TY - JOUR A1 - Moll, Corinna A1 - Reboredo, Jenny A1 - Schwarz, Thomas A1 - Appelt, Antje A1 - Schürlein, Sebastian A1 - Walles, Heike A1 - Nietzer, Sarah T1 - Tissue Engineering of a Human 3D in vitro Tumor Test System JF - Journal of Visualized Experiments N2 - Cancer is one of the leading causes of death worldwide. Current therapeutic strategies are predominantly developed in 2D culture systems, which inadequately reflect physiological conditions in vivo. Biological 3D matrices provide cells an environment in which cells can self-organize, allowing the study of tissue organization and cell differentiation. Such scaffolds can be seeded with a mixture of different cell types to study direct 3D cell-cell-interactions. To mimic the 3D complexity of cancer tumors, our group has developed a 3D in vitro tumor test system. Our 3D tissue test system models the in vivo situation of malignant peripheral nerve sheath tumors (MPNSTs), which we established with our decellularized porcine jejunal segment derived biological vascularized scaffold (BioVaSc). In our model, we reseeded a modified BioVaSc matrix with primary fibroblasts, microvascular endothelial cells (mvECs) and the S462 tumor cell line For static culture, the vascular structure of the BioVaSc is removed and the remaining scaffold is cut open on one side (Small Intestinal Submucosa SIS-Muc). The resulting matrix is then fixed between two metal rings (cell crowns). Another option is to culture the cell-seeded SIS-Muc in a flow bioreactor system that exposes the cells to shear stress. Here, the bioreactor is connected to a peristaltic pump in a self-constructed incubator. A computer regulates the arterial oxygen and nutrient supply via parameters such as blood pressure, temperature, and flow rate. This setup allows for a dynamic culture with either pressure-regulated pulsatile or constant flow. In this study, we could successfully establish both a static and dynamic 3D culture system for MPNSTs. The ability to model cancer tumors in a more natural 3D environment will enable the discovery, testing, and validation of future pharmaceuticals in a human-like model. KW - bioengineering KW - biomedical engineering KW - tissue engineering KW - biotechnology KW - cultured KW - tumor cells KW - cell culture KW - 3D in vitro models KW - bioreactor KW - dynamic culture conditions KW - tumor test system KW - primary cell isolation KW - BioVaSc KW - decellularization KW - equipment and supplies KW - cellular microenvironment KW - culture techniques KW - cell engineering KW - anatomy KW - physiology KW - molecular biology KW - cellular biology Y1 - 2013 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-132277 UR - http://www.jove.com/video/50460 VL - 78 IS - e50460 ER - TY - JOUR A1 - Haeusner, Sebastian A1 - Herbst, Laura A1 - Bittorf, Patrick A1 - Schwarz, Thomas A1 - Henze, Chris A1 - Mauermann, Marc A1 - Ochs, Jelena A1 - Schmitt, Robert A1 - Blache, Ulrich A1 - Wixmerten, Anke A1 - Miot, Sylvie A1 - Martin, Ivan A1 - Pullig, Oliver T1 - From Single Batch to Mass Production–Automated Platform Design Concept for a Phase II Clinical Trial Tissue Engineered Cartilage Product JF - Frontiers in Medicine N2 - Advanced Therapy Medicinal Products (ATMP) provide promising treatment options particularly for unmet clinical needs, such as progressive and chronic diseases where currently no satisfying treatment exists. Especially from the ATMP subclass of Tissue Engineered Products (TEPs), only a few have yet been translated from an academic setting to clinic and beyond. A reason for low numbers of TEPs in current clinical trials and one main key hurdle for TEPs is the cost and labor-intensive manufacturing process. Manual production steps require experienced personnel, are challenging to standardize and to scale up. Automated manufacturing has the potential to overcome these challenges, toward an increasing cost-effectiveness. One major obstacle for automation is the control and risk prevention of cross contaminations, especially when handling parallel production lines of different patient material. These critical steps necessitate validated effective and efficient cleaning procedures in an automated system. In this perspective, possible technologies, concepts and solutions to existing ATMP manufacturing hurdles are discussed on the example of a late clinical phase II trial TEP. In compliance to Good Manufacturing Practice (GMP) guidelines, we propose a dual arm robot based isolator approach. Our novel concept enables complete process automation for adherent cell culture, and the translation of all manual process steps with standard laboratory equipment. Moreover, we discuss novel solutions for automated cleaning, without the need for human intervention. Consequently, our automation concept offers the unique chance to scale up production while becoming more cost-effective, which will ultimately increase TEP availability to a broader number of patients. KW - ATMP KW - tissue engineering KW - GMP KW - manufacturing KW - autologous KW - cartilage regeneration KW - automation & robotics KW - automation Y1 - 2021 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-244631 SN - 2296-858X VL - 8 ER -