Exploring the future of cell therapy research

Fast-Tracking Cell Therapy Research

Cell-based assays play a central role in the development of safe and effective cell therapies. They serve as methods for quantifying the potency of cell therapies, providing readouts for a broad spectrum of cell therapy characteristics, and are essential throughout the process from initial screening to safety and efficacy testing and dosage optimization1,2. Researchers employ a wide range of advanced cell-based assays in determining cell therapy characteristics, including growth assays, killing assays, metabolic profiling, xenograft models, and molecular profiling3–7. However, despite their critical importance, the application of these assays is fraught with challenges that hinder productivity and impede the progress of cell therapy development, highlighting the need for innovative solutions, like Levitation Technology, to streamline the pathway to therapeutic breakthroughs.

Common challenges in cell-based assays: the snowball effect

Poor-quality starting material is a primary cause of failure in cell-based assays, characterized by samples containing cells of suboptimal viability, dead or dying cells, and toxic debris. Utilizing low-quality input material undermines research reproducibility and data validity and can trigger a cascade of adverse effects impacting the assay and associated project8.

To compensate for the reduced viability and quality of the input material, more cells must be loaded into each well, which becomes particularly problematic when dealing with primary samples or difficult-to-culture cells. Additionally, more biological replicates are required to offset failed attempts and subpar outcomes. Each additional replicate incurs further expense, stretches resources, and demands more hands-on time, leading to lower testing throughput and extended development timelines. Even when experiments are completed under these conditions, the results are often highly variable, and data analysis becomes more complex, further affecting the project’s efficiency and deadlines9.

Ultimately, starting with low-quality material expands the scope and cost of projects and diminishes confidence in the outcomes, presenting a significant hurdle to the success of cell therapy projects. Consequently, scientists urgently need effective strategies to overcome these challenges and increase research productivity and outcomes.

Improved sample preparation increases productivity

Overcoming these significant challenges and enhancing the productivity of cell therapy workflows requires an approach that deals with the root of the problem: sample preparation. A comprehensive approach is needed that addresses several key factors: 

  1. Viability. One of the most critical factors in improving cell-based assay outcomes is improving the viability of cells entering the assay by eliminating dead or dying cells in a gentle separation process that does not further impact viability10,11
  2. Debris removal. Aside from skewing viability measurements, the presence of dead cells, debris, and other toxic stimulators can cause high background noise, generate misleading signals, and potentially obscure critical information12,13
  3. Gentle sample preparation. Applying a gentle cell separation approach that does not impact cell viability or integrity, change gene expression, or alter cell population profiles is essential14,15
  4. Efficiency. When selecting the most appropriate sample preparation method for improving lab productivity, efficiency must be prioritized. 

Historically, the most commonly used cell separation techniques have involved centrifugation, bead, bubble, and fluorescence-activated cell sorting (FACS) approaches16–19. Despite their utility, each of these methods has considerable limitations that exacerbate the previously outlined challenges. For example, bead, bubble, and FACS techniques require cell labeling, introducing additional, time-intensive steps into the protocol and posing the risk of cytotoxicity. Furthermore, these methods often fail to eliminate debris from the samples. Centrifugation, conversely, does not require labeling and is capable of removing debris, yet by nature, it places significant stress on the cells, adversely affecting their viability20

Addressing these limitations is crucial for cell-based work and for in vivo models – enhancing efficiency in cultivating cells for patient-derived xenograft (PDX) models, improving outcomes and timelines in cell-based assays, maximizing research ROI, and ultimately boosting productivity in cell therapy research and development.

The answer to increasing productivity – Levitation Technology

In a bid to overcome the challenges posed by existing cell separation processes, LevitasBio developed Levitation TechnologyTM, which offers an efficient means of separating cells, substantially improving the outcomes of cell-based assays and boosting productivity. By utilizing a paramagnetic solution within a magnetic field, this technology levitates live cells, distinguishing them from dead or dying cells without the need for labels. Levitation Technology effectively removes debris from samples, yielding data with an improved signal-to-noise ratio compared to other separation methods. 

In contrast to conventional methods that can impair cell viability and integrity through harsh treatments such as dyes or centrifugation, Levitation Technology offers a gentle alternative. It minimizes cell stress and preserves cell integrity, allowing researchers to enrich for viable cells and major cell-specific subpopulations. Utilizing these high-quality cells in applications such as cell-based assays, CRISPR transductions, or PDX models leads to higher efficiencies, better model growth, and superior assay outcomes. Using high-quality cells or input material opens the door to using fewer cells per reaction. This translates to either testing a wider range of conditions or possibly working with smaller sample sizes.

Moreover, the improved starting material quality means that every sample can be used as a data point, and less filtering or data correction is required in downstream analysis. This significantly shortens timelines, enhances data validity, and avoids wasted resources and research funds. This is particularly crucial in applications like single-cell RNA sequencing, where the analysis of each individual cell is critical, or in drug dosing assays, which demand low variability and clean results across time points to accurately determine a drug’s response. By enriching for high viability and removing contaminants, this maximizes every dollar spent on sequencing and yields high-quality results. The LeviCell® platform enables researchers to achieve all these benefits through three simple steps that can be completed in just 20 minutes. 

Conclusions

Cell-based assays are indispensable for developing effective and safe cell therapies, providing crucial insights into therapeutics’ potency, characteristics, and optimal dosages. The challenges that  stem from poor-quality cell input can limit the entire development process by necessitating increased resources, time, and financial investment while also compromising the accuracy and reliability of results. Levitation Technology offers a much-needed solution for cell therapy researchers by enhancing cell viability and reducing assay variability. This method leads to improved productivity, efficiency, and reliability. Ultimately, Levitation Technology ensures the generation of high-quality samples, yields better results, and faster turn around in development time which can lead to more effective development of cell therapies. To learn more about how LevitasBio’s LeviCell® platform can help you increase productivity and output in cell-based assays, get in touch with one of our expert team members today!

References

1. Capelli C, Cuofano C, Pavoni C, et al. Potency assays and biomarkers for cell-based advanced therapy medicinal products. Front Immunol. 2023;14:1186224. doi:10.3389/fimmu.2023.1186224

2. Salmikangas P, Carlsson B, Klumb C, Reimer T, Thirstrup S. Potency testing of cell and gene therapy products. Front Med. 2023;10:1190016. doi:10.3389/fmed.2023.1190016

3. Eekels JJM, Pasternak AO, Schut AM, Geerts D, Jeeninga RE, Berkhout B. A competitive cell growth assay for the detection of subtle effects of gene transduction on cell proliferation. Gene Ther. 2012;19(11):1058-1064. doi:10.1038/gt.2011.191

4. Kiesgen S, Messinger JC, Chintala NK, Tano Z, Adusumilli PS. Comparative analysis of assays to measure CAR T-cell-mediated cytotoxicity. Nat Protoc. 2021;16(3):1331-1342. doi:10.1038/s41596-020-00467-0

5. Joaquina S, Forcados C, Caulier B, Inderberg EM, Wälchli S. Determination of CAR T cell metabolism in an optimized protocol. Front Bioeng Biotechnol. 2023;11:1207576. doi:10.3389/fbioe.2023.1207576

6. Liu Y, Wu W, Cai C, Zhang H, Shen H, Han Y. Patient-derived xenograft models in cancer therapy: technologies and applications. Signal Transduct Target Ther. 2023;8(1):160. doi:10.1038/s41392-023-01419-2

7. Bode D, Cull AH, Rubio-Lara JA, Kent DG. Exploiting Single-Cell Tools in Gene and Cell Therapy. Front Immunol. 2021;12:702636. doi:10.3389/fimmu.2021.702636

8. Niepel M, Hafner M, Mills CE, et al. A Multi-center Study on the Reproducibility of Drug-Response Assays in Mammalian Cell Lines. Cell Syst. 2019;9(1):35-48.e5. doi:10.1016/j.cels.2019.06.005

9. Caicedo JC, Cooper S, Heigwer F, et al. Data-analysis strategies for image-based cell profiling. Nat Methods. 2017;14(9):849-863. doi:10.1038/nmeth.4397

10. Reddy OL, Stroncek DF, Panch SR. Improving CAR T cell therapy by optimizing critical quality attributes. Semin Hematol. 2020;57(2):33-38. doi:10.1053/j.seminhematol.2020.07.005

11. Li A, Barabadi M, McDonald H, et al. Improving cell viability using counterflow centrifugal elutriation. Cytotherapy. 2022;24(6):650-658. doi:10.1016/j.jcyt.2022.01.008

12. Azqueta A, Stopper H, Zegura B, Dusinska M, Møller P. Do cytotoxicity and cell death cause false positive results in the in vitro comet assay? Mutat Res Toxicol Environ Mutagen. 2022;881:503520. doi:10.1016/j.mrgentox.2022.503520

13. Ölander M, Handin N, Artursson P. Image-Based Quantification of Cell Debris as a Measure of Apoptosis. Anal Chem. 2019;91(9):5548-5552. doi:10.1021/acs.analchem.9b01243

14. Box A, DeLay M, Tighe S, et al. Evaluating the Effects of Cell Sorting on GeneExpression. J Biomol Tech JBT. 2020;31(3):100-111. doi:10.7171/jbt.2020-3103-004

15. Beliakova‐Bethell N, Massanella M, White C, et al. The effect of cell subset isolation method on gene expression in leukocytes. Cytometry A. 2014;85(1):94-104. doi:10.1002/cyto.a.22352

16. Liao X, Makris M, Luo XM. Fluorescence-activated Cell Sorting for Purification of Plasmacytoid Dendritic Cells from the Mouse Bone Marrow. J Vis Exp. 2016;(117):54641. doi:10.3791/54641

17. Tomlinson MJ, Tomlinson S, Yang XB, Kirkham J. Cell separation: Terminology and practical considerations. J Tissue Eng. 2013;4:204173141247269. doi:10.1177/2041731412472690

18. Plouffe BD, Murthy SK, Lewis LH. Fundamentals and application of magnetic particles in cell isolation and enrichment: a review. Rep Prog Phys. 2015;78(1):016601. doi:10.1088/0034-4885/78/1/016601

19. Faridi MA, Ramachandraiah H, Iranmanesh I, Grishenkov D, Wiklund M, Russom A. MicroBubble activated acoustic cell sorting. Biomed Microdevices. 2017;19(2):23. doi:10.1007/s10544-017-0157-4

20. Kim IH, Yang JD, Lee DG, Chung HY, Cho BC. Evaluation of Centrifugation Technique and Effect of Epinephrine on Fat Cell Viability in Autologous Fat Injection. Aesthet Surg J. 2009;29(1):35-39. doi:10.1016/j.asj.2008.09.004