Skip to main content

Implementation and Use of State-of-the-Art, Cell-Based In Vitro Assays

  • Chapter

Part of the book series: Handbook of Experimental Pharmacology ((HEP,volume 232))

Abstract

The impressive advances in the generation and interpretation of functional omics data have greatly contributed to a better understanding of the (patho-)physiology of many biological systems and led to a massive increase in the number of specific targets and phenotypes to investigate in both basic and applied research. The obvious complexity revealed by these studies represents a major challenge to the research community and asks for improved target characterisation strategies with the help of reliable, high-quality assays. Thus, the use of living cells has become an integral part of many research activities because the cellular context more closely represents target-specific interrelations and activity patterns. Although still predominant, the use of traditional two-dimensional (2D) monolayer cell culture models has been gradually complemented by studies based on three-dimensional (3D) spheroid (Sutherland 1988) and other 3D tissue culture systems (Santos et al. 2012; Matsusaki et al. 2014) in an attempt to employ model systems more closely representing the microenvironment of cells in the body. Hence, quite a variety of state-of-the-art cell culture models are available for the generation of novel chemical probes or the identification of starting points for drug development in translational research and pharma drug discovery. In order to cope with these information-rich formats and their increasing technical complexity, cell-based assay development has become a scientific research topic in its own right and is used to ensure the provision of significant, reliable and high-quality data outlasting any discussions related to the current “irreproducibility epidemic” (Dolgin 2014; Prinz et al. 2011; Schatz 2014). At the same time the use of cells in microplate assay formats has become state of the art and greatly facilitates rigorous cell-based assay development by providing the researcher with the opportunity to address the multitude of factors affecting the actual assay results in a systematic fashion and a timely manner. This microplate-based assay development strategy should result in the setting up of more robust and reliable test systems that ensure and increase the confidence in the statistical significance of the actual data generated. And, although assay miniaturisation is essential in order to achieve this, most, if not all, cell-based assays can be easily reformatted and adapted to be used in this format in a straightforward manner. This synopsis aims at summarising valuable, general observations made when implementing a diverse set of functional cellular in vitro assays at Bayer Pharma AG without claiming to deeply review all of the literature available in each and every detail. In addition, phenotypic assays (Moffat et al. 2014) or label-free detection methods (Minor 2008) are not discussed. Although this essay tries to cover the most relevant technological developments in the field, it nevertheless may express personal preferences and peculiarities of the author’s approach to state-of-the-art cell-based assay development. For additional reviews covering the actual field, see Wunder et al. (2008) and Michelini et al. (2010).

This is a preview of subscription content, log in via an institution.

Buying options

Chapter
USD   29.95
Price excludes VAT (USA)
  • Available as PDF
  • Read on any device
  • Instant download
  • Own it forever
eBook
USD   169.00
Price excludes VAT (USA)
  • Available as EPUB and PDF
  • Read on any device
  • Instant download
  • Own it forever
Softcover Book
USD   219.99
Price excludes VAT (USA)
  • Compact, lightweight edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info
Hardcover Book
USD   219.99
Price excludes VAT (USA)
  • Durable hardcover edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info

Tax calculation will be finalised at checkout

Purchases are for personal use only

Learn about institutional subscriptions

Notes

  1. 1.

    That is, the 96-well plate described by Gyula Takátsy in 1951 already

  2. 2.

    Aside from the growing public debate over the disproportionate use of experimental animals in certain research areas

  3. 3.

    See: Foundation of the Society for Biomolecular Sciences (SBS) in 1994 (http://www.slas.org/about/who-we-are/society-for-biomolecular-sciences/) now an integral part of the Society for Laboratory Automation and Screening (SLAS) established in 2010 (https://www.slas.org/).

  4. 4.

    As exemplified by the foundation of (a) the NIH Chemical Genomic Center (NCGC) in 2004, now a part of the National Center For Advancing Translational Science (NCATS; established 2012; http://www.ncats.nih.gov/research/reengineering/ncgc/ncgc.html) or (b) the Innovative Medicines Initiative (IMI; http://www.imi.europa.eu/content/home) established in 2008 as well as (c) the establishment of the National Cancer Institute Chemical Genomics Consortium in 2009 (http://dctd.cancer.gov/CurrentResearch/cbc/20090810_meeting.htm)

  5. 5.

    See Freshney (2000b).

  6. 6.

    The term frozen cell describes the use of freshly resuscitated cells shortly after their recovery from liquid nitrogen storage and should not to be mistaken for the use of division-arrested cells, as exemplified/described by Digan et al. (2005).

  7. 7.

    Demanding for the supply of 1–5 × 108 cells per screening day

  8. 8.

    From routine pharmacological profiling on a weekly basis to approaches based on frozen cells transiently transfected with mutant receptors to test hypotheses from computational chemistry

  9. 9.

    That is, a medium version based on charcoal-treated serum, medium only or even just PBS

  10. 10.

    For chloramphenicol acetyltransferase (CAT), see Devinoy et al. (1991).

  11. 11.

    For secreted alkaline phosphatase (SEAP), see: Cullen and Malim (1992).

  12. 12.

    For secreted urokinase, see Langer et al. (1995).

  13. 13.

    For a discussion of FRET artefacts and signal proper, see (Vogel et al. 2006).

  14. 14.

    For a thorough examination of potential BRET-related artefacts, see Marullo and Bouvier (2007).

  15. 15.

    That is, from milliseconds to seconds and from a few minutes to a few hours

  16. 16.

    A process called complementation that has been reported in 1965 already, see Ullmann et al. (1965).

  17. 17.

    See http://www.discoverx.com/technologies-platforms/enzyme-fragment-complementation-technology/pathhunter-efc-cell-based-assay-platform.

  18. 18.

    For more details, see http://www.discoverx.com/technologies-platforms/enzyme-fragment-complementation-technology/hithunter-efc-biochemical-assay-platform.

  19. 19.

    For dissociation-enhanced lanthanide fluorescent immunoassays (Delfia) marketed by PerkinElmer, see http://www.perkinelmer.com/catalog/category/id/delfia%20trf%20assays%20and%20reagents.

  20. 20.

    The use of lanthanide cryptates is called HTRF (homogeneous, time-resolved FRET) and marketed by the French company Cisbio http://www.cisbio.com/other.

  21. 21.

    For more details, see http://www.cisbio.com/other/drug-discovery as well as http://www.perkinelmer.com.

  22. 22.

    See http://www.cisbio.com/drug-discovery/toolbox-products-and-services.

  23. 23.

    http://www.perkinelmer.com/pages/020/cellularimaging/products/opera.xhtml

  24. 24.

    http://www.gelifesciences.com/webapp/wcs/stores/servlet/catalog/en/GELifeSciences-us/applications/hca-imaging-systems-and-software

  25. 25.

    http://www.lifetechnologies.com/order/catalog/product/ASN00003L

  26. 26.

    http://www.perkinelmer.com/pages/020/cellularimaging/products/default.xhtml

  27. 27.

    http://www.moleculardevices.com/systems/high-content-imaging/imagexpress-micro-xls-widefield-high-content-analysis-system

  28. 28.

    http://ttplabtech.com/phenotypic-screening/acumen/

  29. 29.

    http://www.biotek.com/products/imaging/cytation5_cell_imaging_multi_mode_reader.html

  30. 30.

    That is, average signal intensity, which can be the product of multiple sub-parameters of course

  31. 31.

    In a sense permitting population studies otherwise known from fluorescence-activated cell sorter (FACS) analysis

  32. 32.

    Molecular Probes® Handbook – A Guide to Fluorescent Probes and Labeling Technologies; the printed version is available free of charge via http://www.lifetechnologies.com/de/de/home/references/molecular-probes-the-html.

References

  • Bergsdorf C, Kropp-Goerkis C, Kaehler I, Ketscher L, Boemer U, Parczyk K, Bader B (2008) A one-day, dispense-only IP-One HTRF for high-throughput screening of Galpha q protein coupled receptors: towards cells as reagents. Assay Drug Dev Technol 6:39–53

    Article  CAS  PubMed  Google Scholar 

  • Breslin S, O’Driscoll L (2013) Three-dimensional cell culture: the missing link in drug discovery. Drug Discov Today 18:240–249

    Article  CAS  PubMed  Google Scholar 

  • Burgstaller G, Oehrle B, Koch I, Lindner M, Eickelberg O (2013) Multiplex profiling of cellular invasion in 3D cell culture models. PLoS One 8:e63121

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Cawkill D, Eaglestone SS (2007) Evolution of cell-based reagent provision. Drug Discov Today 12:820–825

    Article  CAS  PubMed  Google Scholar 

  • Cottet M, Falco A, Villier B, Laget M, Zwier JM, Trinquet E, Mouillac B, Pin JP, Durroux T (2015) Multicolor time-resolved Forster resonance energy transfer microscopy reveals the impact of GPCR oligomerization on internalization processes. FASEB J 29:2235–2246

    Article  PubMed  Google Scholar 

  • Cullen BR, Malim MH (1992) Secreted placental alkaline phosphatase as a eukaryotic reporter gene. Methods Enzymol 216:362–368

    Article  CAS  PubMed  Google Scholar 

  • De Wet JR, Wood KV, Helinski DR, DeLuca M (1985) Cloning of firefly luciferase cDNA and the expression of active luciferase in Escherichia coli. Proc Natl Acad Sci U S A 82:7870–7873

    Article  PubMed  PubMed Central  Google Scholar 

  • Denner P, Schmalowsky J, Prechtl S (2008) High-content analysis in preclinical drug discovery. Comb Chem High Throughput Screen 11:216–230

    Article  CAS  PubMed  Google Scholar 

  • Devinoy E, Malienou-N’Gassa R, Thepot D, Puissant D, Houdebine LM (1991) Hormone responsive elements within the upstream sequences of the rabbit whey acidic protein (WAP) gene direct chloramphenicol acetyl transferase (CAT) reporter gene expression in transfected rabbit mammary cells. Mol Cell Endocrinol 81:185–193

    Article  CAS  PubMed  Google Scholar 

  • Digan ME, Pou C, Niu H, Zhang JH (2005) Evaluation of division arrested cells for cell-based high-throughput screening. J Biomol Screen 10:615–623

    Article  CAS  PubMed  Google Scholar 

  • Dolgin E (2014) Drug discoverers chart path to tackling data irreproducibility. Nat Rev Drug Discov 13:875–876

    Article  CAS  PubMed  Google Scholar 

  • Eglen RM (2002) Enzyme fragment complementation: a flexible high throughput screening assay technology. Assay Drug Dev Technol 1:97–104

    Article  CAS  PubMed  Google Scholar 

  • Fennema E, Rivron N, Rouwkema J, van Blitterswijk C, de Boer J (2013) Spheroid culture as a tool for creating 3D complex tissues. Trends Biotechnol 31:108–115

    Article  CAS  PubMed  Google Scholar 

  • Freshney RI (2000a) Introduction. Historical background. In: Freshney RI Culture of animal cells: a manual of basic technique and specialized applications, 4th edn. Wiley-Liss, New York

    Google Scholar 

  • Freshney RI (2000b) Cell lines, routine maintenance, split ratios and growth cycle. In: Freshney RI Culture of animal cells: a manual of basic technique and specialized applications, 4th edn. Wiley-Liss, New York, p 188

    Google Scholar 

  • Friedrich J, Seidel C, Ebner R, Kunz-Schughart LA (2009) Spheroid-based drug screen: considerations and practical approach. Nat Protoc 4:309–324

    Article  CAS  PubMed  Google Scholar 

  • Hill SJ, Baker JG, Rees S (2001) Reporter-gene systems for the study of G-protein-coupled receptors. Curr Opin Pharmacol 1:526–532

    Article  CAS  PubMed  Google Scholar 

  • Hirschhaeuser F, Menne H, Dittfeld C, West J, Mueller-Kiliser W, Kunz-Schughart LA (2010) Multicellular tumor spheroids: an underestimated tool is catching up again. J Biotechnol 148:3–15

    Article  CAS  PubMed  Google Scholar 

  • Hoffman AF, Garippa RJ (2007) A pharmaceutical company user’s perspective on the potential of high content screening in drug discovery. Methods Mol Biol 356:19–31

    Article  CAS  PubMed  Google Scholar 

  • Ioannidis JPA (2005) Why most published research findings are false. PLoS Med 2:e124

    Article  PubMed  PubMed Central  Google Scholar 

  • Kenny HA, Lal-Nag M, White EA, Shen M, Chiang C, Mitra AK, Zhang Y, Curtis M, Shryver EM, Bettis S, Jadhav A, Boxer MB, Li Z, Ferrer M, Lengyel E (2015) Quantitative high throughput screening using a primary three-dimensional organotypic culture predicts in vivo efficacy. Nat Commun 6:6220. doi:10.1038/ncomms7220

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Langer G, Toschi L, Dieckmann J, Schleuning WD (1995) Low-M(r) urokinase-type plasminogen activator as a reporter protein. Gene 161:287–292

    Article  CAS  PubMed  Google Scholar 

  • Larsson LI (1988a) Immunocytochemical detection systems. In: Larsson LI (ed) Immunocytochemistry: theory and practice. CRC, Boca Raton, pp 77–146

    Google Scholar 

  • Larsson LI (1988b) Fixation and tissue pretreatment. In: Larsson LI (ed) Immunocytochemistry: theory and practice. CRC, Boca Raton, pp 41–76

    Google Scholar 

  • Luense S, Denner P, Fernandez-Montalvan A, Hartung I, Husemann M, Stresemann C, Prechtl S (2015) Quantification of histone H3 Lys27 trimethylation (H3K27me3) by high-throughput microscopy enables cellular large-scale screening for small-molecule EZH2 inhibitors. J Biomol Screen 20:190–201

    Article  PubMed  PubMed Central  Google Scholar 

  • Marullo S, Bouvier M (2007) Resonance energy transfer approaches in molecular pharmacology and beyond. Trends Pharmacol Sci 28:362–365

    Article  CAS  PubMed  Google Scholar 

  • Matsusaki M, Case CP, Akashi M (2014) Three-dimensional cell culture technique and pathophysiology. Adv Drug Deliv Rev 74:95–103

    Article  CAS  PubMed  Google Scholar 

  • Michelini E, Cevenini L, Mezzanotte L, Coppa A, Roda A (2010) Cell-based assays: fuelling drug discovery. Anal Bioanal Chem 398:227–238

    Article  CAS  PubMed  Google Scholar 

  • Minor LK (2008) Label-free cell-based functional assays. Comb Chem High Throughput Screen 11:573–580

    Article  CAS  PubMed  Google Scholar 

  • Moffat JG, Rudolph J, Bailey D (2014) Phenotypic screening in cancer drug discovery – past, present, future. Nat Rev Drug Discov 13:588–602

    Article  CAS  PubMed  Google Scholar 

  • Moore JT, Davis ST, Dev IK (1997) The development of β-lactamase as a highly versatile genetic reporter for eukaryotic cells. Anal Biochem 247:203–209

    Article  CAS  PubMed  Google Scholar 

  • Parsons SJ, Rhodes SA, Connor HE, Rees S, Brown J, Giles H (2000) Use of a dual firefly and Renilla luciferase reporter gene assay to simultaneously determine drug selectivity at human corticotrophin releasing hormone 1 and 2 receptors. Anal Biochem 281:187–192

    Article  CAS  PubMed  Google Scholar 

  • Prinz F, Schlange T, Assadulah K (2011) Believe it or not: how much can we rely on published data on potential drug targets? Nat Rev Drug Discov 10:712–713

    Article  CAS  PubMed  Google Scholar 

  • Remington SJ (2011) Green fluorescent protein: a perspective. Protein Sci 9:1509–1519

    Article  Google Scholar 

  • Santos E, Hernández RM, Pedraz JL, Orive G (2012) Novel advances in the design of three-dimensional bio-scaffolds to control cell fate: translation from 2D to 3D. Trends Biotechnol 30:331–340

    Article  CAS  PubMed  Google Scholar 

  • Schatz G (2014) The faces of big science. Nat Rev Mol Cel Biol 15:423–426

    Article  CAS  Google Scholar 

  • Schiele F, Ayaz P, Fernandez-Montalvan A (2014) A universal homogeneous assay for high-throughput determination of binding kinetics. Anal Biochem 468C:42–49

    PubMed  Google Scholar 

  • Schulze J, Moosmayer D, Weiske J, Fernandez-Montalvan A, Herbst C, Jung M, Haendler B, Bader B (2015) Cell-based protein stabilization assays for the detection of interactions between small-molecule inhibitors and BRD4. J Biomol Screen 20:180–189

    Article  PubMed  Google Scholar 

  • Snapp EL (2008) Fluorescent proteins: a cell biologist’s user guide. Trends Cell Biol 19:649–655

    Article  Google Scholar 

  • Stacey GN, Masters JR (2008) Cryopreservation and banking of mammalian cell lines. Nat Protoc 3:1981–1989

    Article  CAS  PubMed  Google Scholar 

  • Sutherland RM (1988) Cell and environment interactions in tumor microregions: the multicell spheroid model. Science 240:177–184

    Article  CAS  PubMed  Google Scholar 

  • Thoma CR, Zimmermann M, Agarkova I, Kelm JM, Krek W (2014) 3D cell culture systems modeling tumor growth determinants in cancer target discovery. Adv Drug Deliv Rev 69–70:29–41

    Article  PubMed  Google Scholar 

  • Ullmann A, Perrin D, Jacob F, Monod J (1965) Identification par Complémentation in vitro et Purification d’un Segment Peptidique de la ß-Galactosidase d’Escherichia coli. J Mol Biol 12:918–923

    Article  CAS  PubMed  Google Scholar 

  • Vogel SS, Thaler C, Koushik SV, Fanciful FRET (2006) Science’s stke. www.stke.org/cgi/content/sigtrans;2006/331/re2

  • Wenzel C, Riefke B, Gründemann S, Krebs A, Christian S, Prinz F, Osterland M, Golfier S, Räse S, Ansari N, Esner M, Bickle M, Pampaloni F, Mattheyer C, Stelzer EH, Parczyk K, Prechtl S, Steigemann P (2014) 3D high-content screening for the identification of compounds that target cells in dormant tumor spheroid regions. Exp Cell Res 323:131–143

    Article  CAS  PubMed  Google Scholar 

  • Wunder F, Kalthof B, Müller T, Hüser J (2008) Functional cell-based assays in microliter volumes for ultra-high throughput screening. Comb Chem High Throughput Screen 11:495–504

    Article  CAS  PubMed  Google Scholar 

  • Yamada KM, Cukierman E (2007) Modelling tissue morphogenesis and cancer in 3D. Cell 130:601–610

    Article  CAS  PubMed  Google Scholar 

  • Zaman GJR, de Roos JADM, Blomenröhr M, van Koppen CJ, Oosterom J (2007) Cryopreserved cell facilitate cell-based drug discovery. Drug Discov Today 12:522–526

    Article  Google Scholar 

  • Zhang JH, Chung TDY, Oldenburg KR (1999) A simple statistical parameter for use in evaluation and validation of high throughput screening assays. J Biomol Screen 4:67–73

    Article  PubMed  Google Scholar 

  • Zwier JM, Roux T, Cottet M, Durroux T, Douzon S, Bdioui S, Gregor N, Bourrier E, Oueslati N, Nicolas L, Tinel N, Boisseau C, Yverneau P, Charrier-Savournin F, Fink M, Trinquet E (2010) A fluorescent ligand-binding alternative using Tag-lite(R) technology. J Biomol Screen 15:1248–1259

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

The author thanks B. Bader, U. Nguyen, K. Parczyk, P. Steigemann and S. Prechtl for critical reading and helpful comments while preparing the manuscript.

Competing Financial Interests The author is an employee of Bayer Pharma AG.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Gernot Langer .

Editor information

Editors and Affiliations

Rights and permissions

Reprints and permissions

Copyright information

© 2015 Springer International Publishing Switzerland

About this chapter

Cite this chapter

Langer, G. (2015). Implementation and Use of State-of-the-Art, Cell-Based In Vitro Assays. In: Nielsch, U., Fuhrmann, U., Jaroch, S. (eds) New Approaches to Drug Discovery. Handbook of Experimental Pharmacology, vol 232. Springer, Cham. https://doi.org/10.1007/164_2015_18

Download citation

Publish with us

Policies and ethics