Keynote speakers

Prof. Dame Molly Stevens, DBE FRS FREng (UK)

John Black Professor of Bionanoscience

University of Oxford

Kavli Institute for Nanoscience Discovery

Department of Physiology, Anatomy and Genetics

Department of Engineering Science

Designing and translating materials for advanced therapeutics and biosensing (virtual talk)

This talk will provide an overview of our recent developments in bioinspired materials for applications in advanced therapeutics and biosensing with focus on establishing translational pipelines to bring our innovations to the clinic [1]. Our group has developed fabrication methods to engineer complex 3D architectures that mimic anisotropic and multiscale tissue structures and generate spatially arranged bioinstructive biochemical cues [2]. I will discuss recent advances in our tunable nanoneedle arrays for multiplexed intracellular biosensing at sub-cellular resolution and modulation of biological processes [3]. We are developing creative solutions for targeted and controlled delivery using microrobots with unique bioinspired characteristics that respond to external stimuli to release a payload [4]. Our therapeutic delivery portfolio includes high molecular weight polymer carriers for enhanced delivery of saRNA therapeutics and photo-responsive nanoreactors inspired in the circadian rhythms [5]. We are exploiting the sensing capabilities of functionalised nanoparticles to engineer nanoprobes for in vivo disease diagnostics that produce a colorimetric response ideal for naked eye read-out and for CRISPR-based preamplification free detection of ncRNAs (CrisprZyme) which we have validated with cardiovascular disease patient samples [6]. I will present advances in Raman spectroscopy for high-throughput label-free characterization of single nanoparticles (SPARTA™) that allow us to integrally analyse a broad range bio-nanomaterials without any modification enabling exciting biosensing applications using extracellular vesicles as disease biomarkers, a growing area of interest in cardiovascular medicine [7]. Finally, I will explore how these versatile technologies can be applied to transformative biomedical innovations and will discuss our efforts in establishing effective translational pipelines to drive our innovations to clinical application while actively engaging in efforts towards the democratisation of healthcare [8].

[1] J. P. K. Armstrong… M. M. Stevens. “A blueprint for translational regenerative medicine.” Science Translational Medicine. 2020. 12(572): eaaz2253. [2] T. von Erlach, … M. M. Stevens. Nature Materials. 2018. 17: 237-242. [3] C. Chiappini… M. M. Stevens, E. Tasciotti. Nature Materials. 2015. 14: 532. [4] X. Song… M. M. Stevens. Advance Materials. 2022. 34(43): 2204791.; R. Sun… M. M. Stevens. Advanced Materials. 2022. 35(13):2207791. [5] A. Blakney, … M. M. Stevens. ACS Nano. 2020, 14(5): 5711-5727.; O. Rifaie-Graham, … M.M. Stevens. Nature Chemistry. 2023. 15: 110–118. [6] C. N. Loynachan, … M. M. Stevens. Nature Nanotechnology. 2019. 14: 883-890.; M. Broto, … M. M. Stevens. Nature Nanotechnology. 2022. 10: 1038. [7] J. Penders, … M. M. Stevens. Nature Communications. 2018, 9: 4256.; J. Penders, … M. M. Stevens. ACS Nano. 2021, 15, 11, 18192–18205; H. Barriga, … M. M. Stevens. Advanced Materials. 2021, 34(26):2200839. [8] A. T. Speidel, … M. M. Stevens. Nature Materials. 2022. DOI: 10.1038/s41563-022-01348-5.

Professor Dame Molly Stevens DBE FREng FRS is John Black Professor of Bionanoscience at the University of Oxford and also holds part-time professorships at Imperial College London and the Karolinska Institute. Molly’s multidisciplinary research balances the investigation of fundamental science with the development of technology to address some of the major healthcare challenges. She is a serial entrepreneur and the founder of several companies in the diagnostics, advanced therapeutics, and regenerative medicine space. Her work has been instrumental in elucidating the bio-material interfaces. She has created a broad portfolio of designer biomaterials for applications in disease diagnostics and regenerative medicine. Her substantial body of work influences research groups around the world (>450 publications, h-index 109, >50k citations, 2018, 2021, 2022 and 2023 Clarivate Analytics Highly Cited Researcher in Cross-Field research).

Prof. Dr. David W. Grainger (USA)

University Distinguished Professor and Department Chair of Biomedical Engineering, Ole and Marty Jensen Endowed Chair, School of Dentistry, and Distinguished Professor of Molecular Pharmaceutics, The University of Utah

Allogenic mesenchymal cell sheet properties in regenerative medicine strategies

Cell sheet tissue engineering is a clinically demonstrated scaffold-free regenerative approach applied in 45 registered clinical trials using a variety of cell types and diseases to date. Cell sheets are produced using commercial thermo-responsive cultureware that facilitates release of confluently cultured cells without proteolytic enzymes (e.g. trypsin) via moderate changes in culture temperature from 37°C to below 32°C. This process preserves endogenous extracellular matrix (ECM) and surface proteins that enable robust cell sheets to spontaneously adhere and integrate with biological surfaces without biomaterial supports.

We have used both human mesenchymal stem cells and differentiated juvenile and adult chondrocytes in different cell sheet designs and regenerative applications. Layered sheets of different cell types and sheets conditioned with various cytokine pretreatments im cultures have been characterized for regenerative and immune tolerance-inducing properties and implanted into preclinical models to assess safety and efficacy. This talk will highlight our recent progress in using scaffold-free cell sheets as regenerative and immuno-modulating tools.

References:
Hu, D. et al, The preclinical and clinical progress of cell sheet engineering in regenerative medicine. Stem Cell Res. Ther. 14, 112 (2023). Thorp, H. et al, Trends in articular cartilage tissue engineering: 3D mesenchymal stem cell sheets as candidates for engineered hyaline-like cartilage. Cells 10, 1–22 (2021). Kondo, M. et al, “Novel Therapies Using Cell Sheets Engineered from Allogenic Mesenchymal Stem/Stromal Cells,” (2020) Emerg. Top. Life Sci., ETLS20200151. Kondo, M. et al, Safety and efficacy of human juvenile chondrocyte-derived cell sheets for osteochondral defect treatment. npj Regen. Med. 6, 1–11 (2021).

David W. Grainger is a University Distinguished Professor and Department Chair of Biomedical Engineering, and Distinguished Professor of Pharmaceutics and Pharmaceutical Chemistry at the University of Utah, USA.  Grainger’s research focuses on improving cell regenerative therapy through allogenic approaches, implanted medical device and clinical diagnostics performance, and nanomaterials toxicity.  His research awards include the 2020 International Award from the European Society for Biomaterials, a 2016 Fulbright Scholar Award (New Zealand), the 2013 Excellence in Surface Science Award (Surfaces in Biomaterials Foundation), the 2007 Clemson Award for Basic Research (Society for Biomaterials), the 2005 American Pharmaceutical Research and Manufacturer’s Association’s “Excellence in Pharmaceutics” award, and 2019 Daniels Fund Award for Education in Research Ethics.  Grainger emphasizes translational approaches to clinical biomaterials, and validation of clinical effectiveness in implants and drug delivery systems for value-based medicine.

Prof. Dr. Suzie H. Pun (USA)

Washington Research Foundation Professor of Bioengineering and Director for the Molecular Engineering and Sciences Institute, University of Washington

Biomaterials for cancer immunotherapy applications

Biological systems have exquisite complexity, responsiveness, and interactions at multiple scales. We have developed synthetic materials inspired by nature to address unmet medical needs. In the first example, unique aptamers with high affinity for T cell markers were discovered and applied as alternatives to antibodies for T cell isolation in the manufacturing process for CAR T cells. In the second example, a polymer was developed that mimics the endosomal release mechanism of adenovirus, selectively displaying a membrane-disrupting peptide in acidic pH. This polymer promotes efficient endosomal release and has been used to deliver biologic drugs such as nucleic acids and peptides into the cell for applications such as cancer immunotherapy.

Suzie H. Pun is the Washington Research Foundation Professor of Bioengineering and Director for the Molecular Engineering and Sciences Institute at University of Washington. She is a fellow of the U.S. National Academy of Inventors (NAI) has been recognized with the Presidential Early Career Award for Scientists and Engineers and the University of Washington’s Marsha Landolt Distinguished Graduate Mentor Award for her dedicated mentoring of students. She currently serves as an Associate Editor for ACS Biomaterials Science and Engineering. Suzie Pun received her B.S. from Stanford University and her Ph.D. from the California Institute of Technology in Chemical Engineering.

Prof. Dr. Massimo Mastrangeli (NL)

Associate Professor, Electronic Components, Technology and Materials

Delft University of Technology

Microelectromechanical organs-on-chip

Organ-on-chip (OoC) technology is rapidly being established as a valid approach to develop in-vitro models of human (patho)physiology of unprecedented relevance. Advances in the technology involve co-development of the biological substrates and the design of supporting hardware enabling eminently microfluidic perfusion, electro-mechanical actuation and electro-chemical sensing.

In this talk, I will introduce the perspective of fully electro-mechanical OoCs devices and of OoC platforms. I will argue that virtually all relevant functions in OoCs can be driven and controlled electro-mechanically, and exemplify how this is best achieved by a seamless integration of electric and fluidic layers in the architecture of the platforms. These efforts are meant to foster ease of use, wider adoption and superior reproducibility of OoC technology.

Massimo Mastrangeli is Associate Professor in the Electronic Components, Technology and Materials (ECTM) group of the Microelectronics department of Delft University of Technology (Delft, NL), where he is developing innovative microelectromechanical organ-on-chip devices and platforms. He got his BSc and MSc degrees cum laude in Electronic Engineering from University of Pisa (IT), and his PhD degree in Materials Engineering from University of Leuven (BE). He held research positions at École Polytechnique Fédérale de Lausanne (EPFL, CH), Université Libre de Bruxelles (BE) and Max Planck Institute for Intelligent Systems (Stuttgart, DE). Massimo is also Guest Lecturer at EPFL and Board Member of the European Organ-on-Chip Society (EUROoCS).

Prof. Dr. Chantal Pichon (FR)

Professor, ART-ARNm  Innovative therapies and Nanomedicine, INSERM University of Orleans

Messenger RNA vaccines and therapeutics

The perspective of using messenger RNA (mRNA) as a therapeutic molecule has first faced some uncertainties due to concerns about its instability and the feasibility of large-scale production. The potential of messenger RNA (mRNA)-based vaccines has been revealed by the success of rapid and adaptable vaccination strategies to fight against COVID-19 pandemic. The achievement of those mRNA vaccines has been made possible through advances in the design of mRNA structure, manufacturing and delivery systems. This success opens up an avenue for the development of innovative mRNA-based therapeutics and vaccines envisioning different applications in immunotherapy, regenerative medicine and gene editing. I will present the key milestones that have led to the production of these vaccines. Current knowledge regarding crucial aspects-structure, stability, formulations, impact of delivery systems and targeting on mRNA translation as well as in vivo mRNA applications will be summarized. Last, I will also present challenges that have to be tackled to fully prove its mettle and to potentiate mRNA therapeutic applications.

Chantal Pichon has an established professorship at the University of Orleans (France). She is leading an  R&D laboratory of INSERM, the French national research institution dedicated to health research. She holds also an Innovation chair of the Institut Universitaire de France (Paris) and an invited professorship at La Charité  Berlin Health Institute funded by Stiftung Charité. C. Pichon is conducting interdisciplinary projects based on chemistry and molecular and cell biology with a crosstalk between basic and applied researches. Her main research activities are dedicated to the use of nucleic acids as therapeutics, especially messenger RNAs. Her lab is developing innovative formulations characterized by the presence of imidazole/imidazolium ring in the lipid or the polymer. They are used for various applications: mucosal vaccination, imune cell-based therapies and protein replacement therapy. C. Pichon has a track-record of 200 articles and 12 filled patents. She obtained 35 academic and private grants including Horizon Europe, FP7, ANR, among others.

Prof. Dr. Khalid Salaita (USA)

Samuel Candler Dobbs Professor of Chemistry and Director for Graduate Studies, Emory University

DNA mechanotechnology: nucleic acids that sense and generate molecular forces enable a new class of biomedical applications and diagnostics

Modern machines, which are composed of force-generating motors, force sensors, and load-bearing structures, enabled the industrial revolution and are foundational to human civilization. Miniature micromachines are used in countless devices including cell phone microphones, implantable biosensors, and car and airplane accelerometers. Further miniaturization to the nanometer scale would enable the design of machines that can manipulate biomolecules and other nanomaterials for applications in medicine, biological research, and material development. Such machines are typically difficult or impossible to build because of their small size.

However, powerful synthetic methods to assemble and modify nucleic acids combined with single molecule force spectroscopy studies have propelled the emergence of a subfield that we call “DNA mechanotechnology”. DNA mechanotechnology is particularly well suited for measuring and controlling molecular forces at the scale of piconewtons (pN). For context, 10 pN is roughly one-billionth the weight of a paper clip, and ~4 pN applied a distance of 1 nm equals the work of 1 kT. Mechanical forces on this scale are important in diverse areas including molecular biophysics, immunology, regenerative medicine, materials science, and nanorobotics. In this talk, I will discuss my group’s efforts at using DNA mechanotechnology for new types of viral diagnostics and in biomedical applications relating to the field of mechanobiology and immunology.

Khalid Salaita is the Samuel Candler Dobbs Professor of Chemistry and Director for Graduate Studies at Emory University. His lab currently investigates the use of nucleic acids as molecular force sensors, smart drugs, and synthetic motors. In recognition of his independent work, Khalid has received a number of awards, most notably: the Alfred P. Sloan Research Fellowship, the Camille-Dreyfus Teacher Scholar award, the NSF Early CAREER award, the Kavli Fellowship, and Merck Future Insight Prize. Khalid is currently the director of the Center on Probes for Molecular Mechanotechnology, and an Associate Editor of SmartMat.

Prof. Dr. Carole Bourquin (CH)

Full Professor, Pharmacology, University of Geneva

Nanomaterials for drug delivery in cancer and inflammation

Engineered nanoparticles have become essential tools in therapeutic and diagnostic applications, particularly for enhancing drug delivery. This presentation discusses recent advances using polymer-coated gold nanoparticles (GNPs) and multifunctional mesoporous silica nanoparticles (MSNs) to target immune cells in order to modulate immune responses.


Our research focused on MSNs as pH-responsive carriers for an anticancer immuno-stimulant. Equipped with a biotin-avidin cap, MSNs efficiently released the drug in acidic environments such as the endosome of phagocytic cells, thus facilitating activation of antigen-presenting cells. In vivo studies demonstrated enhanced dendritic cell activation and improved antigen-specific T-cell responses, by optimizing the pharmacokinetic profile of R848 and reducing systemic exposure.


Additionally, we explored various porous nanoparticles for delivering the hydrophobic anti-inflammatory drug necrosulfonamide to macrophages. Effective delivery and suppression of proinflammatory cytokine secretion were achieved.


Our work on polymer-coated GNPs revealed specific interactions with age-associated B cells without affecting other B cell populations or inducing innate-like immune responses. These findings pave the way for potential clinical applications for targeting age-associated B cells to treat B-cell-mediated autoimmune diseases.

These studies collectively highlight the potential of engineered nanoparticles for targeted immune modulation, presenting promising strategies for treating inflammatory diseases and enhancing cancer immunotherapy.

Carole Bourquin is full professor of Pharmacology at the University of Geneva. She heads the group of Immunopharmacology of Cancer at the Institute of Pharmaceutical Sciences of Western Switzerland. Her research focuses on uncovering mechanisms that control immune activation in cancer, in order to improve the efficacy of cancer immunotherapy in patients. Prof. Bourquin addresses these questions using translational approaches that range from preclinical cancer models to patient-oriented research. An important aspect of her work is the use of nanodelivery systems to target immunomodulatory drugs to their site of action. Prof. Bourquin is also a practicing clinical pharmacologist at the Geneva University Hospital.

Dr. Gregoire Altan-Bonnet (USA)

Senior Investigator and Deputy Chief, Laboratory of Integrative Cancer Immunology, National Cancer Institute, National Institutes of Health (NIH)

Building models of CAR-T signal integration, using automatized/dynamic high-dimensional dynamic profiling

We present an experimental/theoretical pipeline to build quantitative models of leukocyte activation. We introduce a robotic platform to quantify the dynamics of cell differentiation and cytokine production/consumption by T cells ex vivo. These high-dimensional dynamics can be compressed into a 2D model using tools from machine learning. Our model highlights two modalities of T cell activation that enforce adaptive kinetic proofreading of antigen-TCR interactions, and that encode antigen discrimination. We test our model of antigen discrimination across varied immunological settings, including CAR-T and signaling-impaired T cells. To conclude, we highlight the power of lab automation, data integration, machine learning and theoretical modeling to usher new insights in systems immunology.

I have been trained in Statistical Physics and nonlinear dynamics (PhD) and in Immunology (post-doctoral studies). My field of expertise is Systems Immunology. The ImmunoDynamics group I have been heading since 2005, first at Memorial Sloan Kettering and at the NCI since 2016, has been developing experimentally validated quantitative models of different aspects of the immune response. In particular, we have addressed the interplay between the robustness and variability of self/non-self discrimination, as well as the bridging of local and global cytokine regulations in the immune system. We are currently focused on developing quantitative models of leukocyte-leukocyte communications within the cytokine network and exploring applications of machine learning to the field of cancer immunology and virology.

Prof. Dr. Joachim Spatz (DE)

Director, Max Planck Institute for Medical Research; Founding Director, Institute for Molecular Systems Engineering and Full Professor of Biophysical Chemistry, Heidelberg University

Matter to Life: Bottom-up assembly of synthetic cells

The evolution of cellular compartments for spatially and temporally controlled assembly of biological processes was an essential step in developing life by evolution. Synthetic approaches to cellular-like compartments are still lacking well-controlled functionalities, as would be needed for more complex synthetic cells. With the ultimate aim to construct life-like materials such as a living cell, matter-to-life strives to reconstitute cellular phenomena in vitro – disentangled from the complex environment of a cell. In recent years, working towards this ambitious goal gave new insights into the mechanisms governing life. With the fast-growing library of functional modules assembled for synthetic cells, their classification and integration become increasingly important. We will discuss strategies to reverse-engineer and recombine functional parts for synthetic eukaryotes, mimicking the characteristics of nature’s own prototype. Particularly, we will focus on large outer compartments, complex endomembrane systems with organelles and versatile cytoskeletons as hallmarks of eukaryotic life. Moreover, we identify microfluidics and DNA nanotechnology as two highly promising technologies which can achieve the integration of these functional modules into sophisticated multifunctional synthetic cells.

Prof. Dr. Joachim Spatz is Director of the Max Planck Institute for Medical Research. He is the Founding Director of the Institute for Molecular Systems Engineering and Full Professor of Biophysical Chemistry at Heidelberg University. His research encompasses the fields of cellular biophysics, materials science, cell biology, interface science, and physics of soft matter. The primary scientific goal of the Spatz group is to develop technologies, based on physics, chemistry and materials science, for unraveling fundamental problems in cellular science, as well as to construct life-like materials. This includes pathophysiology of cells and cell cohorts by analyzing and manipulating cells on the nanoscale; how to bottom-up assemble synthetic cell functions and materials; the role of growth factors in cellular mechanobiology; and the role of polysaccharides of the extracellular matrix in regulating cell fate. Dr. Spatz earned his Ph.D. in Physics at Ulm University. He is the author of over 600 publications and has received numerous prestigious awards including his recent appointment as a member of the Leopoldina National Academy of Sciences.


Invited speakers

Dr. Inge Wortel (NL)

Assistant Professor, Data Science, Institute for Computing and Information Sciences, Radboud University

When motility matters: simulating host-pathogen interactions during Listeria invasion of the gut epithelium

Listeria monocytogenes (Lm) is a food-borne bacterium that causes severe gut infections, leading to hospitalization or even death. Human infections typically occur with Lm growing at or below room temperature, which is highly motile – yet it remains unclear how such motility affects invasion and immune evasion dynamics.
We addressed this question using complementary imaging and computational models. Imaging Lm interacting with mouse and human intestines, we found that motility facilitated rapid scanning of the gut epithelium, allowing Lm to locate goblet cells permissive to invasion. When we built an in silico model integrating data from various imaging experiments, it consistently predicted that motile bacteria invade twice as efficiently over the first hour of infection. Intriguingly, our imaging experiments also revealed neutrophils interacting with Lm on the apical epithelial surface within hours after infection, suggesting that neutrophils might help limit bacterial burden. Yet our models and data challenge the prevailing view that neutrophils “hunt” for bacteria, instead suggesting that bacterial motility is key in driving neutrophil-Lm interactions, with Lm moving an impressive 50x faster than neutrophils. In conclusion, our findings reveal bacterial motility as a critical factor in host-pathogen interaction dynamics during infection with highly motile bacteria such as Lm.

After a journey through the fields of Chemistry (BSc, 2014), Molecular Mechanisms of Disease (MSc, 2016) and Computational immunology (PhD, 2021), Inge Wortel’s research interest is how tools from across disciplinary boundaries can help us make sense of the complexity of our immune system. How can we get to the essence of dynamical processes in immunology, translating complex datasets into simple insights?

Inge was recently awarded an AiNed Fellowship grant to build a group in the Data Science department at Radboud University (the Netherlands), where she and her team combine simulation models and AI to uncover the when, where, and how of key events in immunoimaging data. In her work, Inge takes a “less is more” approach to build simple models that are “wrong but useful” in debugging our understanding of immune dynamics. Her talk at BIC will showcase how such a simulation model helps pinpoint the key factors governing host-pathogen dynamics in the gut.

Dr. Nicholas Reynolds (AU)

Head, Self-assembled Nanomaterials Lab, La Trobe Institute for Molecular Sciences

Self-healing self-assembled peptide based Bioinks for 3D printing applications

Self-assembled peptides that spontaneously form networks of nanofibrillar materials make attractive candidates for 3D biomaterials as they can form hydrogels that possess extracellular matrix mimicking morphologies, they can be readily synthesised and purified to high levels, and short sequences can be added to improve cellular responses. Typically, such peptide-based hydrogels are held together by weak non-covalent interactions which makes them soft, and unable to withstand significant shear forces without breaking. Thus, self-assembled peptide-based materials are generally considered not to be attractive materials for 3D bioprinting applications. Here I will present recent results from my group, where we have combined a variety of self-assembled peptides with natural polymers to make cell laden hybrid materials that are highly printable (termed bioinks). The bioinks can support the growth of a variety of different cell types, have self-healing properties and have tunable mechanical properties yet remain easily printable even at high stiffnesses. During the development of these bioinks we made several serendipitous discoveries. These discoveries include the fabrication of 3D printable self-propelling particles with potential applications in biosensing, and the collaborative development of a simple plasmonic device to detect collagen deposition from mesenchymal stem cells encapsulated within hydrogels without the need for histological labelling or second harmonic generation (SHG) microscopy. Thus, in addition to presenting our materials we will discuss these exciting discoveries.

Dr. Reynolds is the head of the self-assembled nanomaterials lab at the La Trobe Institute for Molecular Sciences (LIMS) in Melbourne, Australia. He obtained his PhD from the University of Sheffield, and has held positions at the University of Zurich, CSIRO and Swinburne University of Technology. Dr Reynolds’ interdisciplinary research focuses on the design, discovery, characterisation and biological interactions of self-assembled proteins and peptides. He works closely with academics, industry and clinicians on projects spanning biomaterials and understanding disease. Since 2020 Dr. Reynolds has become interested in uncovering the roles of amyloid protein assemblies in viral infections, including COVID-19.

Prof. Dr. Dagmar Iber (CH)

Associate Professor, Department of Biosystems Engineering, ETH Zürich

Morphogenesis of biological interfaces – from 3D cell shapes to 3D tissue shapes

Virtually all biological surfaces are lined by epithelia. Their structural properties are key to the morphogenesis of animals and the integrity of organisms. At the same time, epithelia play a key role in many diseases with more than 90% of all cancers originating from epithelia.

 

In my presentation, I will focus on the physical principles by which cells organise into epithelia and present examples of epithelial morphogenesis in development and disease. To define those physical principles my team develops bespoke computational algorithms and software to leverage data-based computational modelling based on 3D cell and tissue geometries obtained from image segmentation.

Dagmar Iber studied mathematics and biochemistry in Regensburg, Cambridge, and Oxford. She holds Master degrees and PhDs in both disciplines. After three years as a Junior Research Fellow in St John’s College, Oxford, Dagmar became a lecturer in Applied Mathematics at Imperial College London. Dagmar has joined ETH Zurich in 2008 after returning from an investment bank where she worked as an oil option trader for one year.

Dagmar Iber is interested in the principles of self-organisation that transform the linear genetic information contained in animal genomes into complex three-dimensional tissue structures. Her group develops data-based, predictive models to understand the spatio-temporal control of morphogenesis, as well as computational algorithms and simulation frameworks that enable efficient image segmentation, geometry extraction, and the use of geometries and computational models in simulations of morphogenesis at cellular resolution.

Dr. Tilo Netzer (CH)

Head of Commercial Development, Business Unit mRNA, Lonza AG

Optimizing GMP manufacturing of mRNA lipid nanoparticles

mRNA has a lot of potential not only for development of new vaccines but also for development of new therapeutics, e.g. as cell and gene therapies. However, mRNA has two challenges when used as a medicinal product: First, it is prone to degradation by RNAses and second it cannot easily enter into cells because it is large and immunogenic. Thus, most companies developing mRNA medicines encapsulate it into lipid nanoparticle (LNPs) which provides protection, minimizes innate immune activation and facilitates cellular uptake and cytoplasmic release. For encapsulation of mRNA into LNPs the T-mixing approach is commonly used which has been proven to work well for large scale manufacturing of mRNA-LNPs during the Covid-19 pandemic. However, these systems require substantial development work for upscaling from small research scale to clinical and later commercial cGMP scale. Technologies based on microfluidics are easy to set-up for the generation of small research amounts but require expensive single-use materials for cGMP manufacturing. Most recently a mixing technology using an oscillating stream has become available for mRNA encapsulation into LNPs. This technology offers a fast and robust scale-up while providing easy cleaning thus avoiding expensive single-use materials. In the Lonza mRNA Excellence Center in Geleen, NL, we performed a study investigating the influence of various parameters such as flow rates, working concentrations and sizes of the mixing chip on the quality of the manufactured mRNA-LNPs. The results of this study will be presented confirming that this new technology is suitable and beneficial for manufacturing of high quality mRNA-LNPs under GMP conditions in an economic process.

Tilo Netzer is Head of Commercial Development in the Business Unit mRNA at Lonza AG in Basel, Switzerland. In this function he is responsible for building the service offering for Lonza clients such as biopharma companies in the area of mRNA development and manufacturing. This includes the encapsulation of the mRNA in LNPs. He is in this position since middle of 2020 when Lonza started manufacturing of Moderna’s Covid-19 vaccine Spikevax®. Before that time Tilo was head of Preclinical Development at Lonza. Earlier in his career he spent more than 20 years at the German pharma company Merck Serono in different roles in clinical development and at the Pharma consulting company PharmaLex. Tilo is a pharmacist by training and has a PhD in Pharmacology.

Dr. Bart Spee (NL)

Associate Professor , Utrecht University

Liver adult stem cells for advanced in vitro models and whole organ engineering

Liver diseases cause approximately 2 million deaths annually worldwide, and the only effective treatment for end-stage liver disease is liver transplantation. There is also a need for long-lived hepatic in vitro models to better predict drug-induced liver injury. Human liver-derived epithelial organoids are a promising cell source for advanced in vitro models and whole organ engineering. These organoids are bipotential, can be expanded into large numbers of cells with stable genomic profiles, and can differentiate into hepatocyte- and cholangiocyte-like cells in vitro, making them suitable for liver tissue engineering.

 

The presentation will cover basic organoid technology and the benefits of combining organoids with biofabrication techniques. These novel technologies hold great potential for designing in vitro models with complex, customizable architectures. When combined with vasculature, these bioprinted constructs could be used for transplantation. Overall, the bioprinted constructs are viable, recapitulate native liver architecture, and maintain hepatic function. They can be used for testing novel drugs or personalized medicine approaches. In the future, these bioprinted constructs could potentially reduce the waiting list for liver transplants worldwide.

Dr. Spee has more than 20 years’ experience in Molecular Biology, and is (co)author of over 100 peer-reviewed scientific publications. Following a Ph.D. in Molecular Biology on pathways of regeneration and fibrosis of liver diseases, he took up a position as post-doc at the department of pathology at the University of Leuven, Belgium, under guidance of prof. Roskams. Here he used molecular tools combined with the vast tissue bank at the pathology department, resulting in an increased knowledge on the activation of the adult stem cells of the liver including its neoplastic offspring. After an internship at the Laboratory of Experimental Carcinogenesis (NIH, Bethesda, US) with Dr. Thorgeirsson where he worked on cholangiocarcinoma’s, he returned to Utrecht University as an associate professor. Currently he is investigating the use of stem cells including adult stem cells (organoids) for functional recovery of liver diseases. One of the focus points of his research is the creation of physiologically relevant in vitro liver models using biofabrication technology that can be used for drug toxicity testing and personalized medicine approaches. This biofabrication technology is also explored for the development of Advanced Therapy Medicinal Products (ATMPs) such as bioengineered livers as an alternative to organ donation.


Contributed talks

Dr. Jayesh Kulkarni (CA)

Chief Scientific Officer, NanoVation Therapeutics

Extrahepatic nucleic acid delivery enabled by extended pharmacokinetics of lipid nanoparticles

Lipid nanoparticles (LNPs) have demonstrably overcome the in vivo delivery barrier for manipulating targets in the liver; the first-ever approved RNAi therapeutic, Onpattro, was designed to deliver siRNA into the cytosol of hepatocytes. This LNP was designed to rapidly accumulate in the liver following intravenous administration, which limited access to extrahepatic tissues. Numerous methods have been applied to overcome this rapid liver accumulation, including the use of PEGylation (persistent PEG-lipids), modifying the particle surface by conjugation of ligands, or the use of charged lipids; all of which suffer from limited clinical utility due to the stimulation of an adaptive immune response, manufacturing complications, or acute immune stimulation, respectively.

 

Inspired by the development of long-circulating liposomes for small molecule therapeutics, we rationally designed a novel set of LNP formulations to deliver nucleic acids to extrahepatic tissues by extending their circulation time. First, we demonstrated that our formulations display extended circulation profiles, five-fold greater than benchmark LNPs, and exhibit a substantially different morphology. Next, we demonstrated that these novel LNP systems are nucleic acid agnostic with improved extrahepatic delivery over a range of tissues. These findings demonstrate that LNP tissue accumulation can be modulated through compositional change alone without the requirement for additional components, and support the hypothesis that extended circulation times can achieve extrahepatic delivery.

Dr. Kulkarni obtained his PhD from the University of British Columbia and has over 12 years of academic and industry experience in the nanoparticle drug delivery field. He has published over 40 peer-reviewed articles in prestigious journals and co-inventor on numerous patents. Dr. Kulkarni’s research has focused on the role of the various lipid components in LNP and the biophysics that governs particle formation. His work has contributed to clinical translation, including scale-up and manufacturing of LNP systems in accordance with GLP and GMP regulations. Dr. Kulkarni is a leader in the design and development of lipid nanoparticle (LNP) formulations of small molecule and nucleic acid therapeutics. He currently serves as the Chief Scientific Officer of NanoVation Therapeutics, an LNP-RNA formulation developer.

Dr. Bogdan Mateescu (CH)

Group Leader, University of Zürich

Decoding cell-free RNA: Potential for RNA diagnostics and therapeutics

Extracellular RNAs (exRNAs) in biofluids are promising not only as biomarkers for various diseases but also as models for improving therapeutic RNA delivery. As part of the PRISM project, our research focuses on the role of microvesicles and RNA-binding proteins (RBPs) in the stabilization and transport of exRNAs. Utilizing advanced bioinformatics and genetic approaches, we identified novel RBPs with detectable footprints in cell supernatant and human plasma, associated with extracellular vesicles (EVs) and non-EVs carrier classes.

 

To further validate the role of RBPs in exRNA biology, we created a comprehensive atlas by sequencing RNA from cells and conditioned medium of dozens of RBP, revealing their crucial roles in the biogenesis and extracellular stability of specific exRNA biotypes. Additionally, we developed novel pull-down assays, achieving high recovery and purity of specific exRNAs from low-volume biofluid samples. In collaboration with the deMello lab (ETHZ), we also developed microfluidic platforms for isolating cellular compartments and nanosized extracellular vesicles from biofluids.

 

Our new technology can significantly improve the sensitivity and specificity of exRNA-based biomarker assays, enabling more accurate disease detection and monitoring. Furthermore, our enhanced understanding of RNA-stabilizing carriers in biofluids provides valuable insights that can inspire the development of more effective therapeutic RNA delivery systems.

Bogdan Mateescu is a Group Leader at the University of Zürich (UZH), investigating extracellular RNAs in biofluids to develop innovative RNA diagnostics and therapeutics. He earned his Ph.D. from the Pasteur Institute and completed post-doctoral training at Institut Curie, focusing on RNA modulators of ovarian cancers. Since 2013, he has led research in Zürich and is affiliated with the NIH extracellular RNA communication consortium. His group has made key discoveries on RNA-stabilizing carriers in biofluids, such as microvesicles and RNA-binding proteins. He recently joined the UZH innovation hub as an entrepreneur, aiming to translate his research into marketable non-invasive diagnostics.

Dr. Ekaterina Umnyakova (CH)

Researcher, Molecular Pharmacy Group, Department of Pharmaceutical Sciences, University of Basel

Complement modulation on biosurfaces: Click chemistry approach for natural regulators recruitment

Umnyakova1, J. Felsch1 , S. Jaquemai1 , D. Ricklin1

1 Molecular Pharmacy Group, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland.

 

INTRODUCTION: As essential part of innate immunity, the complement system recognizes non-self biomaterials and generates pro-inflammatory effectors to eliminate potential threats. These undesirable defence reactions are enabled by the absence of complement regulators on such surfaces. One option to mitigate adverse complement activation is the recruitment of the abundant natural regulator factor H (FH) to the biomaterial surface using FH-binding peptides. Previously, our group described a cyclic peptide, termed 5C6, as a potent tool capable of effectively recruiting FH via complement control protein domains 5-18 [1] and impair the deposition of complement activation products (C3b/iC3b) on tested surfaces [2]. The goal of this project is to use a click chemistry approach for decorating biosurfaces (e.g., agarose, HMEC-1 endothelial cells) with 5C6 and thus, modulate complement activation diminishing uncontrolled complement-mediated effects.


METHODS: We used alkyne-azide click chemistry to covalently tether 5C6 peptides to the surface. 5C6 peptides were synthesized with the addition of an alkyne/strained alkyne (BCN) group to the N-terminus for the binding to active azido moieties of the target surface. In one example, we used commercially available azido agarose beads as material surface. Upon coating the alkyne-5C6 peptide by click reaction, we accessed FH binding using western blotting. In another setting, we employed metabolic glycoengineering to modify HMEC-1 cell surfaces. Active azido groups were introduced into sialic acid by growing cells in the presence of Ac4ManNAz (Fig.1).

Flow cytometry method was used to assess the ability of «clicked» 5C6 to recruit FH on the surfaces of HMEC-1 cells and the deposition of complement-derived opsonins C3b/iC3b.

Fig. 1: Simplified scheme of experiments


RESULTS: Azido-agarose matrix coated with 5C6 via click chemistry was able to effectively and selectively bind FH from normal human serum. On HMEC-1 cells, the azido sugar was successfully incorporated on cell surface glycans. Both the FH-recruiting and complement-modulating capacities of 5C6 coatings on the cell surface were confirmed by flow cytometry, whereas a sequence-scrambled control peptide showed no activity.


DISCUSSION & CONCLUSIONS: Our results show that click chemistry-based coating of FH-binding peptides may confer a promising strategy for preventing biomaterial-induced immune reactions. This approach potentially allows to modify any biomaterial surfaces, including lipid nanoparticles, liposomes, and filter materials, which are known to trigger complement activation.


REFERENCES: 1 Y.-Q. Wu, et al (2011) J Immunol 186(7):4269-77. 2 C. Bechtler, et al. (2022) Acta Biomater 155:123-38


ACKNOWLEDGEMENTS: This work was supported by intramural funds of the University of Basel and by the Swiss National Science Foundation (grants No. 31003A_176104 and 310030_219969).

Ekaterina Umnyakova currently works at Basel University in the group of Prof.Daniel Ricklin. She studies FH-binding peptides as a potent tools for complement regulation on different biomaterials, e.g. liposomes, polymers, cell surfaces, etc.
Earlier she was working at the department of general pathology and pathological physiology of Institute of Experimental Medicine (Saint Petersburg, Russia) for 12 years studying interactions between antimicrobial peptides and complement proteins and modulation of complement activity with peptides.

Dr. Céline Labouesse (CH)

Researcher, Macromolecular Engineering Laboratory, Institute of Energy and Process Engineering, ETH Zürich

Granular biomaterials as bioactive sponges for the sequestration and release of signaling molecules

B. Emiroglu1 2, A. Singh1, B. Marco-Dufort1, N. Speck1, P. G. Rivano1, J. Oakey3, N. Nakatsuka4, A. J. deMello2, C. Labouesse1, and M. W. Tibbitt1 *

1Macromolecular Engineering Laboratory, Department of Mechanical and Process Engineering, ETH Zurich, Sonneggstrasse 3, 8092 Zurich, Switzerland

2 deMello Laboratory, Department of Chemical and Bioengineering, ETH Zurich, Vladimir-Prelog-Weg, 1-5/10, 8093 Zurich, Switzerland

3Department of Chemical & Biological Engineering, University of Wyoming, 1000 E. University Ave., Laramie, WY, USA

4Laboratory of Biosensors and Bioelectronics, Department of Mechanical and Process Engineering, ETH Zurich, Sonneggstrasse 3, 8092 Zurich, Switzerland

 

INTRODUCTION: A major challenge for the regeneration of chronic wounds is an underlying dysregulation of signaling molecules, including inflammatory cytokines and growth factors. To address this, we propose to use granular biomaterials composed of jammed microgels1, to enable the rapid uptake and delivery of biomolecules, and provide a strategy to locally sequester and release biomolecules2.

 

METHODS: Granular hydrogels were made from 4-arm poly(ethylene glycol) with Norbornene (NB) end groups. These were linked with thiol-functionalized linker molecules using thiol-ene chemistry. Affinity ligands were conjugated using the same chemistry. Various biomolecules were used for release and sequestration assays, measured by enzyme linked immunosorbent assay (ELISA). Cellular response were measured using reporter cell lines.

 

Fig. 1: The granular biomaterial is a modular granular biomaterial composed of individual microgels which have the capacity to sequester or release biomolecules to normalize the signaling environment.

 

RESULTS: Sequestration assays on model biomolecules of different sizes demonstrated that granular hydrogels exhibit faster transport than comparable bulk hydrogels due to enhanced surface area and decreased diffusion lengths. To demonstrate the potential of modular granular hydrogels to modulate local biomolecule concentrations, we engineered microgel scaffolds that can simultaneously sequester excess pro-inflammatory factors and release pro-healing factors. To target specific biomolecules, microgels were functionalized with affinity ligands that bind either to interleukin 6 (IL-6) or to vascular endothelial growth factor A (VEGF-A). Finally, disparate microgels were combined into a single granular biomaterial for simultaneous sequestration of IL-6 and release of VEGF-A.

 

DISCUSSION & CONCLUSIONS: Overall, we demonstrate the potential of modular granular hydrogels to locally tailor the relative concentrations of pro- and anti-inflammatory factors2.

 

REFERENCES:

  1. D. B. Emiroglu et al. (2022) Building block properties govern granular hydrogel mechanics through contact deformations. Science Advances 8, eadd8570
  2. D.B. Emiroglu, Dilara Börte et al. (under revision) Granular hydrogels as bioactive sponges for the sequestration and release of signaling molecules.

 

ACKNOWLEDGEMENTS: The authors thank the Ehrbar group (University of Zurich) for their kind donation of HUVEC-GFP cells. ITC was performed on the equipment of Prof. Nyström’s lab (ETH Zurich), with the support of Dr. Cristina Lupo. This work was supported by ETH Zürich the Helmut Horten Stiftung, and ETH Open project SKINTEGRITY.CH.

The focus of my research is on cell-material interactions, specifically on how mechanical and physical environment influences cell phenotype and cell function. I use biomaterials to build 3D cell scaffolds that mimic the extracellular matrix. I apply these to investigate 3D mechanotransduction of fibroblasts. In addition, I use biomaterials to interface with living tissues, for delivery and sequestration of biomolecules.

Beatriz Moura (PT)

PhD Candidate, Medical Biotechnology, Department of Chemistry, University of Aveiro

Glycoengineered bio-clickable spheroidal units for generating hierarchical living materials

Beatriz S. Moura1, Vítor M. Gaspar1, João F. Mano1

1 Department of Chemistry, CICECO – Aveiro Institute of Materials, University of Aveiro, Campus Universitário de Santiago, 3810-193, Aveiro, Portugal.

 

INTRODUCTION: Spheroids have emerged as valuable platforms in disease modeling and bottom-up tissue engineering applications.[1] Despite major advancements, the assembly and tunability of spheroids remains largely restricted by their random cell aggregation. Uncoupling this random aggregation and cell density from the spheroids’ assembly process can unlock a higher control in size and downstream processability. Moreover, challenges persist in ensuring both their reproducibility and size uniformity. For this purpose, cell surface modification toolboxes are valuable to control cellular coupling and spheroids’ formation in chemistry-controlled time scales, and compatible with biological processes.[2]

 

METHODS: To assemble bio-clickable spherically structured living units, a diverse array of cell types, including stem cells, endothelial cells and cancer cells underwent metabolic glycoengineering with chemically functionalized non-natural mannosamine monosaccharides bearing azide groups. These glycoengineered cells were then suspended in a DBCO-modified extracellular matrix (ECM)-based polymer, and consistent volumes were dispensed onto a superhydrophobic surface. (Fig.1)

 

Fig. 1: Metabolic glycoengineering of cells towards producing bio-clickable living units.

 

RESULTS: Bio-clickable living units can be rapidly assembled and offered improved handleability, as well as heightened cellular viability. This innovative technology was successfully applied across different cellular units, including stem cells, endothelial and cancer cells, showcasing its applicability across diverse biological contexts. Conventional spheroids and beads across the different cell types were compared, revealing that while spheroids exhibit size dependency on cell number, our platform demonstrates rapid assembly and maintains a consistent size independent of cell number. (Fig. 2) The developed beads also displayed self-healing capabilities, allowing for the assembly of intricate multicellular constructs, opening new avenues for the development of complex tissue models. The fabricated constructs showed structural integrity and evolvability over extended culture periods, providing also a highly resilient and handleable platform.

 

Fig. 2: Quantification of living units’ area across different cell densities (n=10). Representative widefield microscopy images of living units (day 0) across the three different cell densities.

 

DISCUSSION & CONCLUSIONS: Beyond providing a faster, more efficient and controllable alternative to spheroid fabrication, this technology provides means to decouple the living units’ dimensions from cell density, which is currently unattainable with conventional scaffold-free spheroid production methodologies. These units aim to unlock on-demand processing for hierarchic living materials, vital for tissue engineering, with reproducibility and controllable cell density.

 

REFERENCES: 1Achilli, T., Meyer, J., & Morgan, J. (2012). Advances in the formation, use and understanding of multi-cellular spheroids. Expert Opinion on Biological Therapy, 12, 1347 – 1360. 2Chen, S., Wu, C., & Chen, H. (2018). Enhanced Growth Activities of Stem Cell Spheroids Based on a Durable and Chemically Defined Surface Modification Coating. ACS applied materials & interfaces, 10 38, 31882-31891.

 

ACKNOWLEDGEMENTS: This work was financed by the European Research Council Advanced Grant REBORN (grant agreement number: H2020-ERC-AdG–883370) and within the scope of the project CICECO Aveiro Institute of Materials, UIDB/50011/2020, UIDP/50011/2020 & LA/P/0006/2020, financed by national funds through the FCT/MEC (PIDDAC). Beatriz Moura  acknowledge individual PhD fellowships from the Portuguese Foundation for Science and Technology (FCT) (2021.08331.BD).

Beatriz Moura obtained her Masters in Bioenginnering from University of Porto in 2020. During her Masters, she did an internship in the Paris Seine Institute of Biology and her master thesis in the Institute of Investigation and Innovation in Health (i3S), in Porto. Her master thesis focused on developing a microfluidic chip to study the interplay between the sensory nervous system and the bone metastatic niche. Currently, she is pursuing a PhD in medical biotechnology from the University of Aveiro, with a research focus on the development of living materials for tissue engineering and therapeutics. She specializes in cell surface modification techniques such as metabolic glycoengineering and click-chemistry, aiming to advance knowledge in this field.

Dr. Vincent Hickl (CH)

Post-doctoral researcher, Biointerfaces, Empa

Single-cell live imaging of bacterial infections at 3D surfaces

Hickl123, A. Khan4, R. M. Rossi3, B. F. B. Silva123, K. Maniura-Weber1

1Laboratory for Biointerfaces, Empa, St. Gallen, Switzerland, 2Center for X-ray Analytics, Empa, St. Gallen, Switzerland, 3Laboratory for Biomimetic Textiles and Membranes, 4University of Illinois Urbana-Champaign, Urbana, IL

 

INTRODUCTION: The development of infections, particularly through the emergence of biofilms, is governed by the self-organization of microbes at surfaces. Limitations of live imaging techniques make collective behaviors in clinically relevant systems difficult to quantify. In particular, widely available analysis methods are generally insufficient for single-cell segmentation in 3D from live imaging. This challenge is compounded by the presence of multiple bacterial species in most clinical systems, and by the systems’ geometric complexity. Here, we present novel experimental and image analysis methods to perform high-fidelity imaging and single-cell segmentation of 3D bacterial colonies.

 

METHODS & RESULTS: The best currently available segmentation tools for microscopy images rely on machine-learning algorithms trained using manually labelled images, which are both time-consuming to produce and prone to human biases. We have adapted image-to-image translation methods to create training data from synthetic microscopy images that require no human labelling. The resulting algorithms achieve precise single-cell segmentation for densely-packed bacteria at surfaces, even in systems relevant for research in the life sciences that are not optimized for image quality.

In particular, we image monolayers of Pseudomonas aeruginosa grown on patterned PDMS films in custom microfluidic devices. PDMS films with precisely controlled 3D geometries are produced via excimer laser ablation to provide a model system in which to study the effects of surface curvature on bacterial collective behaviors. Using our novel segmentation approach, we quantify the spatiotemporal organization of the bacteria in 3D. Additionally, we achieve accurate multi-species, single-cell segmentation in mixed colonies of P. aeruginosa and Staphylococcus aureus without differential staining of the two species.

 

 

CONCLUSION & DISCUSSION: Through quantitative imaging in complex environments, these advances promise to provide new insights into the self-organization of pathogenic bacteria, including biofilm formation. As a result, our live imaging approach through microfluidics may be used as a testing platform to shed light on the mechanisms of novel antibacterial therapies. The use of synthetic images for the creation of bespoke segmentation algorithms without human annotation could be immensely useful for quantitative image analysis throughout the life sciences.

 

REFERENCES:

  1. J. Yan, A. G. Sharo, H. A. Stone, N. S. Wingreen, B. L. Bassler, Proc. Natl. Acad. Sci. U. S. A. 113, e5337–e5343 (2016).
  2. M. C. Duvernoy et al., Nat. Commun. 2018 91. 9, 1–10 (2018).
  3. G. C. L. Wong et al., Phys. Biol. 18, 051501 (2021).
  4. V. Hickl, A. Khan, R. M. Rossi, B. F. B. Silva, K. Maniura-Weber, arXiv:2405.12407 (2024).

I am a postdoc at Empa St. Gallen affiliated with the Biointerfaces Laboratory, the Center for X-ray Analytics, and the Biomimetic Membranes and Textiles Laboratory. I received my PhD in physics from the University of Illinois, where I studied the interactions of oil-degrading bacteria with microscopic droplets of crude oil. Now, I am interested in how the collective behaviors of bacteria influence their pathogenicity and their ability to contend with complex environments. In particular, I am developing novel machine-learning and data science-based approaches to quantify the spatiotemporal organization of bacteria in 3D with single-cell resolution.

Peng Xuan (DE)

PhD Candidate, Helmholtz-Zentrum Dresden-Rossendorf

Investigation of dual-targeting tumor spheroids with CAR-T cell therapy in hydrogel microbeads

Peng,1 Ž. Janićijević,1 L. Loureiro,1 L. Hoffmann,1 P. S. Lee,2 I. Cela,1 B. Kruppke,2 A. Kegler,1 A. Feldmann,1 M. Bachmann,1 L. Baraban1

 1Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Dresden, Germany.

2Max Bergmann Center of Biomaterials and Institute of Materials Science, Technische Universität Dresden, Dresden, Germany

 

INTRODUCTION: Three-dimensional (3D) in vitro cancer models are gaining increasing popularity as pre-clinical platforms for evaluating the efficacy of existing anti-cancer drugs and discovering innovative therapeutic approaches.[1,2] However, it is worth noting that only a minority of 3D cancer models are built on biomaterial-based matrices, and many of these systems do not fully replicate the complex tumor microenvironment, which can significantly impact the overall therapeutic performance.[3] In our research, we have successfully established 3D prostate cancer and fibrosarcoma co-culture models and utilize them to investigate the dual-targeting of malignant tumor cells and fibroblast activation protein-α (FAP), a marker of tumor microenvironment. [4,5]

 

METHODS: Poly(ethylene glycol) diacrylate  hydrogel beads (PEGDA GBs) were prepare by using an in-house-developed and cost-effective T-junction based droplet-microfluidic platform coupled with a 365 nm UV lamp for polymerization. PEGDA GBs were characterized by scanning electron microscopy and Fourier-transform infrared spectroscopy. Stiffness of PEGDA GBs was measured using a MicroTester.

The PSCA modified prostate cancer cell line PC3-PSCA, and the FAP modified fibrosarcoma cell line HT1080 hFAP were used to generated spheroids. Anti-PSCA-E5B9 target molecules (TMs) and anti-FAP-E5B9 TMs were used to target the respective antigens. Calcein AM/PI live/dead staining, H&E staining, and fluorescent immunostaining were used to characterize the spheroids.

 

RESULTS: The PEGDA GBs exhibit good biocompatibility and the stiffness ranging from 300 to 600 Pa suitable for culturing the cells. The size of most PC3-PSCA spheroids increased faster comparing to the HT1080 hFAP spheroids and the co-cultured spheroids of PC3-PSCA and HT1080 hFAP. The co-cultured cancer cells can be effectively killed when adding both anti PSCA and anti FAP TMs.

 

Fig. 1: Representative spheroids in PEGDA GBs after 9 days of culturing.

 

Fig. 2: Representative PI (red,dead cell) staining after co-culturing the spheroids of PC3-PSCA and HT1080 hFAP with CAR T cells (green) for 48 h under different cell culture conditions.

 

DISCUSSION & CONCLUSIONS: The PC3-PSCA and HT1080-hFAP co-culture model is a viable proof-of-concept for assessing the targeting of cancer-associated antigens, including those involved in modulating the tumor microenvironment, such as FAP. Our goal is to develop various hydrogel beads or capsules as tunable bioreactors for the enhancement of cancer modeling accuracy. These advancements will serve as valuable tools for future cancer research endeavors.

 

REFERENCES: 1 J. Drost and H. Clevers (2018) Nat. Rev. Cancer, 18:407-18. 2 M. Hofer and M. P. Lutolf (1995) Nat. Rev. Mater. 6: 402–20. 3 R. Curvello, V. Kast, P. Ordóñez-Morán, et al (2023) Nat. Rev. Mater. 8: 314-30. 4A. Feldmann, C. Arndt, R. Bergmann, et al (2017). Oncotarget. 8: 31368–85. 5L.R. Loureiro, L. Hoffmann, et al. (2023) J. Exp. Clin. Cancer Res. 42: 341.

 

ACKNOWLEDGEMENTS: We would like to thank the support of China Scholarship Council; Deutsche Forschungsgemeinschaft (DFG, Nr.BA4986/8−1), GRK 2767; Helmholtz Initiative and Networking Fund in the project ‘Mhelthera’ (project ID: InterLabs-0031). Furthermore, LB acknowledges the financial support of European Research Council (ERC) in the ERC- Consolidator Grant (ImmunoChip, 101045415).

Currently, I am a Ph.D. candidate at Helmholtz-Zentrum Dresden-Rossendorf (HZDR, Germany), where I specialize in utilizing microfluidic methods to prepare diverse hydrogel microcapsule/bead reactors for 3D tumor cell generation. I have applied these methods to liver, prostate, and epithelial cancer cells, with some of my results published in Biotech. J. (2023) and Adv. Health. Mater. (2024). My ongoing project delves into exploring their utility in UniCAR-T cell therapy as preclinical models. My work experience has fostered my interest in 3D tissue engineering, biofabrication, nanotechnology, cancer therapy, and other related areas. I also collaborate with colleagues on biosensor and nanoparticle therapy projects, I contribute expertise in cell experiments and material synthesis.

Dr. Emanuele Mauri (IT)

Assistant Professor, Applied Physical Chemistry, Politecnico di Milano

Advanced formulation of hyaluronan-based nanogels for targeted therapy in ovarian cancer

Mauri1, C. Colli1, A. Rainer2, L. Rosanò3, D. Moscatelli1

1Dept. Chemistry, Materials and Chemical Engineering “G. Natta”, Politecnico di Milano, Milan, Italy. 2Dept. Engineering, Campus BioMedico di Roma, Rome, Italy. 3Institute of Molecular Biology and Pathology, National Research Council, Rome, Italy

 

INTRODUCTION: Hyaluronan (HA)-based nanogels (NGs) are promising drug delivery system for tumoral treatments. However, the conventional synthetic routes exhibit some limitations in terms of process scalability, ad-hoc NG properties, and batch-to-batch reproducibility [1].

We propose two innovative strategies to overcome these constraints: a mixed emulsion/evaporation technique (MEET) and a droplet-based microfluidics. We have validated our NGs as drug delivery system in ovarian cancer, showing their potential use as CD44-targeting agent due to the selective binding to these receptors.

 

METHODS: NGs are obtained via chemical crosslinking between HA and polyethyleneimine (PEI).

MEET: each polymer is separately dissolved in water, and then dispersed into the same continuous organic phase. The interfacial interaction and the progressive coalescence between the two dispersed phases give rise to the NGs [2].

Droplet-based microfluidics: HA-PEI aqueous solution is prepared as disperse phase and mineral oil used as continuous phase. A hydrodynamic flow-focusing microfluidic device is fabricated and used to form a microemulsion consisting of aqueous dispersed microdroplets working as reactors in which NGs are generated [3].

DOX-loaded NGs are administered to ovarian cancer cells and the therapeutic effect analysed via MTT assay. In vitro assay with a CD44 blocking/neutralizing antibody is performed to validate NGs as CD44-targeting system.

 

RESULTS: Both techniques ensure high reproducibility, low polydispersity and colloidal stability of the NGs. In microfluidic set-up, tuning the flow focusing geometry enables the modulation of the microdroplet diameter and, as a result, NGs size can be finely controlled (range 80-300 nm). The HA NGs exhibit a sustained drug release and are internalized only by cells expressing CD44 receptors. NG-mediated DOX release improves the therapeutic effects vs the administration of non-encapsulated drug.

 

Fig. 1: NG synthesis

 

Fig. 2: Drug release (A) and DOX-induced cytotoxicity in OVCA433 cells at day 7 (B)

 

DISCUSSION & CONCLUSIONS: The methods ensure the formation of NGs with specific targeting features, in a highly controlled manner. In particular, microfluidic platform ensures the in-flow production of NGs with an extremely high reproducibility, encouraging the progress towards an effective use of the technique in tumour treatments.

 

REFERENCES: 1 E. Mauri (2021) Gels, 7:36. 2 E. Limiti (2022) ACS Appl Nano Mater, 5:5544−5557. 3 S.M Giannitelli (2022) Nanoscale, 14:11415.

Emanuele Mauri is Assistant Professor in the framework of Applied Physical Chemistry at Politecnico di Milano. He graduated in Chemical Engineering (2014) and he received a Ph.D. in Industrial Chemistry and Chemical Engineering (2018) at Politecnico di Milano with a thesis on the synthesis of hydrogels for drug and cell delivery in central nervous system. His main research fields are related to the chemical functionalization of polymers and the design of three-dimensional polymeric systems for controlled drug, gene and cell delivery. Special focuses are: functionalization of nanoparticles for drug targeting in inflamed/injured cells of tumoral scenarios, continuous in-flow production of nanoparticles through microfluidic devices to overcome the shortcomings of the conventional batch syntheses, and 3D printing of bioinks for the design of organotypic models.

Jannes Felsch (CH)

PhD Candidate, Pharmaceutical Sciences, University of Basel

Protection of biomaterials against immune-mediated complications: Using a bacterial surface model to evaluate the efficacy of complement regulator-recruiting peptide coatings

Felsch1, G. Sommerfeld1, A. Blagojevic1, E. Umnyakova1, D. Ricklin1

1 Molecular Pharmacy, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland.

 

INTRODUCTION: The complement system confers a central part of host defense. It is organized in three pathways, which converge in a terminal effector pathway that releases anaphylatoxins and forms lytic membrane attack complexes. Although complement activation is critical for protection against pathogens and maintenance of cell homeostasis, biomaterial-induced complement activation can lead to severe adverse effects such as functional loss of grafts or inflammatory reactions.

 

The natural complement regulator factor H (FH) recognizes host cells and impairs the amplification of complement responses, which renders it a potential candidate to prevent complement activation on biomaterial surfaces. However, direct surface modification with FH is technically challenging and expensive. The cyclic peptide 5C6 can bind blood-derived FH in its active orientation. Therefore, surface coating with 5C6 is an elegant method to capture physiological FH and protect surfaces from immune-mediated complications.

 

While biomaterials are considered the prime application of the technology, polymer-coating models are of limited use for screening of next-generation 5C6 analogs. Owing to their susceptibility to complement-mediated damage, bacterial surfaces may serve as surrogate system for biomaterial-induced immune reactions. Herein, we describe the click-chemistry based modification of bacterial surfaces with 5C6 using metabolic labeling and its impact on the complement-mediated lysis in normal human serum (NHS).

 

METHODS: Bacteria (E. coli K12) were incubated in culture medium containing azide-modified carbohydrates (Kdo-N3) to introduce a click handle on bacterial lipopolysaccharides. Upon confirming the presence of surface azides using a clickable fluorophore, an alkyne-modified 5C6 analogue or a sequence-scrambled control (prepared by solid-phase peptide synthesis) were clicked to the surface. The modified bacteria were exposed to NHS and lysis was monitored (reduction of colony-forming units).

 

Fig. 1: Experimental scheme of 5C6-coating of bacterial surfaces as a model system for protection against immune-mediated reactions.

 

RESULTS: Metabolic labeling successfully introduced azide modifications on the surface of E. coli. The azide and peptide modification did not

impair bacterial growth under culture conditions. When exposed to NHS, 5C6-modified bacteria showed substantially enhanced survival compared to unmodified bacteria or to bacteria that were modified with a scrambled peptide analogue (scr).

 

DISCUSSION & CONCLUSIONS: Our study introduces a bacterial survival model as novel test system for assessing complement regulator-recruiting surface coatings. The efficiency of click-chemistry-based coating and use of complement-mediated damage as clear and relevant readout provide important advantages. While the current model may be employed for the testing of coatings and therapeutics, the wide scope of click reactions suggest that the coating strategy can be adapted to biomaterial and/or graft cell surfaces.

 

REFERENCES: 1 Bechtler et al. (2023) Complement-regulatory biomaterial coatings: Activity and selectivity profile of the factor H-binding peptide 5C6, Acta Biomater. 155: 123-138. 2 Ziylan et al. (2023) Evaluation of Kdo-8-N3 incorporation into lipopolysaccharides of various Escherichia coli strains, RSC Chem. Biol. 4: 884-893.

 

ACKNOWLEDGEMENTS: This work is supported by intramural funds of the University of Basel and the Swiss National Science Foundation. (grants 31003A_176104, 310030_219969).

Jannes Felsch graduated with a Master of Science “Pharmaceutical Sciences” with focus on Drug Discovery and Delivery from the Albert-Ludwigs-University of Freiburg (Germany). During his studies, he has been working on various research projects. This includes a liposome project in the Pharmaceutical Technology in Freiburg as well as protein-templated reactions at the FU Berlin, before joining the Molecular Pharmacy group of Prof. Dr. Daniel Ricklin for his master thesis in 2022. In 2023, Jannes Felsch joined the Ricklin lab as a PhD Student. His research focuses on the protection of biomaterials against immune-mediated complications using the FH-capturing cyclic peptide 5C6. Currently, he is working with biomaterials ranging from living systems (eukaryotic cells, bacteria) to artificial surfaces like liposomes.

Tamara Zünd (CH)

PhD Candidate, Lab of Applied Mechanobiology, Department of Health Sciences and Technology, ETH Zürich

Mechanobiological T cell stimulation using nanoporous confinement of microvilli induces T cell signaling leading to CD69 and Il-2 expression

Zünd1, S. Lickert1, T. Kovalchuk1,2, L. Baldi1, E. Klotzsch2, V. Vogel1

1Laboratory of Applied Mechanobiology, Department of Health Sciences and Technology, ETH Zurich, Zürich, Switzerland, 2Institute for Biology, Experimental Biophysics/Mechanobiology, Humboldt-Universität zu Berlin, Berlin, Germany

 


INTRODUCTION: Chimeric antigen receptor (CAR) T cell therapy has revolutionized blood cancer treatment, but the production of this living therapeutic is complicated, leading to sub-optimal drug efficacy. Central to this process is ex vivo T cell activation, which controls gene transfer, proliferation, and differentiation. Recent advances have highlighted the important effects of mechanical stimulation on T cell activation and differentiation1. However, most strategies used in research and clinics rely solely on biochemical triggering of T cells. Here, we present a breakthrough approach that activates T cell signaling via nanoporous substrates, dictating T cell function and phenotype (Fig. 1).


METHODS: Primary human naïve pan T cells were isolated from healthy donors and subjected to experiments measuring ERK and NFкB phosphorylation via flow cytometry. After 24 hours, IL-2 secretion was quantified using Lumit Human IL-2 Immunoassay. CD69 expression was assessed via flow cytometry. For T cell expansion, IL-7 and IL-15 were added to the T cell medium. The T cell phenotype was analysed after 10 days of expansion, targeting CCR7, CD4, CD8, and CD3. Immunofluorescent staining against CD3, CD45, CD28, and LCK, as well as wheat germ agglutinin (WGA), were measured using Airy scan imaging.


RESULTS: We demonstrate accumulations of signaling proteins such as LCK, CD3, and CD28 on microvilli within nanopores (Fig. 2). These signaling hotspots induce high levels of initial ERK and increased levels of NFкB phosphorylation, even surpassing CD3 antibody stimulation (Fig. 3). Surprisingly, stimulation with nanopores is sufficient to fully activate T cells, indicated by CD69 expression and ~5.2 times higher IL-2 secretion compared to biochemical CD3 stimulation, followed by mild expansion with a fold change of ~4. For practical implementation of these findings, we introduced additional CD28 co-stimulation via antibodies to ensure high levels of Il-2 secretion followed by increased expansion rates, resulting in high percentages of CCR7-expressing cells.

 

DISCUSSION & CONCLUSIONS: This unique synergy of mechanical and biochemical co-stimulation facilitates robust proliferation while retaining a high population of memory like phenotype. In summary, our innovative method, utilizing gentle mechanical stimulation instead of harsh biochemical approaches, shows potential for enhancing CAR T cell therapy by promoting the generation of more memory-related T cells.

 

 

Fig. 1: Cartoon depicting a T cell spreading on a nanoporous substrate. T cell form microvilli extending into the nanopores inducing TCR-ligand independent T cell signaling.

 

Fig. 2: Airy scan image showcasing a primary human naïve T cell cultured on a nanoporous substrate. WGA was used as a membrane stain and CD3 as an immunofluorescent marker. The image represents a maximum z-stack projection, highlighting microvilli with TCR complexes inside the nanopores.

 

Fig. 3: Percentage of double-phosphorylated primary human naïve T cells for ERK and NFκB, measured by flow cytometry at various incubation times on substrates with different coatings.

 

REFERENCES: 1 M. Aramesh et.al. (2019) Engineering T-cell activation for immunotherapy by mechanical forces, Current Opinion in Biomedical Engineering.

I‘m Tamara Zünd, currently pursuing a PhD in the Lab of Applied Mechanobiology at ETH Zürich. Before beginning my doctoral studies, I completed my undergraduate education in interdisciplinary sciences at ETH Zürich, where I acquired a solid foundation in biology and biochemistry.

Prof. Dr. Anne Géraldine Guex (CH)

Assistant Professor, Oral Implantology, University Center of Dental Medicine Basel

Mechanical compression and shear induce distinct macrophage polarisation and subsequent mesenchymal stromal cell differentiation

G. Guex1,2, U. Menzel2, Y. Ladner2,3, A. R. Armiento2, M. J. Stoddart2,4
1Department Research, University Center for Dental Medicine Basel, UZB, University of Basel, Switzerland

2AO Research Institute Davos, Davos, Switzerland

3Institute for Biomechanics, ETH Zurich, Zurich, Switzerland
4Department of Orthopedics and Trauma Surgery, Medical Center-Albert-Ludwigs-University of Freiburg, Faculty of Medicine, Freiburg, Germany

 

INTRODUCTION: In-situ, cells are influenced by physical forces, which are often neglected in standard experiments. By use of an in-house built bioreactor to culture cell-scaffold constructs under compression and shear, we overcome limitations of static in vitro culture1. The here presented system allows us to mimic the biomechanical environment during bone fracture healing to study macrophage polarisation under load. We aim to gain in-depth understanding of the correlation between mechanical load and the immune response, and subsequent mesenchymal stromal cell (hMSC) differentiation during callus formation. 


METHODS:
THP-1 monocytes were seeded in fibrin hydrogels. Transition to macrophages was induced with PMA. Macrophages were then cultured in standard medium (M0) or medium with LPS and IFN-γ, or IL-4 to induce an M1 or M2-like phenotype, respectively (named M(LPS) or M(IL-4)). Culture was either under static conditions, or in a multi-well bioreactor at 20% compression and shear at a frequency of 1 Hz for 1 hour per day during 3 days. Conditioned media was collected, mixed 1:2 with fresh culture media, and added to human bone marrow derived MSCs in pellets (3 donors). Expression of ACAN, ADAMTS4, ADAMTS5, ALPL, COL1A1, COL2, COL10, IL-6, IL-8, MMP13, RUNX2, and SOX9 was assessed by RT-qPCR after 9 days in pellet culture.

 

RESULTS: In THP-1 cells, mechanical stimulation induced changes in the gene expression level of both pro- or anti-inflammatory hallmarks. In M(IL-4) conditions, the expression of IL-1β, IL-6, TNF-α; IL-10, CCL18, CD163, and CD206 was increased compared to the static condition. The only exception was NOS2, with a reduced expression in all conditions after loading compared to the static control. In M(LPS) conditions, a less pronounced up or down-regulation in response to mechanical load was observed. The majority of assessed genes in hMSCs remained unchanged, with few exceptions: IL-8 decreased, ADAMTS4, RUNX2, ALPL, and COL2 increased in all conditions treated with medium from mechanically stimulated THP-1 cells.

Fig. 1: A) Schematic of cell-hydrogel loading. A ceramic lever is in direct contact with the hydrogel and applies compression and shear. B) Confocal microscopy image of THP-1 cells in fibrin. Actin skeleton in green, nuclei in blue.

Fig. 2: Expression of selected genes reported as
2-ΔΔCt relative to house-keeping genes and THP-1 harvested on day 0. * p<0.5 compared to all other conditions or how indicated.

 

DISCUSSION & CONCLUSIONS: Our results suggest that mechanical loading induces a complex macrophage phenotype. Further adding to the complexity, MSCs cultured with conditioned media responded in a donor-dependent manner, underpinning clinical observations of patient-specific symptoms to local or systemic inflammation. Ultimately, we aim to suggest an appropriate loading or resting scheme for patients with bone fractures.   

 

REFERENCES: 1 Y. Ladner, H. Kasper, A. R. Armiento, M. J. Stoddart (2023), A multi-well bioreactor for cartilage tissue engineering experiments, iScience 26:107092.

Géraldine Guex has been appointment assistant professor in Oral Implantology at the University Center of Dental Medicine Basel, UZB, in 2023. She obtained her PhD in cardiac tissue engineering from the University of Berne in 2012 and worked as a Postdoc on bone tissue engineering at Imperial College, London for over two years. Upon her return to Switzerland, she was leading projects as Scientist at the Empa in St. Gallen, and the AO Research Institute in Davos where she primarily focused on material-based approaches for skin wound healing, or cartilage and bone tissue engineering, respectively. As a Nanoscientist by training, Géraldine is particularly interested in interdisciplinary projects to understand cell material interactions and control cell fate decision by functional materials.

Prof. Dr. Laura Suter-Dick (CH)

Group leader, Cell biology and in vitro toxicology, Institute for Chemistry and Bioanalytics, FHNW School of Life Sciences

A “plug & play” microphysiological system to quantify the liver fibrosis AOP in vitro

Suter-Dick1, L. Burr2, A. Homsy3

1 University of Applied Sciences and Arts Northwestern Switzerland (FHNW), School of Life Sciences, Muttenz, CH.

2 Centre Suisse d’Electronique et de Microtechnique SA (CSEM), Landquart, CH.

3 Haute Ecole Arc, School of Engineering (HES-SO), La Chaux-de-Fonds, CH.

 

INTRODUCTION: Adverse Outcome Pathways (AOPs) represent a series of biological key events that lead to an adverse outcome [1]. Quantitative AOP models can provide a bridge between the description of events and the prediction of hazard and risk assessment. The liver fibrosis AOP depicts the liver response to chronic injury, leading to the pathological accumulation of extracellular matrix [1]. Liver fibrosis remains a major health concern for which effective therapies are lacking. The processes leading to hepatic fibrosis can be mimicked in vitro [2], [3], but the quantification of the cellular responses leading to either pathological fibrosis or adaptive liver responses represents a major challenge.

 

METHODS: In this project, we developed a novel microphysiological system tailored for the quantitative investigation of the liver fibrosis AOP (AOP-MPS) by housing human cell lines representing hepatocytes, Kupffer cells and hepatic stellate cells cultured in 2D or 3D (Table 1). The AOP-MPS was fabricated by 3D-printing to generate a prototype amenable to injection molding in the future. The “plug & play” design allows switching on-demand between separate mono-cultures cultures interconnected by sequential perfusion. To quantify cellular responses, electrochemical sensors are incorporated for in-line monitoring of glucose, lactate, pH and levels of reactive oxygen species. In addition, the cell cultures can be investigated using immunostaining for cellular markers, such as albumin (hepatocellular function) or αSMA (fibrotic stress fibres).

 

Table 1. Human cell lines in the AOP-MPS.

Cell line

Representing

Cultured as

HepaRG

Hepatocytes

Spheroids (3D)

THP-1

Kupffer Cells

Monolayer (2D)

hTERT-HSC

Stellate Cells

Spheroids (3D)

  

RESULTS: The implemented AOP-MPS was biocompatible and the built-in sensors were placed downstream from the cells to avoid any potential cytotoxicity (Fig. 1). Treatment of the cells (7 days) in the AOP-MPS with pro-fibrotic stimuli resulted in a fibrotic phenotype detected by immunostaining. The individual cellular responses could be quantitatively monitored with the built-in sensors.

 

Fig. 1: Built-in sensors, downstream of the perfused, interconnected cell compartments in the AOP-MPS (prototype).

 

DISCUSSION & CONCLUSIONS: This innovative “plug & play” AOP-MPS facilitates the investigation of cellular interactions leading to liver fibrosis. The quantitative data can support in vitro to in vivo extrapolation, thus positioning it as a valuable tool for advancing liver fibrosis research, including toxicological investigations and drug discovery.

 

REFERENCES: [1] T. Horvat et al. Arch. Toxicol., vol. 91, no. 4, pp. 1523–1543, Apr. 2017. [2] V. Prestigiacomo et al., PloS One, vol. 12, 2017.  [3] I. Mannaerts et al., Biomaterials, vol. 261, p. 120335, Dec. 2020.

 

ACKNOWLEDGEMENTS: Funding by the Swiss National Science Foundation, BRIDGE Discovery Project 40B2-0_187219.

Prof. Laura Suter-Dick is a full professor at the School of Life Sciences, University of Applied Sciences Northwestern Switzerland (FHNW), where she leads the group for Cell Biology and in vitro Toxicology since 2012. She holds a Ph.D. in biology, is a European Registered Toxicologist (ERT) and acquired ample experience in the pharmaceutical industry. She specialized in molecular and mechanistic toxicology, with a strong focus on advanced in vitro systems for toxicity assessment, alternatives to animal methods, and toxicogenomics. Her current research focuses on the application of microfluidics and microphysiological systems to toxicity assessment and disease modeling. She is actively involved in several scientific societies (SST, ESTIV, Eurotox, 3RCC, SCAHT) and currently presides Biotechnet Switzerland.

Dr. Behnam Akhavan (AU)

Senior Lecturer and Leader, Plasma Bio-engineering Research Group, University of Newcastle

Ion-assisted plasma surface bio-functionalisation: Bio-instructive interfaces for implantable medical devices

Akhavan1,2,3, M. Bilek3, Hala Zreiqat3, Anyu Zhang3, Zufu Lu3, L. Martin3

1 School of Engineering, University of Newcastle, Callaghan, NSW 2308, Australia

2 Hunter Medical Research Institute, Precision Medicine Program, NSW 2305, Australia

3 School of Biomedical Engineering, University of Sydney, Sydney, NSW 2006, Australia

 

INTRODUCTION: Implantable medical devices face the challenge of eliciting an optimal biological response for rapid tissue integration. Here, we report the creation of a new class of plasma-engineered bio-instructive interfaces, facilitating the rapid tissue integration of implants. Our approach combines radical-rich coatings with controlled peptide immobilisation and covalent attachment of hydrogel layers. The strategy involves two steps. First, highly robust radical-rich ion-assisted plasma polymerised (IPP) coatings are created on the implants. Second, electric fields, controlled by pH modulation or applied by simple household batteries, ensure specific peptide orientation and density without chemical pre-treatment. The radical-rich coatings also enable hydrogel polymerisation and covalent attachment upon contact, eliminating the need for initiators and mitigating concerns related to toxic initiators in the production of soft biointerfaces.

 

METHODS: The IPP coatings were deposited on a variety of substrates, including titanium, stainless steel, and additively manufactured Ti and Mg-doped bagdédite bioceramic scaffolds as well as a wide range of polymers such as ePTFE, polystyrene and PCL, using a precursor gas mixture of argon, acetylene, and nitrogen. A specifically designed peptide (AcFFMMMAAAAAAAAAADDDDDK-NH2) was used. The IPP-coated surfaces were incubated in the peptide solution using a custom-made well plate with electrodes above and below the samples. Voltages (0-30 V) in either the Eup or Edown directions were applied during immobilization to control the orientation and density of surface-attached peptides. For hydrogel functionalisation, the activated surfaces were incubated in various hydrogel monomer solutions such as GelMa, chitosan, and alginate with no addition of initiators or cross-linkers. The efficacy of the interfaces in regulating the cellular responses was evaluated both in vitro and in vivo.

 

RESULTS: Hydrogel formation, peptide orientation and density on IPP-coated surfaces were assessed using spectroscopic and biological assays, including ToF-SIMS, XPS, ATR-FTIR, ellipsometry, and ELISA. XPS and ToF-SIMS results showed higher concentrations of sulphur-containing groups with increased Eup field strength, indicating success in orienting the peptide (Figure 1). Both pre and post-detergent-washing data from XPS, ToF-SIMS, and ellipsometry demonstrated the radical-rich coatings’ excellent capacity for covalent hydrogel attachment and polymerization without additional reagents. The biofunctionalised interfaces created using this dry and environmentally friendly approach showed a reduced protein desorption and a more sustained osteoblast response both in vitro and in vivo compared to implants modified through conventional physisorption of biomolecules such as BMP-2 (Figure 2).

 


Figure 1. External field controls peptide density and orientation as indicated by XPS and ToF-SIMS data.

 

Figure 2. Covalent protein immobilization through the IPP interlayer.

 

DISCUSSION & CONCLUSIONS: These results provide strong evidence that electric fields on radical-functionalized IPP films effectively immobilize peptides with precise orientation and density. The same coatings further enable reagent-free hydrogel polymerisation and covalent attachment, presenting significant potential as bio-instructive interfaces for surface engineering of rapidly integrating implantable biomedical devices.

Dr. Behnam Akhavan, an Australian Research Council (ARC) DECRA Fellow and a Senior Lecturer of Biomedical Engineering at the University of Newcastle, heads the Plasma Bio-engineering Research Group at the School of Engineering and the Hunter Medical Research Institute (HMRI). Since obtaining his PhD in Advanced Manufacturing from the University of South Australia in 2015, he has held postdoctoral positions at the Max Planck Institute for Polymer Research and Fraunhofer Institute of Microtechnology in Germany, and the University of Sydney. Dr. Akhavan‘s pioneering work in plasma surface bio-engineering, published in over 70 journal articles, has led to innovative applications in healthcare and beyond. He is celebrated by Engineers Australia as one of the nation’s Most Innovative Engineers.

Dr. Jiangtao Zhou (CH)

Staff of Professorship, Food and Soft Materials Science, Department of Health Sciences and Technology, ETH Zürich

An optical fiber-based nanomotion sensor for rapid antibiotic and antifungal susceptibility tests

Jiangtao Zhou1,2, Changrui Liao3, Giovanni Dietler1, Sandor Kasas4,5

1EPF Lausanne, Lausanne, Switzerland. 2 ETH Zurich, Zurich, Switzerland.

3 Shenzhen University, Shenzhen, China.4 University of Lausanne (UNIL), Lausanne, Switzerland

5 VUB-EPFL BioNanotechnology & NanoMedicine, Brussels, Belgium

 

INTRODUCTION: The emergence of antibiotic and antifungal resistant microorganisms represents nowadays a major public health issue that might push humanity into a post-antibiotic/antifungal era. One of the approaches to avoid such a catastrophe is to advance rapid antibiotic and antifungal susceptibility tests [1]. However, traditional sensitivity tests based on the bacterial and fungal proliferation rate take tens of hours for identifying drug-resistant microorganisms. The recent rapid antimicrobial susceptibility devices based on AFM nanomotion sensing of microorganism activities can complete susceptibility test in a time frame of minutes[2], but these sensors reply on the complex and sophisticated AFM instrument that is difficult to parallel in the large-scale test for the diagnosis of bacteria or fungi.

 

METHODS: The process of 2PP printing the optical fiber-based nanomotion sensor by femtosecond laser are three steps (Fig. 1a-b): 1) A drop of photoresist was dropped on the end face of the standard SMF and a coverslip was placed onto the SMF upper surface to stop the photoresist from flowing away. 2) the glass slide described above was then mounted on the femtosecond laser stage for structure printing. The femtosecond laser was focused in the photoresist on the fiber end face through an oil-immersion objective (Zeiss, 63x, NA=1.4). The base and cantilever structures were printed on the SMF end face according to the pre-designed scan path. 3) a drop of the developer made by mixing isopropanol and acetone (the volume ratio is 4:1) was applied to the fiber end face to remove the unexposed photoresist.

 

RESULTS: We present an optical fiber-based nanomotion sensor that shows a high sensitivity of nanomotion detection in antimicrobial susceptibility tests and indicates the potential of parallelization because of its compact feature and simplicity of use[3]. A flexible 3D-printed cantilever as a nanomotion sensing beam was anchored at the distal end of the optical fiber by two-photon polymerization (2PP) technology (Fig. 1c). In our sensor, the vibration of the cantilever onto which the microorganisms are attached is real-time monitored by an in-fiber interferometer. The geometry and nanomechanical properties of our cantilever sensors, i.e., a thickness of about 1 μm and a spring constant of 0.3 N/m, are comparable to that of the AFM nanomotion cantilever. As a proof-of-concept prototype, this novel device shows excellent performance in real-time susceptibility tests of Escherichia coli and Candida albicans to antibiotics and antifungals, respectively. This strategy of a nanomotion sensor with both advantages in fast-response and parallelization may advance this technology toward a next-generation nanomotion sensor for large-scale and rapid antimicrobial susceptibility tests, as well as other technological and biomedical applications.

 

Fig. 1 (a) Cantilever fabrication process. (b) Schematic of cantilever fabrication. (c) SEM images of sensing head and fluorescence image living E. coli. on the cantilever. Scale bars are 20 µm.

 

DISCUSSION & CONCLUSIONS: We presented a high-sensitivity optical fiber-based nanomotion sensor for fast antimicrobial susceptibility tests, with the simplicity of use and the possibility of parallelization. We anticipate our device could be an interesting candidate for next generation nano-motion sensor for antimicrobial susceptibility tests, microorganism viability assays, as well as other biological and biomedical applications.

 

REFERENCES:

[1] Burnham et al. Nature Reviews Microbiology (2017), 15 (11), 697.

[2] Longo et al. Nat. Nanotechnol. 2013, 8, 522.

[3] Zhou et al. Nano Lett. 2024, 24, 10, 2980.

Dr. Jiangtao Zhou is currently working at ETH Zurich as a scientist focusing on the study of atomic force microscopy and amyloid fibrils based biomaterials. Prior to that, Dr. Zhou earned his PhD degree in EPFL with Giovanni Dietler studying the atomic force microscopy and nanomotion sensors.

Alessandro Stumpo (CH)

PhD Candidate, Department of Biomedicine, University of Basel and Institute for Chemistry and Bioanalytics, FHNW School of Life Sciences

Raman microscopy: enhancing EGFR targeting in ovarian cancer cells through surface modifications of gold nanoparticles

Alessandro Stumpo1,2,3, Francis Jacob2, Viola Heinzelmann-Schwarz2, Scott McNeil3, Sina Saxer1

 1University of Applied Sciences and Arts of Northwestern Switzerland (FHNW), Muttenz, Switzerland

2Ovarian Cancer Research, Department of Biomedicine, University Hospital Basel and University of Basel, Basel, Switzerland

3Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland

 

INTRODUCTION: Debulking surgery followed by chemotherapy remains the most efficient ovarian cancer (OC) treatment strategy, with fluorescence guided surgery (FGS) as gold standard for an intraoperative disease visualization. Recently, properly functionalised gold nanoparticles (AuNPs) were proposed as an alternative to fluorescence, exploiting their optical properties by Raman microscopy. The ideal AuNPs surface chemistry necessary to achieve a perfect balance between Raman sensitivity, binding specificity, and AuNPs stability/biocompatibility was previously defined by Burgio et al1. However, due to the limitation of the carbodiimide crosslinker chemistry, the antibodies on the surface are randomly oriented, possibly affecting their binding sites and consequently their capability of recognizing the antigen on the cell surface. We hereby have outlined 60 nm AuNPs with SERS properties using a different antibody binding approach, Light Activated Site-Specific Conjugation (LASIC)2, and tested their capability of better discriminating EGFR-positive and EGFR-negative OC. This method allows for precise control over the orientation of anti-EGFR antibodies, ensuring optimal binding without compromising antibody functionality.

 

Fig.1: Schematic illustration of AuNPs with different antibody binding approach targeting ovarian cancer cells expressing the EGFR protein on the surface (in red). 1,4-Benzenedithiol (1,4 BDT) is used as Raman reporter.

 

METHODS: Raman measurements were carried out with confocal Raman alpha300 R microscope (WiTec, GmbH, Germany) equipped with a 785 nm excitation wavelength. Each cell is analysed individually, and the presence of AuNPs assigned based on the detection of at least two of the main four 1,4 BDT peaks: 350, 730, 1050 and 1550 rel.1/cm.

 

RESULTS: Physicochemical properties (PCs) of AuNPs with both surface chemistries were initially measured, and any change evaluated in terms of their influence on AuNPs stability, biocompatibility, and specificity. Finally, the difference in the antibody’s functionality on the AuNPs surface is evaluated in terms of their capability of distinguishing EGFR-positive (OVCAR8, OVCAR5) and EGFR-negative (TOV-112D, Knock-out OVCAR8) ovarian cancer cells.

 

Fig.2: Raman analysis of OVCAR5 cells treated with AuNPs displaying anti-EGFR antibodies on the surface (LASIC). a, fluorescence microscopy image; Nuclei stained with DAPI. b, Raman image; Background depicted in orange; dbcoAuNPs depicted in red.

 

DISCUSSION & CONCLUSIONS: Our work introduces a surface chemistry capable of increasing AuNPs specificity towards cells expressing a specific antigen and hence of improving Raman signal sensitivity to visualize cancer cells intraoperatively.

 

REFERENCES: 1ACS Appl. Nano Mater. 2020, 3, 3, 2447–2454. 2Bioconjugate Chem. 2015, 26, 8, 1456–1460.

 

ACKNOWLEDGEMENTS: This project is kindly funded by the Swiss Nanoscience Institute (SNI), University of Basel.

I am a Phd candidate affiliated with the University of Basel. My research project is conducted within the NanoLab at the Fachhochschule Nordwestschweiz and in collaboration with the Ovarian Cancer cell research group at the Department of Biomedicine, University of Basel.

Flash posters

Dr. Edith Perret (CH)

Scientist at the Laboratory of Advanced Fibers, Empa St. Gallen

4113: Local drug delivery with melt-spun liquid core fibers

Röthlisberger1, S. Dul1, P. Meier2 G. Giovannini3, R. Hufenus1, E. Perret1,4

1 Laboratory for Advanced Fibers, Empa, St. Gallen, Switzerland

2 Laboratory for Particles-Biology Interactions, Empa, St. Gallen, Switzerland

3 Laboratory for Biomimetic Membranes and Textiles, Empa, St. Gallen, Switzerland

4Center for X-ray Analytics, Empa, Dübendorf, Switzerland

 

INTRODUCTION: Traditional local drug delivery methods often face challenges such as sudden release of medication, limited drug storage and poor mechanical properties. Here, we introduce a novel approach to local drug delivery employing melt-spun drug-loaded liquid-core filaments (LiCoFs). LiCoFs, composed of a drug-containing liquid core enveloped by poly(ε-caprolactone) (PCL), offer a promising solution to these issues. We conducted experiments incorporating fluorescein sodium salt as a model drug dissolved in various liquid cores. For some fibers, the liquid was also exchanged with different drug-containing solutions using a pumping device. We evaluated the thermal, mechanical, and structural characteristics of LiCoFs and conducted extensive drug diffusion trials.

 

METHODS: The fabrication of drug-loaded melt-spun liquid-core filaments (LiCoFs) was carried out using a customized melt-spinning pilot plant.

 

RESULTS: We demonstrate the feasibility of fabricating drug-loaded LiCoFs through melt-spinning. Fluorescein sodium salt was selected as the model drug and was dissolved in various liquids. Different carrier liquids were utilized to assess their spinnability and to investigate how they impact the drug diffusion behavior. Various analytical techniques, such as tensile tests, differential scanning calorimetry (DSC), scanning electron microscopy (SEM), and x-ray analytics, were employed to comprehensively analyze material and fiber properties. Drug release from the filaments was evaluated through diffusion trials using fluorescence spectroscopy. Additionally, we investigated the exchange of liquid cores with other solutions containing drugs (e.g., ibuprofen), labeled proteins (e.g., bovine serum albumin–fluorescein isothiocyanate, BSA-FITC), and chromophores (e.g., methylene blue-containing liquid, Aqua stabil).  The diffusion of the drugs was analyzed using either Ultra Performance Liquid Chromatography (UPLC), fluorescence spectroscopy or ultraviolet-visible (UV-vis) spectroscopy. [1]

 

Fig. 1: Potential medical applications of drug-loaded liquid-core melt-spun fibers

 

DISCUSSION & CONCLUSIONS: Diffusion results indicated that the diffusion mechanisms and rates are influenced by several factors, including temperature, core size/sheath thickness, sheath permeability, type of carrier liquid, properties of the drug molecule, and its affinity to the sheath polymer. These drug-loaded LiCoFs pave the way towards a new generation of medical textiles enabling precise and localized drug delivery.

 

REFERENCES: 1 M. Röthlisberger et al. (2024) Polymer 298, 126885.

https://doi.org/10.1016/j.polymer.2024.126885

 

ACKNOWLEDGEMENTS: We thank Benno Wüst, Markus Hilber, Bianca Panchetti, Patrick Rupper for helping with experiments and people from the center of X-ray analytics of Empa (Antonia Neels, Bruno Silva, Jonathan Avaro, Leonard Krupnik) for valuable discussions.

Since 2018, Dr. Perret has worked as Scientist at the Laboratory of Advanced Fibers, Empa St. Gallen, Switzerland. Her research centres on the development of drug-loaded liquid core fibers for medical applications. She has led various projects about high-performance industrial monofilaments, including liquid-core, bicomponent and monocomponent filaments for technical textiles.

Previously, she was a Postdoc at the University of Fribourg, Switzerland, investigating the physical properties of thin films. She also worked at the Argonne National Laboratory, Chicago, USA on the physical properties of cathodes for fuel cells. She has led projects at large-scale research facilities in the USA, Germany and Switzerland. Dr. Perret obtained her PhD in Physics from the Paul Scherrer Institute, ETH Zurich.

Léo Sifringer (CH)

PhD candidate, Laboratory for Biosensors and Bioelectronics (LBB), Department of Information Technology and Electrical Engineering, ETH Zürich

4165: A soft, implantable nerve model on a chip towards biohybrid regenerative electronics

L.Sifringer1, A. Fratzl2, B. Clément1, P. Chansoria4, S.J. Ihle1, B. Maurer1, L. Mönkemöller1, J. Duru1, C. M. Tringides1, K. Vulić1, A. Beltraminelli1, S. Steffens1, E. Ceylan1, S. Weaver1, G. Amos1, S. Madduri3, M. Zenobi-Wong4, B. Roska2, J. Vörös1 and T.Ruff1

1Laboratory of Biosensors and Bioelectronics, ETH and University of Zurich, Zurich, Switzerland.

2Institute of Molecular and Clinical Ophthalmology Basel, Basel, Switzerland.

3Center for Bioengineering and Regenerative Medicine, University of Basel, Switzerland.

4Laboratory for Tissue Engineering and Biofabrication, ETH Zurich, Switzerland.

 

INTRODUCTION: In this work, we introduce a novel nerve-on-a-chip model designed as a neural interface for deep brain stimulation. Termed as a “biohybrid” approach, it aims to overcome the limitations of standard deep brain implants such as low stimulation resolution. The biohybrid concept leverages on-chip grown retinal neurons to convert electrical signals from a stretchable 2D microelectrode array into synaptic stimulation of a neural target tissue. A photograph of the device can be seen in Figure 1.

 

METHODS: The device consists of two primary components: a soft, stretchable multi-electrode array (MEA) and an axon-guiding microstructure [1]. The MEA, fabricated using a transfer stripping method [2], comprises a PDMS substrate and microstructured platinum tracks. The PDMS microfluidic axon guidance structure is aligned and bonded onto the microelectrode array. Living retinal neurons labeled with a viral vector (mRuby3) are then seeded into 16 seeding wells and cultured under standard cell culture conditions before implantation. The seeding procedure is depicted in Figure 2a.

Fig. 1: a) photograph of the device and b) zoom-in on the microstructure used for neuron culturing

 

RESULTS: We explain the fabrication of the biohybrid neural interface and demonstrate how the retinal ganglion cells seeded into the implant form an artificial optic nerve of up to 3 mm in length (Figure 2b). We demonstrate how axons transit from the biohybrid implant into a nerve-forming bioresorbable collagen tube that will guide axons from the implant towards a neural target structure for sensory reinnervation and synaptic stimulation of the visual thalamus in vivo.

 

Fig. 2: a) Schematic of neuron seeding and growing in the microstructure. b) Fluorescence imaging of retinal neurons growing in the device.

 

We show that retinal spheroids can be stimulated using the stretchable microelectrode array. To assess stimulation-induced signal transmission in the biohybrid implant we present in vitro data on how spikes propagate within the axon guidance channels using CMOS[3] multielectrode arrays. Lastly, we demonstrate that neurons cultured in the device survive for over 3 weeks when implanted in vivo.

 

DISCUSSION & CONCLUSIONS: With this work, we show that this biohybrid approach has the potential to serve as a new kind of neural interface technology. Although further experiments are necessary for in vivo deep-brain stimulation, our results highlight the feasibility of this approach.

 

REFERENCES: 1C. Forró et al., Biosens. Bioelectron. 122:7587, 2018, 2R.F. Tiefenauer et al., ACS Nano 12:2514-2520, 2018, 3J. Duru et al., Front. Neurosc. 16:829884, 2022

 

ACKNOWLEDGEMENTS: This work was supported by the Human Frontiers Science Project (HFSP) postdoc fellowship, the OPO foundation, Swiss National Science Foundation and ETH Zürich.

I am a PhD candidate in the Laboratory for Biosensors and Bioelectronics (LBB) at ETH Zürich.
Originally from an engineering background, I‘m passionate about creating tools that help us understand how the human brain and nervous system work. My work currently focuses on interfacing stretchable electronics and engineered neuronal networks for in vitro and in vivo applications, such as biohybrid implants, and studying the mechanobiology of neuronal networks.

Dr. Romy Marek (CH)

PhD candidate, Laboratory for Biosensors and Bioelectronics (LBB), Department of Information Technology and Electrical Engineering, ETH Zürich

4169: Enhancing the degradation behavior and osseointegration of resorbable Mg-Zn-Ca orthopedic screws by PEO surface treatment

Marek1,2, K. Kaufmann2, M. Honea2,4, T. Imwinkelried3, P. Holweg2, F. Warchomicka4, L. Berger5, A.M. Weinberg2

1 Institute for Medical Engineering and Medical Informatics, School of Life Sciences, FHNW Muttenz, Switzerland.

2 Department of Orthopaedics and Traumatology, Medical University of Graz, Austria.

3 RMS Foundation, Bettlach, Switzerland.

4 Institute for Materials Science, Joining and Forming, Technical University of Graz, Austria.

5 Department of Materials, ETH Zürich, Switzerland.

 

INTRODUCTION: Magnesium (Mg)-based implants are promising to overcome common drawbacks of gold-standard orthopedic implant materials, such as long-term foreign body reactions and stress shielding effects [1]. The major advantage of Mg-alloys poses their ability to degrade within the body, eliminating the need for removal surgeries [2]. However, regulating the degradation rate of Mg is challenging. Accelerated degradation may lead to extensive hydrogen gas accumulation in the surrounding tissues, which compromises bone-to-implant contact (BIC) and screw stability within the bone structure [3]. Ensuring a slow and homogeneous degradation of the implant is crucial to enable a consistent substitution of deteriorating implant material with newly formed tissue. Several factors influence the degradation behavior of Mg-based materials, such as the incorporation of alloying elements, the material’s microstructure, or surface treatments [4]. In this study, the influence of a plasma-electrolytic-oxidation (PEO) surface treatment on the in vivo degradation behavior and osseointegration of the Mg-zinc(Zn)-calcium(Ca) alloy ZX00 (Mg, Zn<0.5, Ca<0.5; in wt.%) was evaluated in a sheep model over 24 weeks.

 

METHODS: A cohort of 37 juvenile sheep was operated for this study. Up to three screws were implanted into both tibial shafts of all sheep, using ZX00-, PEO-treated ZX00 (PEO-ZX00)-, and Ti screws. Euthanasia of the animals was performed 0, 3, 6, 12 or 24 weeks post surgery and their tibiae were extracted. Micro-computed tomography (µCT) scans with a voxel size of 20.3 µm were taken on all the extracted tibiae. To investigate the degradation behavior of ZX00 and PEO-ZX00 screws, the µCT data were evaluated regarding implant and hydrogen gas volume, as well as the degradation rate at the given time points. Furthermore, histological evaluations were carried out on embedded, un-decalcified bone samples, to elaborate the bone quality adjacent to the ZX00-, PEO-ZX00 and Ti screws. To assess the screw stability of the different materials in the bone at the given time points, biomechanical pullout testing was carried out.

 

RESULTS: Evaluation of the ex vivo µCT data revealed a homogeneous degradation behavior for both, ZX00 and PEO-ZX00 screws over the study duration of 24 weeks. A decreased degradation rate was computed for the PEO-ZX00 screws compared to the ZX00 screws and a delayed hydrogen gas formation was detected. Furthermore, histological investigations revealed an enhanced BIC around PEO-ZX00 screws compared to the ZX00 screws and higher pullout forces were measured.

 

Figure 1. Ex vivo µCT images of a ZX00 (a) and a PEO-ZX00 (b) screw 12 weeks after implantation into the tibial shaft of juvenile sheep.

 

DISCUSSION & CONCLUSIONS: The decreased degradation rate and enhanced BIC observed for the PEO-ZX00 screws compared to the ZX00 screws indicates effectiveness of the PEO surface treatment. Further evaluations with PEO-ZX00 screws in fracture models are suggested to verify their ability to promote bone fracture healing.

 

REFERENCES: 1 P. Thomas et al (2011) J. Bone Joint Surg. Am., 93:e61. 2 J. M. Seitz et al (2013) J. of Biom. Mat. Res. A, 102:10, pp 3744-3753. 3 F. Amerstorfer et al. (2016) A. Biomat., 42, pp440-450. 4 M. M. Gawlik et al (2018) Materials (Basel), 11(12):2561

 

ACKNOWLEDGEMENTS: This project has received funding from the European Union´s Horizon 2020 research and innovation programme under the Marie Sklodowska-Curie grant agreement No 811226 and from RMS Foundation (RMS Foundation, 2544 Bettlach, Switzerland).

I currently hold a research position at the Institute for Medical Engineering and Medical Informatics within the School of Life Sciences at FHNW in Muttenz. My work focuses on medical additive manufacturing and implant development, with a special emphasis on resorbable metallic materials.

Paula De Dios Andres (DK)

PhD candidate, Cell Mimicry Group, Interdisciplinary Nanoscience Center (iNANO), Aarhus University

4173: Interfacing artificial cells with hepatocytes

P. de Dios Andres1, B. Städler1

1Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Aarhus, Denmark.

 

 

INTRODUCTION: The collaborative interfacing of biomaterials (i.e., artificial cells) and mammalian cells remains challenging. For example, alginate microgels with encapsulated catalase and coated with poly (L lysine) were co-cultured with hepatocytes, integrating synthetic and biological partners, where the former provides supportive detoxification capabilities.[1] However, the artificial cells were only active for 24 h, the level of integration was limited due to simplistic interface design in uncontrolled cell aggregates.

Here, we use 3D bioprinting of artificial cells and HepG2 cells towards the creation of bionic tissue. Additionally, these artificial cells, were equipped with a small organic compound that acted as an artificial enzyme with CYP450-imitating activity. [2] In addition, the artificial cells were camouflaged with cell membrane vesicles and equipped with and antioxidant to support HepG2 cells against reactive oxygen species. [3]

 

METHODS: Cell membrane vesicles (CMVs) were purified from HepG2 cells by homogenization and centrifugation to separate the membrane from other organelles. Then, the membranes were extruded through 400 and 200 nm membranes and cleaned by size exclusion chromatography (SEC). Alginate microgels were produced using an encapsulator (B-390 from Buchi, Switzerland). These artificial cells were coated with poly(L lysine) (PLL) and loaded with metalloporphyrin that mimics the enzyme activity of CYP1A2 [4], or EUK [5] to have catalase-like and superoxide dismutase-like activity. HepG2 cell aggregates were grown for 2 days before suspension in a composite ink composed of 1.5% w/v alginate, 5% w/v Gelatin metacryloyl (GelMA) and 25% v/v artificial cells. The structures were printed using a bioprinter (BioX) and cultured for up to 35 days. The proliferation of the bionic tissues was analysed by DNA quantification and LIVE/DEAD staining and the cytochrome (CYP1A2) expression was quantified by RT-qPCR. The artificial cells were co-cultured with HepG2 cells in 96-well plates with an ultralow attachment surface in different cell-to-AC ratios. The integration and proliferation of these bionic aggregates was visualized by confocal laser scanning microscopy (CLSM).

The bionic aggregates were exposed to different doses and concentrations of tert-butyl hydroperoxide (tBuOOH) and the reactive oxygen species (ROS) scavenging capabilities of the ACs were demonstrated by assessing the proliferation, DNA quantification and mitochondrial markers of the bionic aggregates.

 

RESULTS: HepG2 cell were cultured in non-adhesive flasks for 48 h, forming HepG2 cell aggregates. These aggregates were 3D bioprinted using either an alginate/gelatin methacryloyl ink or a composite ink made of an alginate/gelatin liquid phase with increasing amount of artificial cells (Figure 1a). The proliferation of the bionic tissues was visualized by CLSM for up to 35 days. Cell membrane vesicles (CMVs) were formed by isolation of the cell membrane of HepG2 cells and they were characterized confirming its vesicular shape and the presence of N-cadherins. Then, alginate microgels were coated with the CMVs resulting in artificial cells (ACs). Following on, the ACs were cocultured with HepG2 cells in different cell-to-AC ratios (Figure 1b) over 5 days and the integration of ACs was visualized by CLSM. Finally, the bionic 3D prints were exposed to different concentrations of tBuOOH and REE in different time intervals, and the survival and CYP1A2 activity was assessed, respectively.

 

Fig. 1: a) Cartoon showing the composite bioink consisting of HepG2 cell aggregates and artificial cells. b) Schematic showing the EUK liposomes and the ACs coated with CMVs and their integration with HepG2 cells.

 

DISCUSSION & CONCLUSIONS: Artificial equipped with metalloporphyrin, imitating the catalytic functions of CYP450 enzymes, were included within 3D bioprinted structures alongside HepG2 cells, which proliferated for at least 35 d. CMV camouflaged artificial cells were more successfully integrated with proliferating HepG2 cells compared to PLL-coated versions. This 3D bionic tissue where the artificial cells supported their biological counterparts within a tissue-mimicking milieu, exhibiting augmented hepatocyte-like function.

 

REFERENCES: 1 Y. Zhang, et al (2017) ACS Omega, 2 (10) 7085–7095; 2I. N. Westensee, L. J. Paffen, et al. (2024) Adv. Healthcare Mater., 2303699; 3P. de Dios Andres et al. (In preparation); 4X. Qian, I. N. Westensee, et al., (2021) Angew. Chem. Int. Ed., 60, 18704.; 5C. Ade et al., (2019) ACS Appl. Polym. Mater., 1, (6), 1532–1539

 

ACKNOWLEDGEMENTS: The authors acknowledge the European Research Council (ERC) under the European Union’s Horizon 2020 research and innovation program (grant agreement No. 818890).

I have a bachelor’s degree in chemistry from the University of Zaragoza, Spain and a MSc in Nanoscience from the University of Aarhus, Denmark where I focused on biomedical nanoscience. The topic of my MSc thesis was the development of dynamic 3D cell models using structured paper chips. Currently, I am pursuing a PhD in Nanoscience at Aarhus University, Denmark, working with Prof. Brigitte Städler in the Cell Mimicry Group. My research evolves around the creation of liver-like tissues by combining bottom-up synthetic biology with 3D bioprinting aiming to develop alternative clinical interventions liver disease or to establish minimal systems to address complex biological questions in the context of hepatology. In this context, artificial cells imitate selected structural and functional properties of the mammalian cells with the goal to support, augment or replace aspects of their living role models.

Dr. Santosh Sadashiv Mathapati (IN)

Associate Professor, Translational Health Science and Technology Institute India

4174: Placental ECM hydrogels: a promising biomaterial

Sunil Gujjar1, Anurag Tyagi1, Saloni Sainger1, Puja Bharti2, Vaibhav Nain1, Pratibha Sood1, Prakash Jayabal1, Jagadish Chandra Sharma3, Priyanka Sharma3, Sanjay Rajput3, Anil Kumar Pandey3, Amit Kumar Pandey3, Prasad Abnave2, Santosh Mathapati1

1Translational Health Science and Technology Institute, Faridabad, Haryana, 121001 India.

2National Centre for Cell Science, Pune, Maharashtra, 411007 India.

3Employees State Insurance Corporation Medical College and Hospital, Faridabad, Haryana, 121012 India

 

INTRODUCTION: The decellularized extracellular matrix (ECM) bioscaffolds offer significant tissue repair and regeneration potential1. Placental tissue, readily available as medical waste, is a particularly promising source for these scaffolds due to its similarity to fetal tissues2. This study investigates a new hydrogel preparation method using placental ECM and lays the groundwork for detailed characterization of the resulting material.

 

METHODS: This study explored the enhancement of perfusion-based placental decellularization using detergents and enzymes. The resulting acellular placenta was then pulverized into a powder (P-ECM) using a cryo-miller and liquid nitrogen. P-ECM hydrogels were formed using 10 mg/ml (10P-ECM), 15 mg/ml (15P-ECM), and 20 mg/ml (20P-ECM) digested in a solution containing 0.1M HCl and 1 mg/ml pepsin at a concentration of 10 mg/ml at RT and neutralized by adding 1M NaOH to induce gelation at 37°C. Finally, the pregel was incubated at 37°C, triggering this solution’s polymerization into a hydrogel. These hydrogels’ physical and biological properties were then evaluated and compared with collagen hydrogels.

 

RESULTS: Decellularization over 72 h showed a reduction in cellular and nucleic acid materials (revealed by H&E and Hoechst staining) compared to shorter treatment periods (24 and 48 h). Histology and biochemical analysis confirmed the preservation of collagen and glycosaminoglycans within the decellularized tissues (72 h) compared to native placental tissue. Pepsin digestion in an acidic solution successfully converted the P-ECM into a pregel solution. Neutralization (with 1M NaOH) and incubation at 37 °C successfully induced gelation of the pregel, forming the final hydrogel. The 15PECM and 20P-ECM hydrogels possess sufficient strength to resist early degradation. Hydrogels with higher crosslinking densities tend to have improved mechanical strength and stability. Therefore, 20P-ECM hydrogel, with its lesser degradability, may exhibit enhanced mechanical properties compared to the other hydrogels. The collagen hydrogel showed a higher storage modulus than 15P-ECM and 20P-ECM hydrogels. Live/dead and MTS assays demonstrated the biocompatibility of P-ECM hydrogels with both endothelial and fibroblast cells (Fig. 1). Proteome profiling of P-ECM hydrogels demonstrated the relative abundance of Serpin E1 and IGFBP1 proteins. The animal study investigating skin wounds found that 20P-ECM treatment demonstrated biocompatibility compared to the control group.

 


Fig. 2 A) Cell biocompatibility for the 20P-ECM and B) Cell migration assay2.

 

DISCUSSION & CONCLUSIONS: This study investigates the decellularization of human placental tissue for hydrogel production, focusing on the impact of ECM concentration on resulting properties.  A perfusion-based decellularization method was used, and the effect of digestion time on hydrogel characteristics was analyzed. P-ECM hydrogels showed process-dependent improvements in biocompatibility and bioburden reduction. While promising for clinical applications, mechanical strength, sterility, and degradation control enhancements are necessary for broader implementation.

 

REFERENCES: 1 Saldin T, Cramer M et al., Extracellular Matrix Hydrogels from Decellularized Tissues: Structure and Function, Acta Biomater. 2017. 2 Gujjar S, Tyagi A, Sainger S, et al., Biocompatible Human Placental Extracellular Matrix Derived Hydrogels. Adv Biol (Weinh). 2024:e2300349.

 

ACKNOWLEDGEMENTS: This work was supported by the Science and Engineering Research Board (Grant ID: SRG/2021/000420), New Delhi, India.

Dr. Santosh S. Mathapati (B.V.Sc., PhD) is an Associate Professor at BRIC-Translational Health Science and Technology Institute, Faridabad, India. Their research expertise lies in tissue engineering and regenerative medicine, particularly biomaterials, tissue interactions, and the development of bioengineered tissues, especially liver models.

Camillo Colli (IT)

PhD Candidate, Chemical Engineering, Chemistry, Materials and Chemical Engineering, Politecnico di Milano

4177: Thermoresponsive UCST nanoparticles for controlled drug delivery in cancer hyperthermia treatment

Colli1, M. Sponchioni1, L. Rosanò2, E. Mauri1, D. Moscatelli1

1Dept. Chemistry, Materials and Chemical Engineering “G. Natta”, Politecnico di Milano, Milan, Italy.

2Institute of Molecular Biology and Pathology, National Research Council (CNR), Rome 00185, Italy.

 

INTRODUCTION: Thermoresponsive polymers are a promising class of materials for the design of smart nanocarrier aimed to drug delivery purposes. Tumour thermotherapies, such as hyperthermic intraperitoneal chemotherapy (ovarian cancer), are still demanding for an approach able to counteract the thermotolerance and drug-resistance of specific tumoral cells, which limits the curative effects.

Here, we proposed the synthesis of thermoresponsive nanoparticles (NPs) with tunable Upper Critical Solution Temperature (UCST) to intracellularly deliver a chemotherapeutic and release it on-demand, through hyperthermia stimulation. In particular we have tuned the UCST of NPs to match the needs of ovarian hyperthermia treatments at 43 °C and demonstrated their efficacy as drug delivery system.

 

METHODS: The block-copolymer used in NP synthesis is composed of an UCST zwitterionic portion and a biodegradable hydrophobic block. The former was obtained via RAFT polymerization of sulfobetaine (SB) and sulfabetaine (ZB), obtaining a copolymer (p(SB-co-ZB)) with a degree of polymerization DP = 200. The lipophilic macromonomer was produced via ring opening polymerization (ROP) of ε-caprolactone initialized with HEMA and Sn(Oct)2, obtaining a DP = 5 (HEMACL5). Finally, NP synthesis was performed via RAFT emulsion polymerization at 65°C adding p(SB-co-ZB)200 to HEMACL5, in a buffer. The cloud point (Tcp) of the NPs was detected via DLS. Paclitaxel (PTX) was encapsulated in the NPs  exploiting the phase-transition (i.e., above UCST) and the drug release was evaluated at 37°C and in hyperthermia condition (43°C). Drug-loaded NPs were administered to thermoresistant SKOV3 cells and the therapeutic effects evaluated via MTT assay.

 

RESULTS: Tuning the ratio SB/ZB, NPs with different UCST values (range 30°-43°C) were obtained. According to the hyperthermia treatment, we chose NPs with Tcp close to 43°C and we observed an induced drug release at that temperature only. The administration of PTX-loaded NPs to cellsboosted the therapeutic effect of the drug after hyperthermia, already within the first 24 h, outperforming the effect of the non-encapsulated drug.

 

Fig. 1: a) NP structure; b) Drug release profile at 37°C and 43°C (hyperthermia); c) drug-induced cytotoxicity in SKOV3 at 24h after hyperthermia and administration of free-drug (PTX) and drug-loaded NPs (NP+PTX).

 

DISCUSSION & CONCLUSIONS: The synthesis of well-defined block copolymers via combination of ROP and RAFT polymerization ensures the formation of NPs with tunable UCST. The use thermoresponsive NPs as drug delivery systems in ovarian cancer hyperthermic treatment represents an innovative strategy to counteract the progression of the disease, still lacking an efficient treatment. 

 

REFERENCES: 1 Sponchioni M. et al (2019), Mater Sci Eng C, 102, 589-605. 2 Kong X. et al (2020), Biomed Pharmacother, 132, 110907. 3 Sponchioni M. et al (2019), Nanoscale, 11, 16582-16591.

Camillo Colli is a PhD student in chemical engineering at Politecnico di Milano. His research focuses on polymeric materials for drug delivery applied to different types of tumors with synthesis of nanoparticles/nanogels in microfluidics, fluid dynamic modeling in microreactors and applications on 2PP 3D printing. He graduated in chemical engineering at Politecnico di Milano, and is a lover of the challenges of treating diseases effectively.

Annina Stuber (CH)

PhD Candidate, Laboratory of Biosensors and Bioelectronics, Deptartment of Information Technology and Electrical Engineering, ETH Zürich

4187: Sensing neurotransmitters with aptamer-modified nanopipettes at the interface of neuronal systems

Annina Stuber1,2 , Yassine Massoud1, Anna Burdina1, Nako Nakatsuka1,2

1Laboratory of Biosensors and Bioelectronics, ETH, Zurich, Switzerland.
2Laboratory of Chemical Nanotechnology, EPFL, Geneva, Switzerland.

 

INTRODUCTION: Developing tools to untangle the complex signalling processes taking place in the brain, is a key step to glean insights into brain (dys)function. Neural communication is composed of a harmony of dynamic electrical and chemical signals. We have developed nanopore sensors that enable the measurement of the local concentration of specific neurotransmitters with nanoscale resolution.

 

METHODS: We covalently bind aptamers inside a quartz nanopipette’s orifice. Aptamers are single-stranded DNA/RNA sequences, artificially engineered to bind a target of interest (e.g., dopamine). Target binding induces aptamer conformational change, rearranging its negatively charged backbone1 within the nanopipette’s opening (Fig. 1). This DNA structure switching alters the ionic current through the nanopore, resulting in a measurable signal change upon target recognition.2

 

 

Fig 1: Schematic representation of (left) an aptamer modified nanopipette measurement setup and (right) aptamers undergoing structural rearrangement upon target binding, inside of the nanopipettes orifice.


RESULTS: We tested the selectivity of the aptamer-modified nanopipettes using analytes structurally similar to dopamine (Fig. 2). While sensor selectivity has been validated, nanopipettes are vulnerable to environmental changes (i.e. ionic content, temperature, pH). Thus, measurements in complex and dynamic environments require direct referencing with a control sensor. We developed a control nanopipette, functionalized with a scrambled DNA sequence, retaining the same chemical signature (bases and charge) in an altered order, thus, inert to the analyte of interest. We illustrate the feasibility of detecting endogenous dopamine in murine brain slices, by implanting a specific and control sensor in proximity to a stimulator3 (Fig. 3a). Post electrical stimulus, we observe a change in current in our dopamine sensor, while the control sensor remains stable (Fig. 3b).

 

Fig 2: Sensor response to dopamine-structurally-similar molecules.

 

 

Fig. 3: a. Schematic of specific and control sensors in brain tissue, positioned near a stimulator. b. Current responses of specific and control sensors after electrical stimulation.

 

DISCUSSION & CONCLUSIONS: We demonstrate the feasibility of measuring neurotransmitters in complex biological environments. We still face spatial resolution challenges in our self-referencing, due to the nanopipette positioning limited by microscope resolution. We are working towards specific and control sensor incorporation, at nanoscale proximity in double-pore nanopipettes, which would allow us to differentiate specific vs. nonspecific interactions when detecting neurotransmitters at the interface of biological systems.

REFERENCES: 1 N. Nakatsuka, et al. Science, 362: 319-324, 2018 2    N. Nakatsuka et al. Anal. Chem., 93 (8), 4033-404, 2021. 3 A. Stuber et al. ACS meas. sci. au, 4,1,92-103, 2023

 

ACKNOWLEDGEMENTS: This work was supported by ETH Zürich. The authors would like to thank Janos Vörös for helpful discussions and Anna Cavaccini and Theofanis Karayannis for their collaboration and experimental aid.

I am in my third year of PhD studies in the Laboratory of Biosensors and Bioelectronics (LBB) at ETH (Zürich, Switzerland). My research is focused on creating chemical sensors capable of measuring neurotransmitters. I am motivated to push future work to develop novel sensing techniques enabling patients to monitor neurological conditions, allowing for improved diagnostic and treatment approaches.

Dr. Shabnam Tarvirdipour (CH)

Post-doctoral scientist, Department of Chemistry, University of Basel

4245: Co-delivery of an antisense oligonucleotide and photosensitizer using a peptide nanocarrier

Tarvirdipour1, M. Skowicki1,2, V. Maffeis1,2, S. N. Abdollahi1, C. A. Schoenenberger1, C. G. Palivan1,2

1 University of Basel, Basel, Switzerland. 2NCCR-Molecular Systems Engineering, Basel, Switzerland. 

 

INTRODUCTION: There is a growing demand for advanced nano-systems that can deliver multiple therapeutic agents while maintaining the functionality of each component 1. The aim of this combined delivery is to enhance the effectiveness of each treatment, promote synergistic effects, and thereby increase the overall efficacy of the delivery system while reducing drug resistance 1. However, implementing combination therapy within a single delivery system is significantly more complex than monotherapy, as it requires platforms with specific physicochemical properties to accommodate different therapeutic agents. The loading capacity, stability, biocompatibility, targeting, and efficient intracellular delivery of nanocarriers must be precisely optimized to achieve a site-specific synergistic effect 2,3. In this study, we investigate a peptidic nanocarrier for its ability to load a photosensitizer (PS) and antisense oligonucleotides (ASO) and deliver both to breast cancer cells 3.

 

METHODS: A multi-compartment micellar platform (MCM) was developed by self-assembly via the solvent exchange method. The supramolecular architecture was assessed using transmission electron microscopy (TEM) and atomic force microscopy (AFM). The size of NLS-MCMs was measured using dynamic light scattering (DLS), and their surface charge was measured with a Zeta Sizer Nano ZSP. Fluorescence correlation spectroscopy (FCS) was employed to analyze ASO entrapment in NLS-MCMs. Cellular uptake was examined on SK-BR-3 cells using confocal laser scanning microscopy (CLSM).

 

RESULTS: TEM and AFM images showed that PS + ASO-loaded NLS-MCMs have a spherical, multi-micellar structure and a raspberry-like surface (Fig. 1A, C, and D). Dynamic light scattering (DLS) showed nanocarriers have an average hydrodynamic diameter (DH) of 112 ± 6 nm with a polydispersity index (PDI) of 0.17 (Fig. 1B). Zeta potential measurements indicated an average surface charge of 23 ± 5 mV, contributing to nanocarriers colloidal stability (Fig. 1E). FCS confirmed ASO entrapment, with diffusion time increasing from 114 ± 2 μs for free ASO to 3705.9 ± 1486 μs for loaded NLS-MCMs, and an average of 12.8 ± 4.3 ASO molecules per NLS-MCM (Fig. 1F). After 24 hours of treatment with PS + ASO-loaded NLS-MCMs, CLSM images revealed efficient cellular uptake (Fig. 2).

 

Fig. 1: Physicochemical characterization of PS + ASO-loaded NLS-MCMs. (A) Ultrastructural morphology, (B) Size distribution, (C) AFM topography, (D) AFM height profile along the dashed orange line, (E) Hydrodynamic size and zeta potential as a function of storage time, and (F) Normalised FCS autocorrelation curves. Scale bars = 200 nm.

 

Fig. 2: Cellular uptake of PS + ASO-loaded NLS-MCMs (upper panels, cells incubated with PS + ASO-loaded NLS-MCMs; lower panels, control). Scale bars, 20 μm.

 

DISCUSSION & CONCLUSIONS: We developed a peptidic multi-micellar platform that efficiently encapsulates a hydrophobic photosensitizer (Ce6) and a negatively charged antisense oligonucleotide (G3139-GAP). This platform utilizes hydrophobic and electrostatic interactions in a one-pot assembly process and includes a positively charged NLS for enhanced cellular uptake and nuclear targeting. Our studies show that combining gene therapy with photodynamic therapy increases target cell sensitivity to destruction upon light activation, achieving a synergistic effect. Co-loading led to higher cytotoxicity and greater therapeutic efficacy compared to free forms or single payloads. This study advances the use of peptide amphiphiles in therapeutic strategies, offering significant implications for biomedical research.

 

REFERENCES: 1 B. G. Carvalho, F. F. Vit, H. F. Carvalho, S. W. Han, L. G. de la Torre (2021) Journal of Materials Chemistry B, 5: 1208-1237. 2 S. Tarvirdipour, X. Huang, V. Mihali, C. A. Schoenenberger, C. G. Palivan (2020) Molecules 15: 3482. 3 S. Tarvirdipour, M. Skowicki, V. Maffeis, S. N. Abdollahi, C. A. Schoenenberger, C. G. Palivan (2024) Journal of Colloid and Interface Science, 664: 338-348.

 

ACKNOWLEDGEMENTS: The authors highly appreciate the financial support provided by the National Centre of Competence in Research–Molecular Systems Engineering (NCCR-MSE), the Swiss National Science Foundation and the University of Basel. 

I completed my PhD in Nanoscience from May 2017 to August 2021 as a joint student at the University of Basel (Department of Chemistry) and ETH Zurich (Department of Biosystem Science and Engineering), under the Swiss Nanoscience Institute (SNI). My thesis focused on the rational design and development of self-assembling peptide amphiphiles for gene therapy. Since August 2021, I have been a postdoctoral scientist at the University of Basel. My research interests lie at the intersection of nanotechnology and biomedicine, where I explore cutting-edge strategies for developing advanced delivery platforms tailored for therapeutic applications. Specifically, I focus on engineering nanocarriers for the co-delivery of genes and photosensitizers to optimize treatment outcomes. Currently, I am also developing peptide-decorated solid supports as innovative antimicrobial platforms, integrating the latest advancements in material science and biotechnology to address the challenges associated with this field.

Dr. Ionel Adrian Dinu (CH)

Senior scientific collaborator, Department of Chemistry, University of Basel

4284: Designing catalytic nanocompartments to support parallel reactions and promote localized cell internalization

Korpidou1, C.G. Palivan1,2, I.A. Dinu1,3

1 Department of Chemistry, University of Basel, Basel, Switzerland.

2 Swiss Nanoscience Institute, University of Basel, Basel, Switzerland.

3 NCCR-Molecular Systems Engineering, Basel, Switzerland.

 

 

INTRODUCTION: Enzyme-based therapies show promise for various pathologies but are limited by tissue specificity and immunogenicity [1]. Herein, we introduce catalytic nanocompartments (CNCs) made of PDMS-b-PMOXA diblock copolymers [2-4], co-encapsulating beta-glucuronidase (GUS) and glucose oxidase (GOx), and decorated with mannose-containing glycooligomers. The enzymes provide multifunctionality through in situ parallel reactions: GUS reactivates the drug hymecromone, while GOx induces cell starvation by depleting glucose and generating cytotoxic H2O2. The insertion of the pore-forming peptide, melittin, into the polymer membrane facilitates diffusion of substrates and products. Enhanced cell-specific internalization of CNCs reduces the viability of HepG2 cells due to the combined effect of the drug and reactive oxygen species.

 

METHODS: Our nanocompartments were prepared by film rehydration from a mixture of unfunctionalized and azide-functionalized PDMS-b-PMOXA diblock copolymers and designed as CNCs by encapsulation of two therapeutically relevant enzymes. For targeting the cells and favor cellular uptake, glycooligomer tethers consisting of eight pendant mannose units were attached to the azide-groups exposed on external interface of CNCs membrane. Using a combination of techniques, the resulting polymersomes and CNCs were thoroughly characterized in terms of shape, size and encapsulation efficiency. Two cell lines were chosen for cell targeting, each possessing different expression profiles of mannose-binding lectins: liver-derived HepG2 cells with high expression levels and cervix-derived HeLa cells expressing lower levels of receptors. To study the effect of simultaneous production of intracellular hymecromone and H2O2 on cell viability, MTS cell proliferation assays were performed.

 

RESULTS: To explore cell uptake specificity of glycooligomer-decorated polymersomes (gly-Ps), HepG2 and HeLa cells were co-cultured and then incubated with Atto647-loaded gly-Ps for 24 h. Analysis of CLSM micrographs and quantification of fluorescence intensity corresponding to Atto647 showed that gly-Ps were about 3-times more abundant in HepG2 cells (Fig. 1). The results highlight the key role of glycooligomers in targeting liver cells over-expressing mannose-binding lectins and the effect of bioorthogonally generated hymecromone and H2O2 on cell viability.

 

Fig. 1: 3D reconstruction of multiple confocal sections of HepG2 (blue nuclei) and HeLa (green nuclei) co-cultured cells incubated with gly-Ps (yellow). Cell membrane (red), scale bar:10 μm.

 

DISCUSSION & CONCLUSIONS: This elegant approach improves the enzyme-based therapeutics by co-encapsulating two enzymes in synthetic nanocompartments, and further functionalized with glycooligomer tethers. These targeted CNCs efficiently catalysed two independent reactions in parallel, achieving controlled localization at the target site. Selective internalization into HepG2 cells was facilitated by the glycooligomers on the CNC surface, which interact with overexpressed mannose-binding receptors. The synergistic effect of hymecromone and H2O2 significantly reduced HepG2 cell viability. This study offers an optimal strategy for drug synergism and contributes to the demand for innovative, combinatorial strategies in cancer therapy.

 

REFERENCES: 1 M. de la Fuente, L. Lombardero, A. Gómez-González, et al (2021) Int J Mol Sci 22:9181. 2 M. Korpidou, V. Maffeis, I.A. Dinu, et al (2022) J Mater Chem B 10:3916-26. 3 A. Guinart, M. Korpidou, D. Doeller, et al (2023) PNAS 120:e2301279120. 4 M. Korpidou, J. Becker, S. Tarvirdipour, et al (2024) Biomacromolecules DOI: 10.1021/acs.biomac.4c00526.

 

ACKNOWLEDGEMENTS: The funding from University of Basel, NCCR—Molecular Systems Engineering, and BIOMOLMACS, a European project funded by the European Union´s Horizon 2020 research and innovation program under the Marie Skłodowska-Curie grant agreement no. 859416 is gratefully acknowledged.

My research focus is oriented towards the development of multifunctional polymer materials by combining polymers with small molecules into supramolecular structures at the nano- and microscale. Recent and current research topics are focused on three directions: (1) polymer carriers and catalytic nano-/micro-compartments with stimuli-triggered functionalities for applications in water purification, food packaging, medicine, etc.; (2) nano-/micro-structured composite hydrogels and cryogels for high-throughput recognition and separation of ions and biomolecules by entrapment/attachment of polymer self-assemblies or nano-/microgels on hydrogel or cryogel networks; (3) self-organized pervaporation membranes with improved separation performances in dehydration of alcohols.

Start-up pitches

Dr. Chris Steffi (CH)

Co-Founder and CSO, CompagOs

Personalizing bone health with 3D printed organoids

CompagOs, a Swiss Spin-off from ETH Zürich, aims to transform bone health by leveraging advanced 3D bioprinting technology to develop Bon3OID™, our reproducible in vitro bone models. These models are used for basic research, drug efficacy, and personalized medicine, focusing on bone biology, rare bone diseases, and bone oncology. Our primary focus is on cancerous bone conditions, aiming to enhance early diagnosis.

Looking forward, we aspire to redefine personalized bone healthcare, setting new standards for diagnostics to enable the prognosis of a disease with the potential to predict treatment responses. Our diagnostic tool, Bon3OID™-Dx, is designed to detect cancer metastasis from blood earlier than traditional clinical imaging by mimicking the unique diseased bone condition in an organotypic patient-specific bone model.

In addition, we are committed to reducing reliance on animal testing by creating accessible, 3D human bone organotypic models for researchers worldwide.

Dr. Chris Steffi is a Postdoctoral Research Fellow at ETH Zurich and one of the co-founders of CompagOs, an ETH Zurich Spin-off. During her PhD and postdoctoral work at the National University of Singapore, she developed in vitro models to study drug delivery and biomaterial functionalization for bone tissue engineering. Currently, Dr. Steffi focuses on in vitro bone models for fracture phenotypes and osteosarcoma for personalized medicine, expansion, and biobanking of rare disease samples. She is a recipient of the NUSMed Postdoctoral Fellowship, NUS Research Fellowship, and the Council of Scientific & Industrial Research Catch Them Young scholarship.

Dr. Frank Bonnet (CH)

CEO, Biononous

AI-powered solutions for screening, sorting and dispensing of small biological entities

Bionomous is a spin-off from the École Polytechnique de Lausanne. Founded in 2019, the company currently employs 15 people in Switzerland and 1 person in the United States. The company develops and commercializes laboratory instruments that allow the automatic screening, sorting and plating of small biological entities, which are used as models in research, to facilitate researchers‘ work while increasing the efficiency of their work. The company‘s primary application is sorting eggs of fish research models, such as zebrafish, but it also has clients using the machine to sort flower seeds, plant cells aggregates and organoids. To date, the company has successfully commercialized its products in several countries, including in Europe, the United States, and China.

Frank Bonnet holds a Master‘s degree and a PhD in Robotics from the École Polytechnique Fédérale de Lausanne. During his thesis work, Frank developed robotic systems for studying biological systems, which later led to the technology behind Bionomous. After receiving the award for the best thesis in robotics in Europe, Frank decided to co-found Bionomous in 2019. Since then, Frank has served as the CEO and Chairman of the company.

Dr. Deana Mohr (CH)

CEO, MUVON Therapeutics

Helping patients regain control with personalized muscle regeneration

MUVON Therapeutics is a clinical stage Life Science Spin-off from the University of Zurich developing a novel therapy platform for the regeneration of skeletal muscle tissue based on autologous cells. We aim to provide safe, effective and affordable treatments to millions of patients suffering from seriously debilitating diseases by not only repairing damaged tissue but also increase the regenerative potential of weakened muscles. Our initial area of focus is the treatment of stress urinary incontinence in women, supporting them respectfully throughout their journey to a healthy life.

Deana has been central to the development of the science behind MUVON from the very beginning. The results of her seminal PhD thesis (Radiopharmaceutical Sciences at ETH Zurich) were the final step before the authorization of the muscle tissue-engineering project for clinical translation which inspired her to proceed with further education in CAS Clinical Trial Management, build an international consortium, and prepare a Horizon2020 grant proposal (2016) for the translation of the project from bench-to-bedside, effectively establishing the MUS.I.C. project.

 

Deana co-founded MUVON Therapeutics in 2020 and serves as its CEO. In 2021 Deana was awarded the Venture Leaders Award by Venturelab as one of the 10 best Medtech Start Ups selected to represent the Swiss National Startup Team in Boston, USA. Deana was named one of the top 30 Rising Leaders Across The Biopharma, Medtech and Health Technology Sectors by InVivo in 2022. Additionally, she had the privilege to present her discoveries and research findings to a Nobel Prize winner and pioneer of cell therapy Prof. Yamanaka, sharing a common goal of bringing regenerative therapies to the people in need.