky DNA adducts formed from chemical carcinogens dictate structure, reactivity, and mechanism of chemical-biological reactions; therefore, their identification is central to evaluating and mitigating cancer risk. Natural food components, or others associated with certain food preparations or metabolic conversions, initiate potentially damaging genetic mutations after forming DNA adducts, which contribute critically to carcinogenesis, despite the fact that they are typically repaired biochemically and they are formed at extremely low levels. This situation places significant limitations on our ability to understand the role of formation, repair, and mutagenesis on the basis of the complex DNA reactivity profiles of food components. The long-term goals of this research are to contribute basic knowledge and advanced experimental tools required to understand, on the basis of chemical structure, the contributions of chronic, potentially adverse, dietary chemical carcinogen exposure to cancer development. It is proposed that a new class of synthetic nucleosides, devised on the basis of preliminary discoveries made in the independent laboratory of the applicant, will serve as molecular probes for bulky DNA adducts and can be effectively used to study and AMPlify, i.e. as a sensitive diagnostic tool, low levels of chemically-specific modes of DNA damage. The proposed research is a chemical biology-based approach to the study of carcinogenesis. Experiments involve chemical synthesis, thermodynamic and kinetic characterization DNA-DNA and enzyme-DNA interactions, and nanoparticle-based molecular probes. The proposal describes a potentially ground-breaking approach for profiling the biological reactivities of chemical carcinogens, and we expect to gain fundamental knowledge and chemical tools that can contribute to the prevention of diseases influenced by gene-environment interactions.
Max ERC Funding
1 500 000 €
Duration
Start date: 2010-09-01, End date: 2015-08-31
Project acronym DrugsInPregnancy
Project Effects of Medication Use in Pregnancy on Infant Neurodevelopment
Researcher (PI) Hedvig Marie Egeland Nordeng
Host Institution (HI) UNIVERSITETET I OSLO
Call Details Starting Grant (StG), LS7, ERC-2014-STG
Summary Currently, thousands of pregnant women in the EU and worldwide are being increasingly prescribed medications for which we do not have sufficient information on fetal safety. I hypothesize that our current understanding of safety pharmacology is oversimplified and that medication prescribed during pregnancy may play an unrecognized role in the development of neurodevelopmental disorders.
In this research proposal we have the unique opportunity to use a large population-based birth cohort including over 100 000 mother-child pairs and biological data to study how medications may act on the offspring. This offers novel and innovative pharmaceutical insight into the safety of medications.
By linking several nationwide registries (National Prescription Data Base, Norwegian Patient Registry, Medical Birth Registry) to a population-based birth cohort (n=108 000) we specifically aim to 1) estimate the effect of prenatal exposure to psychotropics and analgesics on neurodevelopment in young children using a range of methodological approaches to strengthen causal inference.
With these data made available, we will 2) determine whether fetal exposure to specific medications results in epigenetic events (i.e. changes in DNA methylation) in the child, and 3) determine whether such changes increase the risks of neurodevelopmental disorders in childhood.
The recent availability of large scale human data, possibility of register linkages and genome-wide mapping of DNA methylation at affordable costs makes this research proposal now possible. The size and richness of data including over hundred thousand pregnancies and existence of biological material makes this project unique. The final outcome will be fundamentally new knowledge about how medications affect the developing unborn child and will open up new horizons and opportunities for future research in a new field of “pharmaco-epigenetics” and enhance our understanding of origins of neurodevelopmental disorders.
Summary
Currently, thousands of pregnant women in the EU and worldwide are being increasingly prescribed medications for which we do not have sufficient information on fetal safety. I hypothesize that our current understanding of safety pharmacology is oversimplified and that medication prescribed during pregnancy may play an unrecognized role in the development of neurodevelopmental disorders.
In this research proposal we have the unique opportunity to use a large population-based birth cohort including over 100 000 mother-child pairs and biological data to study how medications may act on the offspring. This offers novel and innovative pharmaceutical insight into the safety of medications.
By linking several nationwide registries (National Prescription Data Base, Norwegian Patient Registry, Medical Birth Registry) to a population-based birth cohort (n=108 000) we specifically aim to 1) estimate the effect of prenatal exposure to psychotropics and analgesics on neurodevelopment in young children using a range of methodological approaches to strengthen causal inference.
With these data made available, we will 2) determine whether fetal exposure to specific medications results in epigenetic events (i.e. changes in DNA methylation) in the child, and 3) determine whether such changes increase the risks of neurodevelopmental disorders in childhood.
The recent availability of large scale human data, possibility of register linkages and genome-wide mapping of DNA methylation at affordable costs makes this research proposal now possible. The size and richness of data including over hundred thousand pregnancies and existence of biological material makes this project unique. The final outcome will be fundamentally new knowledge about how medications affect the developing unborn child and will open up new horizons and opportunities for future research in a new field of “pharmaco-epigenetics” and enhance our understanding of origins of neurodevelopmental disorders.
Max ERC Funding
1 499 439 €
Duration
Start date: 2015-09-01, End date: 2020-08-31
Project acronym ImPRESS
Project Imaging Perfusion Restrictions from Extracellular Solid Stress
Researcher (PI) Kyrre Eeg Emblem
Host Institution (HI) OSLO UNIVERSITETSSYKEHUS HF
Call Details Starting Grant (StG), LS7, ERC-2017-STG
Summary Even the perfect cancer drug must reach its target to have an effect. The ImPRESS project main objective is to develop a novel imaging paradigm coined Restricted Perfusion Imaging (RPI) to reveal - for the first time in humans - vascular restrictions in solid cancers caused by mechanical solid stress, and use RPI to demonstrate that alleviating this force will repair the cancerous microenvironment and improve therapeutic response. Delivery of anti-cancer drugs to the tumor is critically dependent on a functional vascular bed. Developing biomarkers that can measure how mechanical forces in a solid tumor impair perfusion and promotes therapy resistance is essential for treatment of disease.
The ImPRESS project is based on the following observations; (I) pre-clinical work suggests that therapies targeting the tumor microenvironment and extracellular matrix may enhance drug delivery by decompressing tumor vessels; (II) results from animal models may not be transferable because compressive forces in human tumors in vivo can be many times higher; and (III) there are no available imaging technologies for medical diagnostics of solid stress in human cancers. Using RPI, ImPRESS will conduct a comprehensive series of innovative studies in brain cancer patients to answer three key questions: (Q1) Can we image vascular restrictions in human cancers and map how the vasculature changes with tumor growth or treatment? (Q2) Can we use medical engineering to image solid stress in vivo? (Q3) Can RPI show that matrix-depleting drugs improve patient response to conventional chemo- and radiation therapy as well as new targeted therapies?
The ImPRESS project holds a unique position to answer these questions by our unrivaled experience with advanced imaging of cancer patients. With successful delivery, ImPRESS will have a direct impact on patient treatment and establish an imaging paradigm that will pave the way for new scientific knowledge on how to revitalize cancer therapies.
Summary
Even the perfect cancer drug must reach its target to have an effect. The ImPRESS project main objective is to develop a novel imaging paradigm coined Restricted Perfusion Imaging (RPI) to reveal - for the first time in humans - vascular restrictions in solid cancers caused by mechanical solid stress, and use RPI to demonstrate that alleviating this force will repair the cancerous microenvironment and improve therapeutic response. Delivery of anti-cancer drugs to the tumor is critically dependent on a functional vascular bed. Developing biomarkers that can measure how mechanical forces in a solid tumor impair perfusion and promotes therapy resistance is essential for treatment of disease.
The ImPRESS project is based on the following observations; (I) pre-clinical work suggests that therapies targeting the tumor microenvironment and extracellular matrix may enhance drug delivery by decompressing tumor vessels; (II) results from animal models may not be transferable because compressive forces in human tumors in vivo can be many times higher; and (III) there are no available imaging technologies for medical diagnostics of solid stress in human cancers. Using RPI, ImPRESS will conduct a comprehensive series of innovative studies in brain cancer patients to answer three key questions: (Q1) Can we image vascular restrictions in human cancers and map how the vasculature changes with tumor growth or treatment? (Q2) Can we use medical engineering to image solid stress in vivo? (Q3) Can RPI show that matrix-depleting drugs improve patient response to conventional chemo- and radiation therapy as well as new targeted therapies?
The ImPRESS project holds a unique position to answer these questions by our unrivaled experience with advanced imaging of cancer patients. With successful delivery, ImPRESS will have a direct impact on patient treatment and establish an imaging paradigm that will pave the way for new scientific knowledge on how to revitalize cancer therapies.
Max ERC Funding
1 499 638 €
Duration
Start date: 2018-01-01, End date: 2022-12-31
Project acronym MOBILIZE
Project Improving health in people with multimorbidity: a paradigm shift in health care from disease-based curative models to personalized exercise therapy and self-management
Researcher (PI) Søren T. SKOU
Host Institution (HI) SYDDANSK UNIVERSITET
Call Details Starting Grant (StG), LS7, ERC-2018-STG
Summary The goal of this proposal is to support the paradigm shift in the health care of people with multiple chronic conditions in Europe from a focus on disease-based curative models to holistic person-centered self-care through personalized,supervised exercise therapy and education.
The problem:The impact of multimorbidity on the individual and society is massive and much greater than the impact of single chronic conditions alone. However, effective treatments are missing and research and health care reinforce an inefficient and burdensome single-disease framework.
The solution:Exercise has the potential to disrupt the ‘vicious cycle’ of systemic inflammation associated with chronic conditions and improve health in multimorbidity. A personalized exercise and education program aimed at supporting subsequent self-management by the individual will be developed in an interdisciplinary collaboration, building on evidence from biomarkers, patient involvement and methodological expertise. Self-reported,physiological and societal effects will be investigated in a randomized controlled trial comparing the personalized program with standard single-disease models of care. Scientific and public dissemination and implementation ensuring significant personal and societal benefit is fundamental to the proposal.
The proposal is associated with high risk, as the current disease-based curative models involve treatment by several highly specialized health care providers, while the new person-centered self-management model is centered on a personalized program delivered by one health care provider.
The ground-breaking nature of this proposal lies in its potential to revolutionize how health care is organized for people with multimorbidity, by giving them one primary care provider, and how we use non-surgical treatment in health care and science by bringing the concept of precision medicine into multimorbidity and utilizing it to improve treatment outcome with exercise therapy as the model.
Summary
The goal of this proposal is to support the paradigm shift in the health care of people with multiple chronic conditions in Europe from a focus on disease-based curative models to holistic person-centered self-care through personalized,supervised exercise therapy and education.
The problem:The impact of multimorbidity on the individual and society is massive and much greater than the impact of single chronic conditions alone. However, effective treatments are missing and research and health care reinforce an inefficient and burdensome single-disease framework.
The solution:Exercise has the potential to disrupt the ‘vicious cycle’ of systemic inflammation associated with chronic conditions and improve health in multimorbidity. A personalized exercise and education program aimed at supporting subsequent self-management by the individual will be developed in an interdisciplinary collaboration, building on evidence from biomarkers, patient involvement and methodological expertise. Self-reported,physiological and societal effects will be investigated in a randomized controlled trial comparing the personalized program with standard single-disease models of care. Scientific and public dissemination and implementation ensuring significant personal and societal benefit is fundamental to the proposal.
The proposal is associated with high risk, as the current disease-based curative models involve treatment by several highly specialized health care providers, while the new person-centered self-management model is centered on a personalized program delivered by one health care provider.
The ground-breaking nature of this proposal lies in its potential to revolutionize how health care is organized for people with multimorbidity, by giving them one primary care provider, and how we use non-surgical treatment in health care and science by bringing the concept of precision medicine into multimorbidity and utilizing it to improve treatment outcome with exercise therapy as the model.
Max ERC Funding
1 499 230 €
Duration
Start date: 2019-08-01, End date: 2024-07-31
Project acronym NanoSCAN
Project Developing multi-modality nanomedicines for targeted annotation of oncogenic signaling pathways
Researcher (PI) Jason Philip Holland
Host Institution (HI) UNIVERSITAT ZURICH
Call Details Starting Grant (StG), LS7, ERC-2015-STG
Summary Spatial and temporal changes in the underlying biochemistry of cancer control disease progression and response/resistance to treatment. Developing methods to detect changes in oncogenic signaling at an early stage is vital to further our understanding of cancer, and will advance the next generation of anti-cancer therapies. Nanomedicine is the medical application of nanotechnology to diagnose or treat disease. In light of the recent introduction of tools like Positron Emission Tomography/Magnetic Resonance Imaging (PET/MRI) scanners, there is now a new opportunity to develop hybrid imaging protocols that can simultaneously take advantage of the functional and anatomic information available from PET/MRI to address changes in oncogenic signaling pathways. The work outlined in this interdisciplinary ERC Project is designed to advance new chemistry and imaging methods to measure changes in oncogenic signaling in various cancers before, during and after treatment using PET/MRI. The long-term goals are to expand the scope and utility of radiolabelled nanomedicines as dual-modality PET/MRI probes for detecting changes in oncogenic signaling in various cancers and develop efficient methods for translating new technologies to the clinic. Successful completion of this ERC Project has the potential to transform personalised clinical management of cancer patients via advanced PET/MRI detection of oncogenic signaling processes.
Summary
Spatial and temporal changes in the underlying biochemistry of cancer control disease progression and response/resistance to treatment. Developing methods to detect changes in oncogenic signaling at an early stage is vital to further our understanding of cancer, and will advance the next generation of anti-cancer therapies. Nanomedicine is the medical application of nanotechnology to diagnose or treat disease. In light of the recent introduction of tools like Positron Emission Tomography/Magnetic Resonance Imaging (PET/MRI) scanners, there is now a new opportunity to develop hybrid imaging protocols that can simultaneously take advantage of the functional and anatomic information available from PET/MRI to address changes in oncogenic signaling pathways. The work outlined in this interdisciplinary ERC Project is designed to advance new chemistry and imaging methods to measure changes in oncogenic signaling in various cancers before, during and after treatment using PET/MRI. The long-term goals are to expand the scope and utility of radiolabelled nanomedicines as dual-modality PET/MRI probes for detecting changes in oncogenic signaling in various cancers and develop efficient methods for translating new technologies to the clinic. Successful completion of this ERC Project has the potential to transform personalised clinical management of cancer patients via advanced PET/MRI detection of oncogenic signaling processes.
Max ERC Funding
1 700 000 €
Duration
Start date: 2016-09-01, End date: 2021-08-31
Project acronym NASTAR
Project Nano-Sensitizer Cancer Cell Targeted Radiotherapy
Researcher (PI) Thomas Lars Andresen
Host Institution (HI) DANMARKS TEKNISKE UNIVERSITET
Call Details Starting Grant (StG), LS7, ERC-2012-StG_20111109
Summary Targeted chemotherapy in combination with external beam radiation therapy (radiotherapy) is a promising approach to significantly improve the therapeutic outcome for cancer patients. To achieve this, it is essential to develop drug delivery technology that specifically delivers the chemotherapeutic drugs to cancerous tissue. Radiotherapy is an indispensable part of modern cancer treatment; however, despite efforts in improving planning and execution of treatment, the unbalance between therapeutic benefit and side effects limits cure rates, and new approaches are needed to bring to fruition the full potential of radiotherapy. Today, there is considerable focus on systemically administered radiosensitizers for enhancing the effect of radiotherapy and clinical investigations have shown promising results; however, radiosensitizer use is hampered by considerable side effects due to lack of drug targeting to the cancerous tissue.
In the first phase of this project, the aim is to develop tumor targeted nanocarrier delivery systems of radiosensitizers to enhance their therapeutic potential and provide a more efficient and site-directed effect of radiotherapy. In the second phase of the project, nanocarriers for tumor specific delivery of checkpoint inhibitors of cancer cell repair mechanisms will be investigated as an additional targeting strategy for sensitizing cancer cells to radiotherapy. The idea is to circumvent cell cycle checkpoints of DNA damage induced by tumor radiation and thereby enhance mitotic catastrophe. This approach will in combination with the delivery of conventional radiosensitizer drugs, further lower the radiation dose needed to induce irreversible damage to cancer cells.
Thus, the project aims to develop targeted nanocarriers for high precision delivery of radiosensitizing drugs to cancerous tissue for enhancing the effect of radiotherapy. We aim to demonstrate the applicability and clinical potential of this new approach within the project period
Summary
Targeted chemotherapy in combination with external beam radiation therapy (radiotherapy) is a promising approach to significantly improve the therapeutic outcome for cancer patients. To achieve this, it is essential to develop drug delivery technology that specifically delivers the chemotherapeutic drugs to cancerous tissue. Radiotherapy is an indispensable part of modern cancer treatment; however, despite efforts in improving planning and execution of treatment, the unbalance between therapeutic benefit and side effects limits cure rates, and new approaches are needed to bring to fruition the full potential of radiotherapy. Today, there is considerable focus on systemically administered radiosensitizers for enhancing the effect of radiotherapy and clinical investigations have shown promising results; however, radiosensitizer use is hampered by considerable side effects due to lack of drug targeting to the cancerous tissue.
In the first phase of this project, the aim is to develop tumor targeted nanocarrier delivery systems of radiosensitizers to enhance their therapeutic potential and provide a more efficient and site-directed effect of radiotherapy. In the second phase of the project, nanocarriers for tumor specific delivery of checkpoint inhibitors of cancer cell repair mechanisms will be investigated as an additional targeting strategy for sensitizing cancer cells to radiotherapy. The idea is to circumvent cell cycle checkpoints of DNA damage induced by tumor radiation and thereby enhance mitotic catastrophe. This approach will in combination with the delivery of conventional radiosensitizer drugs, further lower the radiation dose needed to induce irreversible damage to cancer cells.
Thus, the project aims to develop targeted nanocarriers for high precision delivery of radiosensitizing drugs to cancerous tissue for enhancing the effect of radiotherapy. We aim to demonstrate the applicability and clinical potential of this new approach within the project period
Max ERC Funding
1 498 731 €
Duration
Start date: 2013-04-01, End date: 2019-02-28
Project acronym OPTIM
Project Optimized drug combinations for effective cancer treatment: a personalised approach.
Researcher (PI) Patrycja*Monika Nowak-Sliwinska
Host Institution (HI) UNIVERSITE DE GENEVE
Call Details Starting Grant (StG), LS7, ERC-2015-STG
Summary This project aims to improve the treatment of metastasized colorectal carcinoma (mCRC), as treatment options after first line chemotherapy are desperately needed. The key to improvement of cancer therapy resides in optimal combination of drugs. Optimally combining drugs is non-trivial due to the large number of possibilities, especially when more than two drugs are combined at various doses. In the current research program I propose to use a differential evolution guided stochastic search algorithm to guide the way in finding optimal combination therapies. In previous research I have applied this feedback system control (FSC) technique to navigate through the enormous parametric space of nine angiostatic drugs at four doses. The straightforward iterative approach of in vitro cell viability testing and algorithm-based analysis identified optimal synergistic low-dose drug combinations. In vivo translation by maintaining the drug dose ratio led to effective anti-cancer activity, without evidence of side-effects.
A new screen for optimal targeted combination treatment of advanced CRC will be performed. A series of 7 genetically different human CRC cell lines will be used in this screen, thus simulating personalized treatment. The optimized combinations will be ‘ratiometrically’ translated into orthotopic and metastasizing preclinical CRC mouse models and tested in parallel to standard chemotherapy regimens. Development of a method for a personalized screen using freshly isolated tumor cells will prepare the technology for application in the clinic.
Using an innovative strategy I previously identified a series of novel markers of the tumor endothelium. After validation of these targets, this project aims for the design of new drugs to be used in a screen for optimal combination therapy for mCRC. The translational and multidisciplinary nature of the current proposal aims for preparing an improved therapeutic combination regimen for testing in cancer patients.
Summary
This project aims to improve the treatment of metastasized colorectal carcinoma (mCRC), as treatment options after first line chemotherapy are desperately needed. The key to improvement of cancer therapy resides in optimal combination of drugs. Optimally combining drugs is non-trivial due to the large number of possibilities, especially when more than two drugs are combined at various doses. In the current research program I propose to use a differential evolution guided stochastic search algorithm to guide the way in finding optimal combination therapies. In previous research I have applied this feedback system control (FSC) technique to navigate through the enormous parametric space of nine angiostatic drugs at four doses. The straightforward iterative approach of in vitro cell viability testing and algorithm-based analysis identified optimal synergistic low-dose drug combinations. In vivo translation by maintaining the drug dose ratio led to effective anti-cancer activity, without evidence of side-effects.
A new screen for optimal targeted combination treatment of advanced CRC will be performed. A series of 7 genetically different human CRC cell lines will be used in this screen, thus simulating personalized treatment. The optimized combinations will be ‘ratiometrically’ translated into orthotopic and metastasizing preclinical CRC mouse models and tested in parallel to standard chemotherapy regimens. Development of a method for a personalized screen using freshly isolated tumor cells will prepare the technology for application in the clinic.
Using an innovative strategy I previously identified a series of novel markers of the tumor endothelium. After validation of these targets, this project aims for the design of new drugs to be used in a screen for optimal combination therapy for mCRC. The translational and multidisciplinary nature of the current proposal aims for preparing an improved therapeutic combination regimen for testing in cancer patients.
Max ERC Funding
1 199 436 €
Duration
Start date: 2016-05-01, End date: 2020-04-30
Project acronym PhaseX
Project Phase contrast X-ray imaging for medicine
Researcher (PI) Marco Stampanoni
Host Institution (HI) EIDGENOESSISCHE TECHNISCHE HOCHSCHULE ZUERICH
Call Details Starting Grant (StG), LS7, ERC-2012-StG_20111109
Summary The goal of this grant is to introduce grating-based phase contrast enhanced X-ray imaging as a novel diagnostic tool in human medicine. We will first gain fundamental knowledge to master high-energy grating-based phase contrast imaging and develop novel radiological equipment at the demonstrator/prototype level to perform phase contrast enhanced investigation in human patients in-vivo. In a second phase, we will investigate in depth how our novel method applies to a few pivotal medical fields like mammography, vascular- and musculoskeletal diseases. In these areas there is a strong need for radiological investigations with improved density discrimination, specifically for early breast cancer detection, plaques visualization and cartilage, tendons or ligaments imaging. No hospital or clinical infrastructure in the world can perform phase contrast enhanced X-ray radiological investigations: therefore our discoveries will constitute an enormous impact in this field.
Summary
The goal of this grant is to introduce grating-based phase contrast enhanced X-ray imaging as a novel diagnostic tool in human medicine. We will first gain fundamental knowledge to master high-energy grating-based phase contrast imaging and develop novel radiological equipment at the demonstrator/prototype level to perform phase contrast enhanced investigation in human patients in-vivo. In a second phase, we will investigate in depth how our novel method applies to a few pivotal medical fields like mammography, vascular- and musculoskeletal diseases. In these areas there is a strong need for radiological investigations with improved density discrimination, specifically for early breast cancer detection, plaques visualization and cartilage, tendons or ligaments imaging. No hospital or clinical infrastructure in the world can perform phase contrast enhanced X-ray radiological investigations: therefore our discoveries will constitute an enormous impact in this field.
Max ERC Funding
1 499 300 €
Duration
Start date: 2012-11-01, End date: 2017-10-31
Project acronym PICS THERAPY
Project Manipulation of senescence pathways for cancer therapy: from experimental models to clinic
Researcher (PI) Andrea Alimonti
Host Institution (HI) Ente Ospedaliero Cantonale
Call Details Starting Grant (StG), LS7, ERC-2010-StG_20091118
Summary This proposal aims to harness a novel type of senescence that we have identified in response to acute Pten inactivation, and which we believe offers a radical therapeutic approach to target the quiescent cancer stem cell in vivo. In characterizing Pten loss Induced Cellular Senescence, which we have named PICS for short, we have discovered that PICS is distinct from other forms of cellular senescence including oncogene-induced senescence (OIS) and replicative senescence. These distinct differences are characterized by a lack of DNA damage and hyper-replication, breaking the current dogma for senescence induction. The ability to induce senescence, an irreversible growth arrest, in cells by targeting Pten signaling, without a requirement for hyper-replication and DNA damage opens up the possibility to target quiescent cells, including stem cells, that have a low proliferative index. This approach has tremendous therapeutic potential and represents one of the most exciting developments for the advancement of prostate cancer therapy in recent years. Through the manipulation of senescence induction pathways we will identify PICS enhancing drugs and redefine the paradigm for cancer therapy. By developing novel mouse models that target prostate stem cells we will evaluate these PICS pro-senescence drugs in a pre-clinical setting. Finally, these results will be cross referenced with data from human prostate stem cells and we will lay the ground work to translate this to the clinical setting, further developing the clinical potential of these findings to eradicate prostate cancer.
Summary
This proposal aims to harness a novel type of senescence that we have identified in response to acute Pten inactivation, and which we believe offers a radical therapeutic approach to target the quiescent cancer stem cell in vivo. In characterizing Pten loss Induced Cellular Senescence, which we have named PICS for short, we have discovered that PICS is distinct from other forms of cellular senescence including oncogene-induced senescence (OIS) and replicative senescence. These distinct differences are characterized by a lack of DNA damage and hyper-replication, breaking the current dogma for senescence induction. The ability to induce senescence, an irreversible growth arrest, in cells by targeting Pten signaling, without a requirement for hyper-replication and DNA damage opens up the possibility to target quiescent cells, including stem cells, that have a low proliferative index. This approach has tremendous therapeutic potential and represents one of the most exciting developments for the advancement of prostate cancer therapy in recent years. Through the manipulation of senescence induction pathways we will identify PICS enhancing drugs and redefine the paradigm for cancer therapy. By developing novel mouse models that target prostate stem cells we will evaluate these PICS pro-senescence drugs in a pre-clinical setting. Finally, these results will be cross referenced with data from human prostate stem cells and we will lay the ground work to translate this to the clinical setting, further developing the clinical potential of these findings to eradicate prostate cancer.
Max ERC Funding
1 500 000 €
Duration
Start date: 2011-04-01, End date: 2016-03-31
Project acronym PROGEURO
Project Programming effects of early life stress exposure to bereavement on future health: a perspective in 6.7 million subjects in 3 European countries
Researcher (PI) Jiong Li
Host Institution (HI) AARHUS UNIVERSITET
Call Details Starting Grant (StG), LS7, ERC-2010-StG_20091118
Summary There have been significant challenges in the research areas of: 1. Foetal PROGramming. The widely use of growth variables as the indicators of foetal environment remains the major methodological limitation. And no research in humans has been able to examine the biomarkers at different programming stages from exposure itself to disease in one single study. 2. Stress. It remains difficult to assess stress and to obtain data on long-term health in a large study. The biological programming effects of prenatal stress need to be elucidated. 3. Bereavement. There is a significant knowledge gap in health of children bereaved by the death of a close relative. 4. Register-based research. To combine the multi-national data is necessary to understand the aetiology and the impact of rare disease and the effects of certain risk factors. But such a first attempt will face many obstacles.
The novel approaches in this study are designed to meet all the above challenges. The study uses data from 21 national databases in Denmark, Sweden, and Finland. The first component is a population-based cohort of 6.7 million children. Its objective is to examine the programming effects of an early stress exposure, bereavement during prenatal or early years in life, on a wide range of health outcomes. The second biological component is a proof of concept for foetal programming, examining biomarkers along the pathway from prenatal stress to disease.
The study is feasible only in EUROpean settings, which will strengthen the European leadership in epidemiology and public health. It may start a new era for joint European research in public health. The challenges may lead to difficulties and uncertainties for the study, which could also be the source of new scientific insights, hypotheses, and theories.
Summary
There have been significant challenges in the research areas of: 1. Foetal PROGramming. The widely use of growth variables as the indicators of foetal environment remains the major methodological limitation. And no research in humans has been able to examine the biomarkers at different programming stages from exposure itself to disease in one single study. 2. Stress. It remains difficult to assess stress and to obtain data on long-term health in a large study. The biological programming effects of prenatal stress need to be elucidated. 3. Bereavement. There is a significant knowledge gap in health of children bereaved by the death of a close relative. 4. Register-based research. To combine the multi-national data is necessary to understand the aetiology and the impact of rare disease and the effects of certain risk factors. But such a first attempt will face many obstacles.
The novel approaches in this study are designed to meet all the above challenges. The study uses data from 21 national databases in Denmark, Sweden, and Finland. The first component is a population-based cohort of 6.7 million children. Its objective is to examine the programming effects of an early stress exposure, bereavement during prenatal or early years in life, on a wide range of health outcomes. The second biological component is a proof of concept for foetal programming, examining biomarkers along the pathway from prenatal stress to disease.
The study is feasible only in EUROpean settings, which will strengthen the European leadership in epidemiology and public health. It may start a new era for joint European research in public health. The challenges may lead to difficulties and uncertainties for the study, which could also be the source of new scientific insights, hypotheses, and theories.
Max ERC Funding
1 449 950 €
Duration
Start date: 2011-01-01, End date: 2016-06-30
Project acronym ProstOmics
Project 'Tissue is the issue': a multi-omics approach to improve prostate cancer diagnosis
Researcher (PI) May-Britt Tessem
Host Institution (HI) NORGES TEKNISK-NATURVITENSKAPELIGE UNIVERSITET NTNU
Call Details Starting Grant (StG), LS7, ERC-2017-STG
Summary Overtreatment in prostate cancer (PCa) is a burden for health care economy and for quality of life. Correct diagnosis of early stage PCa is challenging given the limitations of the currently available clinical tools and the biological understanding of PCa. In this inter-disciplinary project, I propose an innovative approach enabling several cutting-edge ‘omics’ technologies (spatial metabolomics, genomics, transcriptomics) as well as histopathology to be performed on the same tissue sample. My goal is to reveal the molecular mechanisms of novel, but also promising metabolite biomarkers (citrate, polyamines, succinate and zinc) and their connection to recurrence, tissue heterogeneity and immune responses in complex human tissues. Such markers can personalize PCa diagnosis, open up new treatment strategies and fundamentally change the view of how to analyze tissue samples in the future. Furthermore, I want to demonstrate that citrate and polyamines are reliable prognostic markers that can be analyzed both in tissue and in patients in vivo by MR spectroscopic imaging. This work is made possible by the availability of high-quality fresh frozen tissue biobanks of prostatectomy biopsies with 5-10 years of follow-up data (N=1000)/slices (N=1000) and targeted in vivo snap-shot biopsies from clinical MR guided procedures (N=100). Among other techniques, I will implement high speed MALDI imaging (RapifleX MALDI TissueTyper) to the multi-omics protocol to study the spatial distribution and provide high resolution metabolic maps for each cell type, and which can be matched to both histopathology and MR Imaging. Multi-disciplinary platforms on large cohorts are needed to explore the clinical potential of the suggested molecular mechanisms. I expect that this ambitious proposal will address the diagnostic challenges of PCa and will further inspire the clinic and scientific community to follow the multi-omics approach within diagnosis and cancer research.
Summary
Overtreatment in prostate cancer (PCa) is a burden for health care economy and for quality of life. Correct diagnosis of early stage PCa is challenging given the limitations of the currently available clinical tools and the biological understanding of PCa. In this inter-disciplinary project, I propose an innovative approach enabling several cutting-edge ‘omics’ technologies (spatial metabolomics, genomics, transcriptomics) as well as histopathology to be performed on the same tissue sample. My goal is to reveal the molecular mechanisms of novel, but also promising metabolite biomarkers (citrate, polyamines, succinate and zinc) and their connection to recurrence, tissue heterogeneity and immune responses in complex human tissues. Such markers can personalize PCa diagnosis, open up new treatment strategies and fundamentally change the view of how to analyze tissue samples in the future. Furthermore, I want to demonstrate that citrate and polyamines are reliable prognostic markers that can be analyzed both in tissue and in patients in vivo by MR spectroscopic imaging. This work is made possible by the availability of high-quality fresh frozen tissue biobanks of prostatectomy biopsies with 5-10 years of follow-up data (N=1000)/slices (N=1000) and targeted in vivo snap-shot biopsies from clinical MR guided procedures (N=100). Among other techniques, I will implement high speed MALDI imaging (RapifleX MALDI TissueTyper) to the multi-omics protocol to study the spatial distribution and provide high resolution metabolic maps for each cell type, and which can be matched to both histopathology and MR Imaging. Multi-disciplinary platforms on large cohorts are needed to explore the clinical potential of the suggested molecular mechanisms. I expect that this ambitious proposal will address the diagnostic challenges of PCa and will further inspire the clinic and scientific community to follow the multi-omics approach within diagnosis and cancer research.
Max ERC Funding
1 500 000 €
Duration
Start date: 2018-02-01, End date: 2023-01-31
Project acronym QUIET
Project Health consequences of noise exposure from road traffic
Researcher (PI) Mette Sørensen
Host Institution (HI) KRAEFTENS BEKAEMPELSE
Call Details Starting Grant (StG), LS7, ERC-2011-StG_20101109
Summary There is growing public concern about adverse effects of traffic noise on health, as research has found that traffic noise increases the risk for cardiovascular diseases. Noise is thought to act as a stressor and disturbs sleep. Though this potentially could increase the risk for other major diseases, noise effects on other than the cardiovascular diseases are virtually unexplored.
The main objective of this project is to investigate if long-term exposure to road traffic noise is detrimental to various health outcomes in susceptible groups, i.e. children and elderly. Outcomes in children include low birth weight, infections and cognitive performance, and in elderly outcomes include diabetes, cancer, cancer survival, health-related quality of life and health behaviour.
The basis of this proposal is two unique Danish cohorts of, respectively, 57,053 elderly and 101,042 children (a national birth cohort). Historic and present residential addresses for all cohort members will be obtained through linkage with the nationwide Central Population Registry, and exposure to road traffic noise and air pollution will be calculated by validated models at all addresses.
The health outcomes will be obtained from cohort interviews/questionnaires or found through linkage with unique, nationwide, population-based health registers, such as the Danish National Hospital Registry, the Diabetes Registry and the Cancer Registry.
Data will be analysed using a number of statistical analyses depending on design and the character of the endpoint variable. All analyses will be adjusted for potential confounders such as air pollution, smoking and education.
Within the EU, 30% of the population lives at locations where the 55dB WHO noise limit is exceeded. Knowledge of harmful effects of noise is, however, limited. The results of the proposed research have a high potential to influence the content and time schedule of noise action plans in the EU member states.
Summary
There is growing public concern about adverse effects of traffic noise on health, as research has found that traffic noise increases the risk for cardiovascular diseases. Noise is thought to act as a stressor and disturbs sleep. Though this potentially could increase the risk for other major diseases, noise effects on other than the cardiovascular diseases are virtually unexplored.
The main objective of this project is to investigate if long-term exposure to road traffic noise is detrimental to various health outcomes in susceptible groups, i.e. children and elderly. Outcomes in children include low birth weight, infections and cognitive performance, and in elderly outcomes include diabetes, cancer, cancer survival, health-related quality of life and health behaviour.
The basis of this proposal is two unique Danish cohorts of, respectively, 57,053 elderly and 101,042 children (a national birth cohort). Historic and present residential addresses for all cohort members will be obtained through linkage with the nationwide Central Population Registry, and exposure to road traffic noise and air pollution will be calculated by validated models at all addresses.
The health outcomes will be obtained from cohort interviews/questionnaires or found through linkage with unique, nationwide, population-based health registers, such as the Danish National Hospital Registry, the Diabetes Registry and the Cancer Registry.
Data will be analysed using a number of statistical analyses depending on design and the character of the endpoint variable. All analyses will be adjusted for potential confounders such as air pollution, smoking and education.
Within the EU, 30% of the population lives at locations where the 55dB WHO noise limit is exceeded. Knowledge of harmful effects of noise is, however, limited. The results of the proposed research have a high potential to influence the content and time schedule of noise action plans in the EU member states.
Max ERC Funding
1 334 890 €
Duration
Start date: 2012-03-01, End date: 2017-02-28