3D Cell Culture and Bioprocess User Days 2022 | Corning

Join the Discussion on Future Applications of 3D Cell Culture and of Bioprocess

To capture the energy and excitement in the fields of 3D Cell Culture and of Bioprocess, Corning is bringing together industry leaders and innovators to share ideas and information, and to discuss future applications.

Join us for two days to:


  • Hear from industrial and academic researchers about their research and work
  • Meet and network with peers and thought leaders in the field
  • Share experiences with other users to overcome experimental limitations and get insights into currently used protocols and procedures
  • Obtain scientific support for selecting and using the appropriate products
  • Learn about what’s new from Corning and get up to speed with new products that might enhance research

June 8


Speakers | 3D Cell Culture User Day

Hansjoerg Keller

Hansjoerg Keller, Ph.D.

Senior Principal Scientist, Musculoskeletal Disease Area, Novartis Institutes for BioMedical Research

 

Contractile 3D bioprinted human skeletal muscle models in multiwell plates for in vitro micro-physiological drug profiling

 

Human in vitro micro-physiological systems (MPS) are crucial elements in preclinical drug discovery to reduce high attrition rates of new drugs in the clinic. We developed Matrigel 3D bioprinting of contractile skeletal muscle models in 24-well plates using microvalve-based drop-on-demand printing of primary human precursor cells. Human muscle models of a few millimeters in length and a few hundred microme-ters in thickness were cultured on an agarose substrate between two inserted attachment posts. After a week, they exhibited aligned and striated myofibers, which contracted upon electrical pulse stimulation (EPS). Prolonged EPS-induced contractions stimulated IL-6 myokine expression and activated the Akt hypertrophy pathway indicating applicability of this system as an in vitro exercise model. Furthermore, we could show acute force increases within minutes by the known muscle stimulators caffeine and tro-ponin C activator Tirasemtiv using the models in an organ-bath force transducer contractility assay set-up. Finally, we built an instrument (MUSTANG) for automated electrical excitation and contractile force measurements of the muscle models in multiwells including an EPS system for 24-well plates consisting of two U-shaped Pt electrodes per well and an automated imaging system for attachment posts move-ments as a readout for contractile force of the muscle models. The new human skeletal muscle MPS has the potential to greatly benefit preclinical in vitro functional compound profiling to develop drugs for various genetic and degenerative muscle wasting diseases where there is currently a complete lack of efficacious medication.

Albano Meli

Albano Meli, Ph.D.

Senior Research Associate, CRCN INSERM Montpellier

 

Patient-specific iPSC biotechnology and neurocardiac organ-on-a-chip to model inherited familial arrhythmia

 

My current research topic focuses on modeling neurocardiac inherited disorders and on understanding the fundamental processes that trigger them. I have been using the patient-specific induced pluripotent stem cell biotechnology in order to deeply get into the patient cellular and molecular context, obtain quantitative functional results and test potential therapeutic molecules. I am particularly interested in investigating the impact of mutations on genes coding for ion channels that are associated with cardiopathies, fatal arrhythmias and neurobehavioral disorders such as epilepsy and autism. The 3D organoid and organ-on-chip technology, together with the patient-specific iPSC technology, now offers an unique opportunity to further understand how patient dysfunctional multicellular organs communicate together and how to offer personalized medicine in this context.

Wojciech Senkowski

Wojciech Senkowski, Ph.D.

Assistant Professor, Biotech Research & Innovation Centre, University of Copenhagen

 

A platform for efficient establishment, expansion and drug response profiling of high-grade serous ovarian cancer organoids

 

The broad research use of organoids from high-grade serous ovarian carcinoma (HGSC) has been hampered by low culture success rates and limited availability of fresh tumor material. At the meeting, I will present a novel approach for HGSC organoid model development, validation and profiling. We established robust cultures from viably cryopreserved surgical material, at success rates significantly higher than what has been described before for fresh surgical material. The organoids grow robustly and exponentially for long periods of time (up to a year tested), are pure tumor cell cultures and retain the genetic and phenotypic identity and heterogeneity of the originating tumor cells. As a proof-of-concept of the versatility of the organoid models developed, we apply them for drug response profiling in a 384-well microplate high-throughput format. We demonstrate that in vitro drug responses correlate with clinical responses and that physiologic Human Plasma-Like Medium (HPLM) enhances the predictivity of the results. Taken together, this work facilitates the application of HGSC organoids in basic and translational ovarian cancer research.

Francesco Boccellato

Francesco Boccellato

Principal Investigator & Leadership Fellow, Ludwig Institute for Cancer Research, University of Oxford

 

Stem-cell driven models to understand epithelial response to infections and cancer initiation

 

The forefront of the gastrointestinal mucosa consists mainly of a continuous polarised epithelial monolayer, protected by mucus. This strong defence barrier can be colonized by pathogens arousing a chronic inflammatory state. This exceptional colonization ability is associated with an increased risk of developing adenocarcinomas at the sites of infection. We have regenerated organoids and we have developed a new functional epithelial monolayer culture called “mucosoids”. The mucosoids are the human multi-lineage stem-cell-based in-vitro equivalent of a real mucosa. They mimic the function of a homeostatic epithelial barrier including accumulation of mucus on the apical side. Use of human mucosoid cultures reveals novel insight into epithelial homeostasis and response to bacterial infections.

Farzin Pourfarzad

Farzin Pourfarzad, Ph.D.

Director of Research and Development, Hubrecht Organoid Technology (HUB)

 

Hub Organoids: A Translatable Patient-Derived In Vitro Platform For Disease Modelling And Drug Development

 

Hubrecht Organoid Technology (HUB) has developed 3D culture systems to establish and expand human and animal epithelial tissue from a variety of organs both healthy and diseased, such as cancer. The organoid technology is based on the work of Hans Clevers that identified adult stem cells in many human tissues including, but not limited to, intestine, liver, pancreas, breast, and lung. Organoid cultures have long term expansion capacity, are genetically and phenotypically stable and retain biological and functional properties of the original tissue (Barker et al., Nature 2007; Sato et al., Nature 2009, 2011; Gastroenterology 2011; Huch et al., Nature 2013; Karthaus et al., Cell 2014; Cell 2015; Boj et al., Cell 2015, Sachs et al. EMBO 2019). Organoids recapitulate the original tissue response to external stimuli and therefore provide a unique and robust model for drug development, diagnostics, and patient drug response stratification for personalized medicine (van de Wetering et al., Cell 2015, Sachs et al., Cell 2018; Driehuis et al., PNAS 2019). Currently, HUB is developing co-culture assays to combine organoid technology with immune cells for immune oncology-based treatments and immunotherapeutic compounds for inflammatory and autoimmune diseases. 

HUB has built a comprehensive Living Biobank of well-characterized organoids from different healthy, disease, and cancerous tissues of multiple organs. In combination with the Living Biobank and Organoid technology, HUB has a unique platform to develop assays for preclinical drug discovery,  efficacy, and safety evaluation.

Mikael Garcia

Mikael Garcia, Ph.D.

Field Application Scientist, Corning Life Sciences

 

Moving Towards High Throughput 3D Cell Culture for Drug Screening and Precision Medicine

 

In vitro cell cultures and animal experiment models are crucial instruments in basic research and preclinical studies. Cells grown in 3D more closely mimic in vivo behavior in tissues and organs than in 2D. 3D cell culture involves expanding cells in a volumetric space as aggregates, spheroids, or organoids. This technique creates a more accurate in vitro environment and provides an alternative to in vivo models for fundamental cell biology and physiology research. The complexities of assay development and challenges associated with validating and transferring assays into high throughput modes are well known bottlenecks in the drug discovery process, especially for those using 3D cell culture. For more than 25 years, Corning has delivered innovations that have advanced the science of 3D cell culture breaking through the barriers to creating more in vivo-like environments and predictive models. In this presentation, I will discuss how Corning products can help you from starting with 3D cell culture an relevant models, to scale up and to move toward high throughput screening.

Nicolas Aznar

Nicolas Aznar, Ph.D.

Group Leader, Cancer Research Center of Lyon, CRCL Lyon

 

Counteracting organoid stemness exhaustion by controlling stem cell microenvironment for organoid culture standardization and robust disease modelling.

 

Stem cells (SCs) and their direct derived organoids offer promising perspectives in regenerative medicine as a source of healthy cells in order to replace their impaired counterparts within a given tissue. Furthermore, cancer-derived organoid culture offers promising perspectives in disease modeling and cancer stem cells (CSCs) studies known to drive tumor initiation, metastatic dissemination, drug resistance, and patients’ relapse. Indeed, high SC signature scores statistically associate with a high risk of tumor relapse in colorectal cancer patients. Therefore, the characterization of mechanisms maintaining stem cell phenotypes is crucial for understanding the SC biology in normal and cancerous tissues and for developing new therapeutic approaches to target them. Because of tremendous amount of publicly available protocols, isolating SCs doesn’t constitute a critical step anymore. Nonetheless, due to their sensitivity to microenvironmental changes, cultivating SCs in 3D to generate organoids still remains challenging and researchers face technical issues to standardize and maintain stemness phenotype to generate robust data. To overcome these obstacles, standard SC culture conditions need to be further improved to maintain their culture in long term concomitant with durable genetic and phenotypic stabilities.  In order to perform in vitro SC studies more efficiently, we have developed a unique and innovative 3D cell culture device (patent pending) improving drastically the intestinal SCs culture quality and allowing the characterization of the gene landscape and regulatory signaling pathways. These fine-tuned culture conditions led us to successfully generate long term “high quality” intestine/colon and lung mouse organoids from normal tissue with limited cell drift and enriched in SC population. Leaning on this technology, we take advantage of our “savoir faire” today to step up the organoid technology by cultivating high quality patient-derived organoids (from normal and tumoral tissues) to predict drug response and sensitivity to chemotherapy.

Phillip Wright

Phillip Wright, Ph.D.

Associate Principal Scientist, Cyprotex

 

3D Organ Models in De-Risking Safety Liabilities

 

3D cultures have advanced the field of tox screening in a multitude of ways. Not only do they represent tissues of origin more accurately compared to their 2D counterpart, they often increase the accuracy of the tox screening process. Here we present a variety of in vitro 3D cell cultures, and how they have improved the safety of compound screening.

June 9


Speakers | Bioprocess User Day

Hawaa Cumar

Hawaa Cumar

Team Lead, eXmoor Pharma Concepts

 

Switching from adherent to suspension

 

This talk will look at the process flow through when switching from an adherent to suspension platform at small scale.

Churlaud Guillaume

Guillaume Churlaud, Ph.D.

Senior Manager Quality Control, Advanced Therapy Medicinal Products, MEARY Center

 

The manufacturing resources needed to make autologous ATMPs accessible at affordable costs to the patients

 

Advanced therapy medicinal products (ATMPs) is an emerging new class of pharmaceutical drugs. These cell and gene therapy products are complicated and very expensive to manufacture, and are thus currently not easily accessible to physicians and patients. Technological solutions have been provided for large batches of allogeneic products, that seem to have now a profitable business model. This is not the case for individual batches of autologous products, whereas these drugs are a source of therapeutic hope for many patients. Technological solutions must be provided to control the production costs of these autologous therapies. Here we address the needs and possible ways to reduce costs. We also provide a feedback on the manufacturing processes developed in the MEARY center compared to the manufacturing processes that have not been developed due to lack of technical solutions or too expensive costs.

Maria Begoña Castro

Maria Begoña Castro

Founding Partner & Scientific Director, Histocells Regenerative Medicine

 

Histocell Regenerative Medicine: adapting ATMPs to Clinical Phase III

 

Histocell is a company that emerged in 2004 from a cell biology research group at the University of the Basque Country in northern Spain, focused on the development of new products in the area of ​​regenerative and restorative medicine. Throughout these years, Histocell has developed its own ATMP and MD products, focused on tissue repair and we have faced the translation of the technology to the GMP field, its industrial scaling and its arrival to the clinic. At this time, apart from the clinical progress of our own products and the evolution in our own technologies, we are immersed in the start-up of our new industrial plant for ATMPs and the adaptation of the processes to adjust to the requirements of clinical phases IIb and III. In this sense, we have had the opportunity to collaborate with Corning, in the adaptation of their X-Wash system, for the adaptation to a ready-to-use protocol of our ATMP developed for the acute phase of spinal cord injuries of traumatic origin, we will share our experience.

Marie Berger

Marie Berger

Application Engineer, Myriade

 

Videodrop: new tool for viral vector bioproduction follow-up

 

Today, Gene and Cell therapies are booming. However, due to the lack of solutions allowing rapid & continuous monitoring throughout the bioproduction process, Biotech’ companies are working “almost blind”. They often have to wait several hours or even days to get some results (ex:P24 Elisa tests). Therefore, there is a major need for new "in-process" quality control tools. 

 

Myriade developed VIDEODROP: an innovative nanoscale imaging technology. VIDEODROP makes it possible to measure the size & concentration of nanoparticles in real time (40s), in a single drop (5µL) without labeling & no purification in the range of 80 nm - 500nm and visualize aggregates & debris up to 10 microns.  

 

During the bioproduction process, VIDEODROP is used for the quantification of the physical titer. Thanks to its rapidity of measurement, it allows to continuously monitor processes, directly perform yield calculation after Harvesting, Clarification, Purification, Concentration, or Diafiltration, and thus enhance process optimization and formulation screening being able to work directly on real samples in a non-denaturant way at any stage of the bioproduction 

 

Thereby, Videodrop is a promising tool for fast in-process controls, continuous monitoring, and final product characterization of complex viral vector solutions of Lentiviruses, adenoviruses, retrovirus & extracellular vesicles. 

Ana Fernandes-Platzgummer

Ana Fernandes-Platzgummer, Ph.D.

Research Scientist, Institute for Bioengineering and Biosciences (iBB)

 

Design and operation of a fully controlled manufacturing platform for mesenchymal stromal cells and their derived extracellular vesicles

 

The therapeutic effects of human mesenchymal stromal cells (MSC) have been attributed mostly to their paracrine activity exerted through small-secreted extracellular vesicles (EVs). It is expected that the combined and/or alternate use of MSC and derived EVs will strengthen the use of MSC-based therapies and will rapidly progress towards clinical studies.  Currently, the production of MSC and their derived EVs (MSC-EVs) is performed in laborious/time-consuming static culture systems with limited manufacturing capacity using serum-containing media. Additionally, several differences were observed, in terms of the cargo of EVs (proteins and nucleic acids), between EVs isolated from culture supernatants of MSC expanded under different culture conditions, stressing the importance of controlling all culture process parameters to obtain consistent EVs content. In this context, it is crucial to design and develop a scalable, robust and in-depth manufacturing process for human MSC and their derived EVs. In this work, we will describe a fully controlled system for MSC/EVs production, using defined reagents compliant with GMP standards, and the Integration of a novel scalable purification process for the EV isolation and purification comprising unit operations that are robust, selective and cost-effective. The manufacturing platform to be established herein is expected to pave a new way for the development of stem cell-based engineering, eliminating time- and labour-consuming procedures aiming at the scalable production of EVs secreted from well-defined MSC populations in order to boost their medical uses.

Yoshi Shyu

Yoshi Shyu, Ph.D.

Global Director of Applications, Corning Life Sciences

 

Leveraging Novel Adherent Technology for Intensified Cell and Gene Therapy Manufacturing

 

Cell and gene therapies (CGTs) are breakthrough medical innovations that are transforming how we treat and potentially cure certain diseases. With more than 2,600 CGT clinical trials underway, the manufacturing processes for both gene delivery vehicles (such as viral vectors) and therapeutic cells (such as stem cells) continue to be bottlenecks for the pharmaceutical industry. There is an urgent need for automated, scalable, and cost-efficient manufacturing platforms to meet the rapidly increasing clinical demands.

 

The Corning Ascent FBR System is designed to combine the greater yield efficiency, flexibility, and viable cell harvest capability of adherent bioproduction platforms with the scale, automation, and closed system features of suspension manufacturing systems. Key features of the Ascent FBR System include specially treated and packed polymer mesh that can enable uniform low-shear fluid flow through the bioreactor bed, uniform high-density cell growth, >90% transfection efficiency, and high vector genomes yield/cm2. The ability to harvest viable cells from the Ascent FBR System enables its use in seed train for manufacturing scale as well as for other CGT workflows such as lentivirus, MSC and PSC, in addition to viral vaccine and other biologic production applications.

In conclusion, the Ascent FBR system is purposely designed to help address manufacturing bottlenecks by providing an automated, high-yield, scalable adherent bioreactor platform for CGT workflows.