2022 Virtual 3D Cell Culture Summit

3D cell culture is exploding. To capture the energy and excitement of this moment in research, Corning is bringing together industry leaders and innovators for a global virtual 3D Cell Culture Summit to share ideas and information, and to discuss the future of 3D applications, including spheroid, organoid and tissue models.

Objectives

  • Gather leading researchers from across the globe to discuss 3D applications as well as novel and advanced models.
  • Introduce new users to best practices and tips to get started in 3D cell culture.
  • Foster discussion among industry leaders on 3D workflow optimization, trends, and innovation.

Agenda

Speakers and Panelists

Amanda Linkous

Amanda Linkous, Ph.D.

Scientific Center Manager, NCI Center for Cancer Systems Biology of Small Cell Lung Cancer

 

Organoids as a Model of Small Cell Lung Cancer Brain Metastasis

 

Small cell lung cancer (SCLC) is a highly aggressive, neuroendocrine tumor that accounts for approximately 15% of all lung cancer cases. Known for its ability to disseminate early, with a strong propensity to metastasize to the brain, SCLC is a recalcitrant cancer with very poor survival (5-year survival is less than 5%). Using a systems biology approach, we have developed 3D organoid models to capture the cell-cell interactions that underlie SCLC brain metastases.

Amy Kauffman

Amy Kauffman, Ph.D.

Sr. Development Engineer, Corning Life Sciences

 

CLS Practicum: Corning® Elplasia® Flask

 

3D cell culture models have been shown to better mimic the in vivo environment compared to traditional 2D monolayer methods. Spheroids, which are 3D round aggregates of cells, are becoming critical physiological models for cancer research and advanced therapy development for compound screening and basic science investigations. While there are various techniques for producing high quality spheroids, scaling up to obtain large quantities can be inefficient and result in spheroids of variable size and shape. To solve this challenge, the Corning Elplasia Flask is a new high-density static cell culture platform that can support the generation of ~12,000 spheroids in the footprint similar to a T-75 flask. Our data shows consistency in both diameter and circularity of spheroids produced in the Elplasia Flask in comparison to the variability observed with alternative methods. This platform is compatible with many cell types, typical to T-Flask culture, and can support research and development of life-saving treatments. Furthermore, this platform offers a unique opportunity for biomolecule production, such as extracellular vesicles, from 3D cell aggregates that share a common media reservoir. The Corning Elplasia Flask supports 3D cell culture model breakthroughs that 2D cell culture simply cannot.

Bram Herpers

Bram Herpers, Ph.D.

Senior Director, Crown Bioscience, Netherlands B.V.

 

Image-based organoid screening identifies a therapeutic EGFR x LGR5 bispecific antibody

 

Patient-derived organoids (PDOs) are established by stimulating the maintenance and proliferation of (cancer) stem cells with WNT pathway activators and receptor tyrosine kinase (RTK) agonists in an extracellular matrix environment. Here we describe a large-scale image-based screen with dual-targeting (RTKxWNT) bispecific antibodies in a diverse panel or (metastatic) colorectal cancer organoids and (matched) normal organoid models. Out of more than 500 antibodies a potent EGFRxLGR5 bispecific antibody, MCLA-158, was identified. The antibody inhibited the outgrowth of a large number of different CRC PDO models, including KRAS mutant models, but shows minimal toxicity towards healthy colon organoids. Functionally, the antibody blocks RTK signaling by triggering EGFR degradation in an LGR5-dependent manner and was shown to block metastasis initiation and to be able to suppress the outgrowth of in vivo tumor models of several other cancer types. This study highlights Crown Bioscience’s application of organoids throughout the drug discovery process –from high throughput screening to patient stratification and mechanism of action studies.

Elizabeth Abraham

Elizabeth Abraham, Ph.D.

Market Manager, 3D Cell Culture, Corning Life Sciences

 

Dr. Elizabeth Abraham is currently the Market Manager for 3D Cell Culture and has been employed at Corning Life Sciences (CLS) since 2008. During this time, she has held multiple senior roles in various functions including R&D, Project Management and Business Operations. She led the development of various new products for 2D and 3D cell culture including organoid platforms as well as cell therapy, translating voice of customer into new concepts and commercializing them globally. She has authored 23 journal articles, technical notes and is an inventor on 11 patents.

Francesco Boccellato

Francesco Boccellato, Ph.D.

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

 

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 bymucus. This strong defense 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 regeneratedorganoids and we have developed a new functional epithelial monolayer culture called “mucosoids”. The mucosoidsare 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.

Hansjoerg Keller

Hansjoerg Keller, Ph.D.

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

 

3D Bioprinted Human Skeletal Muscle Models 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 micrometers 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 troponin 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 movements 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.

Hilary Sherman

Hilary Sherman

Senior Scientist, Corning Life Sciences

 

CLS Practicum: Corning® Matribot® Bioprinter Printed Domes for Organoid Drug Testing

 

This session will demonstrate the use of the Corning Matribot Bioprinter to accurately and consistently dispense pancreatic cancer organoids for drug testing. Dispensed organoid cultures were assessed for toxicity to several chemotherapeutics that are traditionally used for treating pancreatic cancer. This data demonstrates an automated way to screen for the best drug choice for an individual’s pancreatic cancer.

Sylvia Boj

Sylvia Boj, Ph.D.

Chief Scientific Officer, HUB Organoids

 

Patient-derived Organoids: A Platform to Improve Drug Discovery and Development

 

Since the original discovery of LGR5+ adult stem cells, the technology that allowed the development of the first “mini-gut in a dish” has further been developed, optimized, and standardized for a range of organs and disease types to allow the development of living biobanks of patient-derived organoids, or HUB Organoids®. These mini organs grown ex vivo are stable in long-term culture and can be expanded and cryopreserved for multiple applications. Thanks to accurately predicting patient response, HUB Organoids have already been used to test drug efficacy and toxicity and stratify patient populations based on in vitro sensitivity data. HUB Organoids faithfully recapitulate original patient disease allowing to reduce animal usage, accelerate drug development timelines, and optimize patient selections in clinical trials, thus contributing to reducing the high attrition rate of new drugs and making personalized medicine a reality. In this presentation Dr. Sylvia Boj will present key case studies on the applications of HUB Organoids to drug discovery and development in a variety of disease indications.

Wojciech Senkowski

Wojciech Senkowski, Ph.D.

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

 

A Platform for Establishment, Expansion and 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.