B I O P R O C E S S TECHNICAL Strategies for Producing Commercial Cell Lines One Biotech Company’s Experience Sue Clarke, Janette Dillon, Ann Smith, and Elizabeth Sotheran L orantis Ltd. (Cambridge, United Kingdom) is a discovery company developing products for antigen-specific immunotherapy, ASPECT, based on the Notch signaling pathway, which has been shown to play a key role in regulating immune responses. The Notch receptor and its associated ligands represent a family of highly conserved transmembrane proteins expressed during embryonic development in both vertebrates and invertebrates (1). Binding of a Notch ligand to a Notch receptor activates signaling cascades that alter the transcriptional program of the target cell, ultimately determining its fate. Our ASPECT platforms exploit the company’s proprietary position covering the Notch signaling pathway in immune cells and have broad clinical applications. PRODUCT: CELL-CULTURED PRODUCTS PROCESS FOCUS: PRODUCTION, ANALYTICAL METHODS DEVELOPMENT WHO SHOULD READ: PROCESS ENGINEERS, MANUFACTURING, Host Cells and Expression System: KEYWORDS: CELL CULTURE, SERUMFREE MEDIA, ASSAYS, CHO CELL LINES LEVEL: INTERMEDIATE BioProcess International The products in our current preclinical pipeline are derived from the Notch binding protein, Delta-1. To evaluate its biological efficacy, we chose to pursue several approaches. One was to produce a commercial cell line that secretes a recombinant protein derived from the extracellular domain of Delta-1 fused to an antibody constant region. This report summarizes the cell culture strategies we used to speed up the development of a suitable cell line — and ultimately the time to clinic. INITIAL CHOICES ANALYTICAL PERSONNEL 48 PHOTODISC (WWW.PHOTODISC.COM) APRIL 2004 Production of a commercial cell line involves several key decisions and activities. First you must choose the host cell type and expression system. We decided to use the glutamine synthetase (GS) gene expression system from Lonza Biologics (www.lonzabiologics.com) using CHO-K1 cells. GS is the enzyme responsible for biosynthesis of glutamine using glutamate and ammonium as substrates. When a cell line expresses GS, it can survive in a glutamine-free medium. The activity of the GS gene can also be selectively inhibited by methionine sulphoxamide (MSX). CHO-K1 cells produce endogenous GS. But they can be used to produce stable cell lines by transfecting in a GS gene and using glutamine-free medium plus MSX (at sufficient levels to inhibit the endogenous enzyme) to provide selection pressure. Use of a mammalian host cell should give our protein the appropriate posttranslational modifications. The advantages of the GS gene expression system are many. It is used throughout the industry and therefore comes with a wealth of accumulated knowledge and available experience. A fully qualified master cell bank is provided, which is a critical starting material for the whole process. GS has been used for licensed products, Figure 1: Notch ligand signaling activty pressure, maintaining segregation and thereby reducing the risk of cross-contamination and the introduction of adventitious agents. All materials are logged in by batch number, allowing full traceability. All such activities must be well documented to provide traceability and meet regulatory guidelines (2). A summary of data is not only needed for clinical submissions; it is also extremely useful when transferring the cell line to a contract manufacturer. Plasmid DNA: Table 1: Plating efficiency in four 96-well plates Expected cells/well 10 5 1 Frequency of observed cells/well 0 71 166 332 1 91 115 42 >1 222 103 10 and consequently, regulatory authorities are familiar with it. Although it carries an inherent cost, that is money well spent for a startup company. We continue to enhance the quality of our activities by sourcing raw materials, particularly those of animal origin, from reputable suppliers that can provide necessary supporting documentation. The most critical, of course, is a BSEfree source of fetal bovine serum (FBS). Our batch was specifically chosen to create stable cell lines. It is low in IgG, which aids the purification process, it supports clonal cell growth, and it is dialysed and confirmed to be glutamine free. Tissue culture procedures for our cell line are carried out in an isolated room under positive The second most important starting material is the plasmid DNA. Traditionally you would want to confirm that the correct protein is secreted, usually by transient expression, before embarking on stable cell line work. Because we are confident in both the vectors and our proteins, we now set up a stable transfection and use a bulk culture supernatant for the initial analysis at the same time we dispense a duplicate culture into 96-well tissue culture plates (plate out the culture) at a variety of lowdensity concentrations and leave the cells to grow for at least three weeks. While they grow, we fully sequence the plasmid and confirm the quality and biological activity of the secreted protein. Running these procedures in parallel can therefore save time, but it does carry a risk factor. Several methods are available for the actual transfection, and we have successfully used both electroporation and a lipid carrier such as Lipofectamine from Gibco BRL (www.gibcoBRL.com). ANALYTICAL METHODS To be able to select the desired clone in your cell line, you need the right analytical methods to measure the quantity, quality, and biological activity of the product. If you are working with novel proteins, that can be challenging because standard Circle Reader Service No. 128 CLONING Figure 2: Typical growth profile reagents may be unavailable. Reagent production must therefore be considered in the overall project timeline. Because our product is a fusion protein, we have been able to use the antibody constant region to aid us in both the quantitative analysis and subsequent purification. We use standard Western blots as a qualitative assessment for Delta, and we had to make our own polyclonal antibodies for that purpose. The efficacy of the final molecule is, of course, paramount. We made a stable CHO reporter cell line to test for that purpose. Cells are engineered to overexpress a Notch2 membrane-bound protein and also have a core binding factor 1 (CBF1)–driven luciferase construct for easy monitoring of Notch signaling (3). When the secreted recombinant ligand or test product is mobilized to an assay plate, the Delta portion of this fusion protein binds the Notch presented by the reporter cells. That in turn allows cleavage and activation of the luciferase reporter gene (Figure 1). Using the above assay, we demonstrate good activation of the reporter cell, with culture supernatants in the 10–30 µg/mL concentration range. Further in vitro analysis of our later clones demonstrated that our fusion protein delivers an active Notch signal and modulates the cytokine profiles of mouse CD4+ T cells (4). As mentioned earlier, the plated50 BioProcess International AND SELECTION Cloning: We tried two different APRIL 2004 out transfections are ready for assay after several weeks of incubation. Ideally you would assay several hundred clones and take a reasonable number of them on through further selection and cloning steps. However, that process can be extremely labor intensive. A more realistic task (given the limited resources of a small biotech company) would be to reduce significantly the number of cultures assessed. The final endpoint does not change: You still need a highproducing, stable clone. Cloning and selection strategies therefore become vital. Figure 3: Stability study cloning and plating strategies to reach our endpoint. The first approach took about 12 months from gene to final stable cell line. We initially plated out the cells at a nonclonal dilution and tested all the wells (12 ⫻ 96). We then expanded a selection of wells and performed further assays, while preparing cryopreserved stocks as back-up. The best clone was subjected to a limiting dilution cloning. The last two steps were repeated, and four of the resulting clones were selected as candidates to adapt to suspension. Each candidate can take up to two months to clone, select, and freeze. The second method we used aimed to achieve a confidence of clonality at an earlier stage. Table 1 shows the numbers of clones that were microscopically observed when plating the cells three to five days posttransfection. It can been seen from these results that the lower dilutions — five and one expected cells/well — gave us enough wells that appeared as a single colony. The probability that these single colonies come from one cell is increased at the lower dilutions. Transfection efficiency also affects the actual Figure 4: Analytical cloning above. So we used analytical cloning for both clones and found that #0304 was 96% positive, but #0342 was only 56% positive for protein production. Careful observation of the plates microscopically gave us more information. Had we needed to, we could have subcultured the clones at a lower confluency range but higher productivity (Figure 4). number of cells that survive in the selection medium at this initial phase. We therefore took the highest producing clones from the lower dilution plates to increase the chance of clonality. In this experiment, we performed one additional round of limiting dilution cloning to select the final clones. Note here that the best producer in the static flask may not be the best producer once adapted to suspension, so a wider choice is always the preference. This second method reduced the timeline by six months and still yielded a good quality cell line. Selection: Whenever possible, selection of the optimal clone should take into consideration the eventual production process. So the last stage added to both of the above methods is taking the final clones and adapting them to both suspension and a protein-free medium to make them more readily scalable. Again, realistically only a few can probably be handled. We adapted the clones by reducing the percentage of FBS in the adherent culture to 2% and then inoculating a 500-mL spinner or shake flask with 4–5 ⫻ 105 cells/mL in protein-free Ex-Cell 325 medium from JRH Biosciences (www.jrhbiosciences.com), giving a final concentration of 1% FBS. Then, additions of 325 medium alone were made, so the cells slowly (about three months) adapted to the new culture system. Figure 2 is a 52 BioProcess International APRIL 2004 typical growth profile of an adapted clone followed for 17 days. Once adapted, the final clones must be tested for stability to ensure that they are robust enough for a manufacturing campaign — normally at least 50–60 population doublings. A stock culture is maintained and defined in terms of population doublings (5). At regular intervals (about 10 weeks apart), a normally prepared production vessel set up from the stock flask is used to assess productivity in relation to the overall population generations. Figure 3 shows that for a sixweek study, two of the clones looked stable. However, when we continued the study for a further few weeks, clone #LC06-0342 lost productivity. Analytical Cloning: A faster method for testing stability was to perform an analytical cloning as an assessment of population drift. Ideally, 100% of a given population produces the target protein. If a population has nonproducers, they could outgrow the producers, causing an instability in the culture over time. An analytical cloning therefore examines the distribution of cells at a concentration that permits all apparently single-cell wells to be tested for protein expression. This method takes only three weeks at most — two weeks for the clones to grow and a week for analysis — instead of the ten weeks required by the method described MAKE IT QUICK In summary, building in quality from the start and having the appropriate assays available make it possible for a small biotech company to produce a suitable cell line for a phase I study. Timelines can be significantly reduced — even when resources are limited — by the simple cloning strategies described here. REFERENCES 1 McKenzie, GJ; et al. Notch Signaling in the Regulation of Peripheral T-Cell Function. Seminars in Cell & Developmental Biology 2003, 14: 127–134. 2 Center for Biologics Evaluation and Research. Guidance for Industry: Q5D Quality of Biotechnological/Biological Products — Derivation and Characterisation of Cell Substrates Used for Production of Biotechnological/Biological Products. US Food and Drug Administration, 1998; www.fda.gov/cber/qdlns/qualbiot.txt. 3 Wong KK; et.al. Notch Ligation by Delta 1 Inhibits Peripheral Immune Responses to Transplantation Antigens By a CD8+ Cell Dependent Mechanism. J. Clinical Investigation, in press. 4 Young L; et al. The Notch Ligand Delta 1 Selectively Enhances the Production of IL-10 in Murine T Cells. Immunology 2002, 107, Supplement 1: 1–7. 5 McAteer, JA; Davis JM. Basic Cell Culture Techniques and the Maintenance of Cell Lines. In Basic Cell Culture, second edition (University Press: Oxford, UK, 2001), p. 166. Corresponding author Sue Clarke is head of manufacturing development; and Janette Dillon, Ann Smith, and Elizabeth Sotheran are development scientists at Lorantis Ltd., 410 Cambridge Science Park, Cambridge CB4 0PE, UK; 44-1223-702500, fax 44-1223-702599; sue.clarke@ lorantis.com, www.lorantis.com.
© Copyright 2026 Paperzz