Mammalian display was originally conceived for affinity maturation of single-chain variable fragments (scFv) expressed on the surface of human cells (
1- Ho M.
- Nagata S.
- Pastan I.
Isolation of anti-CD22 Fv with high affinity by Fv display on human cells.
) and has been further developed for screening full-length antibody cell surface–expressed libraries (
2- Akamatsu Y.
- Pakabunto K.
- Xu Z.
- Zhang Y.
- Tsurushita N.
Whole IgG surface display on mammalian cells: application to isolation of neutralizing chicken monoclonal anti-IL-12 antibodies.
,
3- Zhou C.
- Jacobsen F.W.
- Cai L.
- Chen Q.
- Shen W.D.
Development of a novel mammalian cell surface antibody display platform.
). The use of mammalian cell display (
4- Zhou Y.
- Chen Z.-R.
- Li C.-Z.
- He W.
- Liu S.
- Jiang S.
- Ma W.-L.
- Tan W.
- Zhou C.
A novel strategy for rapid construction of libraries of full-length antibodies highly expressed on mammalian cell surfaces.
) has some important advantages over other display systems (
e.g. phage/yeast), in particular in relation to the manufacturability of the identified antibodies; in mammalian display, antibodies are produced using the endogenous eukaryotic secretion machinery enabling correct folding and biophysical properties and are therefore more likely to be compatible with mammalian cell production systems. However, a disadvantage of using mammalian display is that only a relatively small library size (usually up to 10
7) can be interrogated. The library sizes that are available for mammalian systems are typically limited by low transfection efficiency, although recent advances have improved this, for example by using CRISPR–Cas9 integration methods (
5- Parola C.
- Neumeier D.
- Friedensohn S.
- Csepregi L.
- Di Tacchio M.
- Mason D.M.
- Reddy S.T.
Antibody discovery and engineering by enhanced CRISPR–Cas9 integration of variable gene cassette libraries in mammalian cells.
,
6- Parthiban K.
- Perera R.L.
- Sattar M.
- Huang Y.
- Mayle S.
- Masters E.
- Griffiths D.
- Surade S.
- Leah R.
- Dyson M.R.
- McCafferty J.
A comprehensive search of functional sequence space using large mammalian display libraries created by gene editing.
). Alternatively, library size can be effectively expanded by first utilizing phage display (
7- Frenzel A.
- Schirrmann T.
- Hust M.
Phage display-derived human antibodies in clinical development and therapy.
,
8- Frenzel A.
- Kügler J.
- Helmsing S.
- Meier D.
- Schirrmann T.
- Hust M.
- Dübel S.
Designing human antibodies by phage display.
) to screen much larger naïve libraries before converting to mammalian cell display after one or two rounds of selection or by using libraries derived from immunized animals, in which initial antibody selection and maturation has occurred
in vivo (
6- Parthiban K.
- Perera R.L.
- Sattar M.
- Huang Y.
- Mayle S.
- Masters E.
- Griffiths D.
- Surade S.
- Leah R.
- Dyson M.R.
- McCafferty J.
A comprehensive search of functional sequence space using large mammalian display libraries created by gene editing.
). With current mammalian display methods, the cells displaying the antibodies are incubated with the target antigen, which must be available in a soluble format and used either free in solution or bound to paramagnetic beads (
1- Ho M.
- Nagata S.
- Pastan I.
Isolation of anti-CD22 Fv with high affinity by Fv display on human cells.
,
9- Beerli R.R.
- Bauer M.
- Buser R.B.
- Gwerder M.
- Muntwiler S.
- Maurer P.
- Saudan P.
- Bachmann M.F.
Isolation of human monoclonal antibodies by mammalian cell display.
,
10- Bowers P.M.
- Horlick R.A.
- Kehry M.R.
- Neben T.Y.
- Tomlinson G.L.
- Altobell L.
- Zhang X.
- Macomber J.L.
- Krapf I.P.
- Wu B.F.
- McConnell A.D.
- Chau B.
- Berkebile A.D.
- Hare E.
- Verdino P.
- et al.
Mammalian cell display for the discovery and optimization of antibody therapeutics.
). The latter system can be advantageous in enhancing antigen avidity, thus allowing for the selection of cells that express low affinity antibodies (
11- Breous-Nystrom E.
- Schultze K.
- Meier M.
- Flueck L.
- Holzer C.
- Boll M.
- Seibert V.
- Schuster A.
- Blanusa M.
- Schaefer V.
- Grawunder U.
- Martin-Parras L.
- van Dijk M.A.
Retrocyte Display® technology: generation and screening of a high diversity cellular antibody library.
). Because purified antigen must be applied to cells in solution or coupled to particles, the target is typically restricted to proteins or protein domains that are soluble and relatively stable; thus, identifying antibodies to membrane proteins remains challenging. Although whole-cell panning methods can be used with phage display to enrich for phage that bind to complex membrane proteins (
12- Nikfarjam S.
- Tohidkia M.R.
- Mehdipour T.
- Soleimani R.
- Rahimi A.A.R.
- Nouri M.
Successful application of whole cell panning for isolation of phage antibody fragments specific to differentiated gastric cancer cells.
,
13- Jones M.L.
- Alfaleh M.A.
- Kumble S.
- Zhang S.
- Osborne G.W.
- Yeh M.
- Arora N.
- Hou J.J.
- Howard C.B.
- Chin D.Y.
- Mahler S.M.
Targeting membrane proteins for antibody discovery using phage display.
), the phage require reformatting, and this process can frequently ablate binding activity, thereby reducing the number of positive binders for subsequent analysis. This process also nonspecifically enriches for phage against unrelated, nontarget, cell surface proteins.
The technology to screen large naïve libraries within a mammalian setting would clearly confer a significant advantage by improving the compatibility and developability of identified antibodies with mammalian cell manufacturing systems, without the requirement to use two distinct discovery methods. Furthermore, because many important therapeutic targets are membrane proteins, the ability to screen against a membrane antigen in its native configuration within a cellular membrane environment would ensure that only physiologically relevant epitopes are presented, thus giving a greater likelihood of identifying functional antibodies. To provide a mammalian display technique with both these major advantages, we here describe a method by which we package large antibody libraries with diversities of ∼10
9 into lentiviral particles and use these to transduce CHO cells that have been engineered to express the target membrane protein. This allows much larger library sizes to be sampled than with existing methods (by at least 100-fold) and is only limited by the number of cells that can be cultured in the laboratory. Our strategy results in individual CHO cells expressing the target antigen on their cell surface while co-expressing and secreting a variant from the high diversity scFv library. Thereby, CHO cells that express an scFv variant capable of binding the target undergo self-labeling, thus allowing them to be isolated, reselected, and eventually sequenced. Reported strategies have identified co-expression of scFvs and antigen in a single cell culture as a method to screen for antibodies, for example in a bacterial display system (
14- Guo X.
- Cao H.
- Wang Y.
- Liu Y.
- Chen Y.
- Wang N.
- Jiang S.
- Zhang S.
- Wu Q.
- Li T.
- Zhang Y.
- Zhou B.
- Yin J.
- Li D.
- Ren G.
Screening scFv antibodies against infectious bursal disease virus by co-expression of antigen and antibody in the bacteria display system.
) and for affinity maturation in mammalian cells (
15- Eguchi A.
- Nakakido M.
- Nagatoishi S.
- Kuroda D.
- Tsumoto K.
- Nagamune T.
- Kawahara M.
An epitope-directed antibody affinity maturation system utilizing mammalian cell survival as readout.
). However, we describe for the first time the screening of a large naïve scFv library fully in a mammalian system and identify binders to a membrane protein antigen presented on the cell surface.
Discussion
Our approach has successfully identified scFvs against EpCAM using a naïve library that mimics the natural naïve human repertoire. This was achieved completely in a mammalian CHO cell line with no requirement for performing initial panning steps in other systems. This has the potential to be exploited as a novel approach to identifying new therapeutic antibodies against other therapeutic membrane proteins in their native environment in vitro and in a mammalian cell system routinely used for antibody expression and manufacture, thus ensuring the compatibility of the discovered antibodies with these systems.
The use of lentiviruses enabled transduction of large numbers of mammalian cells, and their preference for regions of open chromatin (
22- Baranyi L.
- Roy A.
- Embree H.D.
- Dropulic B.
Lentiviral vector-mediated genetic modification of cell substrates for the manufacture of proteins and other biologics.
) was expected to ensure that the majority of the library was sufficiently expressed for detection of self-labeling. The dynamics of the transduction (
i.e. the number of cells transduced by none, one, or more particles) were expected to follow a Poisson distribution and using a MOI of 1.0 theoretically resulted in 37% of cells with no events, 37% cells with one integration event, 18% cells with two integration events, and 8% cells with more than two integration events. In the current selection campaign described in this work, we did not isolate any cells with more than one integration event; however, in other campaigns (data not shown), we have isolated positive binders from cells with two integration events where both scFvs were expressed. This required resolution of the correct binder, which did not overly add to the complexity of the process, although resolving a situation with more than two integration events would be more challenging; because the frequency of more than two integration events is relatively low, we do not expect this to be problematic for selection campaigns. Furthermore, we are working to fine-tune the MOI to define the condition that provides the best compromise between the number of multiple integration events, the number of cells with no integration event and the total number of cells that can be successfully handled for the first selection step. In this work, the initial selection step was done with 4 × 10
9 cells, and we believe that this can be routinely increased to at least 10
10 with the implementation of further automation.
The major challenges of this approach lie in the potential for cross-labeling (where antibody secreted by one cell binds to the surface of another cell) and off-target labeling (where scFv specific for CHO cell surface proteins self-label or cross-label). We do not believe that cross-labeling is significantly problematic during early selections because the numbers of antigen-positive cells in the initial population of cells is extremely low, but the significance of the problem increases as the proportion of antigen-positive cells increases during selection. Therefore, we implemented two strategies to mitigate against these issues: the use of decoy cells expressing antigen only to reduce cross-labeling, combined with FRET as a method for reducing off-target labeling. Of the 93 clones selected after a second round of FRET-based cell sorting, 66 clones secreted an scFv that appeared to bind to the CHO-X parental cell line, whereas 26 were found to give an increased self-labeling signal in the presence of cell surface EpCAM (
Fig. S2). Only one of the clones secreted an scFv that did not bind either to CHO-X or CHO–EpCAM cells, which suggested that cross-labeling had been effectively suppressed. In contrast, the FRET method to reduce the selection of off-target binders appears to have been only moderately successful, and there is still room for improvement for the avoidance of selection of cells secreting an off-target binder. Potentially, improving the intensity of the FRET signal and using a more stringent FACS-gating strategy will reduce the number of off-target binders. Nevertheless, the current rate of selection of off-target labeled cells is manageable. The FRET approach described herein also employed the use of a second antibody that was already known to bind the target; this may not be possible in instances in which the target is particularly challenging. However, the genetic addition of an established epitope tag has been successfully used as an alternative strategy.
One further issue we encountered was the selection of three antibodies that appeared to bind strongly to CHO–EpCAM cells with no signal against CHO-X cells (
Fig. S3); one antibody in particular was heavily selected for. When these antibodies were recloned as scFv, they bound weakly to CHO–EpCAM cells but did not bind when expressed as whole IgG. The reasons for this are currently unclear.
Although the final number of initial antigen-positive cells selected was low (
26- Vander Heiden J.A.
- Yaari G.
- Uduman M.
- Stern J.N.
- O'Connor K.C.
- Hafler D.A.
- Vigneault F.
- Kleinstein S.H.
PRESTO: a toolkit for processing high-throughput sequencing raw reads of lymphocyte receptor repertoires.
), this was primarily caused by low efficiency outgrowth of single-cell clones following FACS, which was as low as 20% (data not shown). Improvements in cell outgrowth and increasing the numbers of cells sorted will increase the chances of selecting true antigen-positive cells and increase the diversity of sequences selected for. Further improvements in library design and creation are also required to increase the probability of selecting diverse sequences; the current library had a maximum theoretical diversity of ∼8.0 × 10
8, whereas the number of independent clones was calculated to be ∼6 × 10
8. As mentioned above, improvements in cell handling should allow us to transduce ∼10
10 cells with Poisson statistics dictating that ∼5.5 × 10
9 cells will contain one or two integration events; thus, interrogation of libraries of at least 10
9 is feasible. Although this is not as large as some phage display libraries that have been created (
23- Söderlind E.
- Strandberg L.
- Jirholt P.
- Kobayashi N.
- Alexeiva V.
- Åberg A.-M.
- Nilsson A.
- Jansson B.
- Ohlin M.
- Wingren C.
- Danielsson L.
- Carlsson R.
- Borrebaeck C.A.
Recombining germline-derived CDR sequences for creating diverse single-framework antibody libraries.
,
24- Prassler J.
- Thiel S.
- Pracht C.
- Polzer A.
- Peters S.
- Bauer M.
- Nörenberg S.
- Stark Y.
- Kölln J.
- Popp A.
- Urlinger S.
- Enzelberger M.
HuCAL PLATINUM, a synthetic fab library optimized for sequence diversity and superior performance in mammalian expression systems.
,
25- Tiller T.
- Schuster I.
- Deppe D.
- Siegers K.
- Strohner R.
- Herrmann T.
- Berenguer M.
- Poujol D.
- Stehle J.
- Stark Y.
- Heßling M.
- Daubert D.
- Felderer K.
- Kaden S.
- Kölln J.
- et al.
A fully synthetic human Fab antibody library based on fixed VH/VL framework pairings with favorable biophysical properties.
)
, our goal has been to create libraries that contain a high proportion of functional sequences that are designed to avoid sequence motifs that may be problematic during manufacturing. Despite these limitations, we were able to isolate a specific EpCAM binder with subnanomolar affinity.
We believe that this system has a particular utility for identifying scFv binders against membrane proteins for CAR-T cell applications. To this end, we reformatted the top three binders as second-generation CARs, which were transduced into both CD3+ T cells isolated from human PBMC and Jurkat cells. We were able to demonstrate that the two higher-affinity scFv CAR constructs (SP14-C8 and SP12-E10) were capable of triggering antigen-specific activation of the primary T cells and directing potent cytotoxicity against CHO–EpCAM cells. SP14-C8 CAR-transduced Jurkat cells were also potently activated in the presence of an antigen-positive MCF-7 breast cancer cell line and CHO–EpCAM cells, although the SP12-E10 CAR was not active in this assay, possibly due to the activation kinetics of the cells, the constraints imposed by a CAR-T configuration compared with that of a soluble scFv, or the lower affinity of this antibody.
In conclusion, we believe that the novel display method reported here is the first platform enabling the discovery of target specific scFvs from an untrained library in a fully mammalian cell system. This platform offers the opportunity to identify novel antibodies against membrane proteins in a physiological setting, with desirable properties for use either in a CAR-T cell setting or as a whole mAb therapy.
Materials and methods
Construct design
A self-inactivating pSF lentivector was modified to accommodate subcloning of PciI/XbaI EpCAM DNA (accession no. BC014785.1) fragments. The pSF lentivector contains a cytomegalovirus promoter with TetO sites that drives the expression of the transgene as well as a TetR gene via an spleen focus forming virus promoter downstream of the transgene, allowing control of the target protein by doxycycline. A puromycin resistance gene was incorporated following the TetR gene and separated by an encephalomyocarditis virus (EMCV) IRES sequence to enable antibiotic selection following transduction. Finally, the woodchuck hepatitis virus post-transcriptional regulatory element was incorporated downstream of the puromycin resistance gene.
For the scFv library construction, a self-inactivating pSF lentivector was modified to accommodate subcloning of KpnI/XhoI scFv DNA fragments. The pSF lentivector has a spleen focus forming virus promoter that drives the expression of the transgene followed by the woodchuck hepatitis virus post-transcriptional regulatory element downstream. The EpCAM-binding scFv-positive control was derived from sequence 75 from U.S. patent 7435549. The CHESS scFv library was constructed by randomly combining all known functional human germline V, D, and J segments that had been synthesized de novo (TWIST Bioscience, San Francisco, CA. USA). Human germline Vκ and Vλ elements flanked by SpeI/SapI restriction sites were ligated to human germline Jκ and Jλ elements flanked by SapI/PciI restriction sites. A signal sequence was incorporated upstream of the light chain segment and a 15-amino acid GS-linker downstream. The heavy chains were constructed from all the known human germline VH sequences flanked by XbaI and SapI restriction sites, which were ligated to the D and JH segments that had been constructed from an oligonucleotide pool annealed and PCR-amplified with adapter sequences flanked by SapI/XhoI restriction sites. Nucleotides were added to the 5′ ends of the D segments to ensure representation of all three reading frames. All assembled segments were transformed via electroporation into competent Escherichia coli (MC1061 F− electrocompetent cells; Cambridge Bioscience, catalog no. 60514-2) and titred to determine the number of individual transformants. We are willing to supply this CHESS scFv library described to anyone who requests it for a stated purpose, subject to a material transfer agreement.
Next-generation sequence analysis of scFv library
Library amplicons were sequenced by GENEWIZ using paired-end reads on an Illumina based platform. Next-generation sequencing (NGS) reads were processed using the Immcantation pipeline (
26- Vander Heiden J.A.
- Yaari G.
- Uduman M.
- Stern J.N.
- O'Connor K.C.
- Hafler D.A.
- Vigneault F.
- Kleinstein S.H.
PRESTO: a toolkit for processing high-throughput sequencing raw reads of lymphocyte receptor repertoires.
) alongside custom R packages. IgBLAST (
27- Ye J.
- Ma N.
- Madden T.L.
- Ostell J.M.
IgBLAST: an immunoglobulin variable domain sequence analysis tool.
) was used to perform antibody-based annotation on NGS reads. The Immcantation R package, Alakazam (
28- Gupta N.T.
- Vander Heiden J.A.
- Uduman M.
- Gadala-Maria D.
- Yaari G.
- Kleinstein S.H.
Change-O: a toolkit for analyzing large-scale B cell immunoglobulin repertoire sequencing data.
), was used to load tabulated NGS data into R, and the composition of the CDR regions within the scFv library population was determined. Sequence logos and histograms were produced using ggseqlogo and ggplot2, respectively (
29ggplot2: Elegant Graphics for Data Analysis.
,
30Ggseqlogo: a versatile R package for drawing sequence logos.
).
Cell lines
OXGENE in-house CHO cells (CHO-X) were maintained and expanded for experiments in complete growth medium containing ProCHO 5 (Lonza, catalog no. BE12-766Q) supplemented with 4 mm UltraGlutamine (Lonza, catalog no. BE17-605E/U1) in shaking flasks (Corning) at 37 °C under 5% CO2 (125 rpm in a Climo-Shaker ISF1-XC incubator, Kuhner AG).
A CHO-X cell pool stably expressing full-length EpCAM was initially generated by transduction of CHO-X at a density of 0.3 × 106 cells/ml with a lentivirus packaging the EpCAM gene and a puromycin-resistant marker at MOI of 2.5. 72 h after transduction, the medium was replaced, and the cells were further cultured in medium containing puromycin (3 µg/ml) for 14 days. After recovery and expansion of the pool, clonal cell lines were generated by cell surface staining with an EpCAM FITC antibody (BD Biosciences, catalog no. 347197) and single-cell sorting on a SH800 Cell Sorter (Sony). The 64% highest-expressing cells were single cell sorted into 96-well plates. The clones were manually picked on day 18 and expanded. CHO clones were screened for EpCAM expression by flow cytometry 35 days after sorting, and clone SP1D12 was chosen for further work based on EpCAM expression levels.
Adherent HEK-293T cells European Collection of Authenticated Cell Cultures (ECACC) were grown in Dulbecco's modified Eagle's medium (Sigma–Aldrich, catalog no. D5796) supplemented with 10% FBS (Thermo Fisher Scientific, catalog no. 10500-064) in a static incubator (CB 160, Binder) at 37 °C under 5% CO2. In-house generated HEK-293 EBNA suspension adapted cells (293OX-EBNA) were grown in BalanCD HEK293 (Irvine Scientific, catalog no. 91165) supplemented with 4 mm UltraGlutamine at 37 °C under 8% CO2 (125 rpm in a Climo-Shaker ISF1-XC incubator).
Jurkat cells (CLS) were maintained in RPMI 1640 (Sigma, catalog no. R8758) supplemented with 10% FBS in a static incubator (CB 160, Binder) at 37 °C under 5% CO2. MCF-7 cells were maintained in minimal essential medium (Sigma–Aldrich, catalog no. M4655), 2 mm GlutaMAX (Thermo Fisher Scientific, catalog no. 35050061), sodium pyruvate (Thermo Fisher Scientific, catalog no. 11360-070), nonessential amino acids (Thermo Fisher Scientific, catalog no. 11140035), and 10% FBS (Thermo Fisher Scientific, catalog no. 10500-064) in a static incubator (CB 160, Binder) at 37 °C under 5% CO2.
Lentiviral preparation
The CHESS scFv synthetic library was packaged into replication-incompetent lentiviral particles in HEK-293T cells. 2 × 107 cells were seeded 3 days before transfection in hyperflasks (Corning, catalog no. 10020). The cells were transfected with 760 µg of total DNA (CHESS library and lentiviral packaging vectors) and 1900 µg of branched polyethyleneimine transfection reagent (Sigma–Aldrich, catalog no. 408727). The resulting lentiviral supernatants were harvested 3 days later, clarified by centrifugation at 800 × g for 10 min, and stored frozen at −80 °C. The physical titer of the lentiviral preparations was determined by quantitative PCR, and the infectious titer on CHO–EpCAM cells of lentiviruses encoding GFP was determined by flow cytometry. The infectious titer of the library lentiviral preparation was calculated by using the physical/infectious titer ratio determined for the GFP lentiviruses.
Library transduction
1.35 × 109 CHO–EpCAM cells adjusted to a cell density of 0.9 × 106 cells/ml were transduced with lentiviral particles packaging the scFv library at MOI = 1. Cell culture medium was then supplemented with 1 µg/ml doxycycline (Sigma–Aldrich, catalog no. D3072) to induce EpCAM expression. After 3 days the medium was changed, and the cells were incubated overnight at 25 °C in culture medium with 5 µg/ml doxycycline before magnetic selection (MACS).
Magnetic activated cell sorting
MACS was done using an autoMACS Pro Separator (Miltenyi Biotec) according to the manufacturer's instructions. MACS buffer consisting of PBS, pH 7.4 (Thermo Fisher Scientific, catalog no. 14190-094), with 10% FBS and 2 mm EDTA (Sigma–Aldrich, catalog no. 03690) was used throughout the process. For the first round of selection, 4 × 109 CHO–EpCAM cells transduced with the lentiviral library were labeled with 400 μl of PE-conjugated anti-HA antibody (1 μl/10 × 106 cells, Miltenyi Biotec, catalog no. 130-120-717) for 30 min at 4 °C followed by labeling with 4 ml of anti-PE microbeads (10 μl/10 × 106 cells, Miltenyi Biotec, catalog no. 130-048-801) for 25 min at 4 °C and subsequent positive cell selection using the AutoMACS instrument (program Depl025). In subsequent rounds, the process was repeated with quantities of detection reagents adjusted for the input quantity of cells.
Between rounds, the enriched fraction of cells was allowed to recover for 3 days before reinduction with 1 µg/ml doxycycline for a further 3 days. The culture medium containing doxycycline was then replaced, and cells were incubated overnight at 25 °C prior to the subsequent sort.
FACS selection of FRET-positive cells
CHO–EpCAM cells were labeled with CellTrackerTM Blue CMAC dye (Thermo Fisher Scientific, catalog no. C2110) and added in excess (9:1) to either the output from the third round of MACS selection or the output from the first FACS pool sort. Briefly, 5 × 107 cells were centrifuged (300 × g for 5 min) and resuspended in PBS, pH 7.4, containing 1% BSA and 150 μm CMAC. The cell suspension was incubated for 40 min at 37 °C in a shaking incubator for intracellular labeling. CMAC-labeled cells were washed once with PBS, pH 7.4, 1% BSA to remove excess dye. Cell suspensions of capturing decoy cells, as well as the cell sample to sort were prepared at 0.4 × 106 cells/ml in culture medium containing doxycycline.
FACS on these induced cells was performed using an SH800 cell sorter (Sony) with Cell Sorter software (Sony). In the first round of FACS, 5 × 107 cells were washed once with culture medium containing 2 mm EDTA and stained in the same medium with 5 nm anti-HA-PE antibody (Miltenyi Biotec) and 30 nm anti-EpCAM–Alexa Fluor 647 antibody (Biolegend, catalog no. 324212) for 30 min at 4 °C. Control cells were single stained with each antibody separately under the same conditions. The cells were washed with culture medium containing EDTA and finally resuspended in the same buffer for analysis and sorting. Live cells were gated based on scatter properties; for simplicity a cell viability dye was not used. The sort gate was drawn on a bidimensional dot plot showing the emission signal of Alexa Fluor 647 versus PE.
For the first sort, 36,000 cells were sorted into 50% conditioned growth medium, 2.5% ClonaCellTM–CHO ACF supplement (STEMCELL Technologies, catalog no. 03820), 40% fresh growth medium and allowed to recover and expand. A second round of FRET selection was performed following the same protocol except that MACS buffer was used throughout the process, and the cell suspension was sorted into single cells in 75% conditioned growth medium, 2.5% ClonaCellTM, 15% fresh growth medium. The cells were expanded and supplemented with 50% fresh medium after 2 weeks, followed by transfer to fresh growth medium after about 3 weeks.
Flow cytometry analysis
Flow cytometry immunostaining experiments were performed using an AttuneTM NxT flow cytometer (Thermo Fisher Scientific) equipped with two lasers of 488 and 561 nm, connected to an AttuneTM NxT acoustic focusing cytometer autosampler (Thermo Fisher Scientific). The samples were stained in and sampled from U-bottom 96-well plates (Corning). The cell samples were harvested and washed once with MACS buffer before incubation for 30 min at 4 °C with the corresponding antibody diluted in MACS buffer. The cells were washed twice with the same buffer before final resuspension. The cells were gated based on scatter properties, and doublets were excluded by plotting FSC-H versus FSC-A. At least 10,000 events on the single cells gate were acquired. Flow cytometry data were analyzed using AttuneTM NxT software (Thermo Fisher Scientific).
Expression of recovered scFv and whole IgG
ScFvs isolated from clonal CHO–EpCAM cells were recovered by PCR and recloned either as scFv into the lentiviral expression plasmid as described above or as whole IgG1 into a dual expression vector, where the heavy and light chains were expressed from separate gene cassettes on the same plasmid, which also contained an EBV OriP element. Plasmid DNA was prepared and used to transfect suspension 293OX-EBNA cells using FectoPro (Polyplus, catalog no. 116-001) according to the manufacturer's instructions in a volume of either 4 ml for supernatant analysis or 200 ml for purification of whole IgG. Protein A purification was done using 1 ml of HiTrap MabSelect SuRe columns (GE Healthcare, catalog no. 11003493) according to the manufacturer's instructions. Purified antibodies were desalted using Zeba Spin columns (Thermo Fisher Scientific, catalog no. 89891) and quantified by absorbance at 280 nm.
CHO–EpCAM cell binding of scFv and whole IgG1 antibodies derived from anti-EpCAM hits
The supernatants from either scFv or whole IgG transfections were incubated with either control CHO-X cells or CHO–EpCAM cells for 1 h at 37 °C in a shaking incubator and then were washed twice with MACS buffer before resuspension and incubation with either anti-HA–PE or anti-Fc–PE at 4 °C for 30 min. Finally, the cells were washed a further two times before analysis on an Attune NxT flow cytometer.
For the EC50 determination, titrations (0.1 nm to 10 μm in doubling dilutions) of purified whole IgG1 antibodies derived from the clones SP14-C8, SP17-F7, and SP12-E10 were applied to either CHO-X control cells or CHO–EpCAM cells and incubated for 1 h at 37 °C in a shaking incubator and then washed twice with MACS buffer before resuspension and incubation with anti-Fc–PE (Biolegend, catalog no. 409304) at 4 °C for 30 min. Finally, the cells were washed a further two times before analysis on an Attune NxT flow cytometer. The cells were gated based on scatter properties, and doublets were excluded by plotting FSC-H versus FSC-A. At least 10,000 events on the single cells gate were acquired. Flow cytometry data were analyzed using AttuneTM NxT software.
Affinity measurements using surface plasmon resonance
The affinity of purified whole IgG EpCAM-positive antibody variants for the EpCAM ECD was determined by surface plasmon resonance (SPR) measurements on a Biacore T200 instrument (GE Healthcare). Each of the anti-EPCAM antibody variants was immobilized onto the surface of a protein A sensor chip. The assays were run in PBST buffer (0.05% Tween 20, PBS, pH 7.4), and recombinant EpCAM-ECD (AcroBiosystems, catalog no. EPM-H5223) was assayed for binding at concentrations between 0.3 nm and 10 μm, followed by a single dissociation step (binding parameters were determined with the Langmuir single binding site model using Biacore T200 evaluation software 2.0.3 (GE Healthcare)).
The second SPR experiment was performed by EpCAM-ECD (AcroBiosystems, catalog no. EPM-H5223) immobilized onto the surface of a sensor chip by an anti–His tag capture antibody. The assays were run in PBST buffer (0.05% Tween 20, PBS, pH 7.4) and recombinant anti-EpCAM antibodies were assayed for binding at concentrations between 1 nm and 3.33 μm, followed by a single dissociation step (binding parameters were determined with the Langmuir single binding site model using Biacore T200 evaluation software 2.0.3 (GE Healthcare)).
Processing of peripheral blood mononuclear cells and T-cell isolation
Human peripheral blood from anonymized healthy donors was obtained from the National Health Service Blood and Transfusion Service. PBMCs were purified by Ficoll (GE Healthcare, catalog no. 17144002) density gradient centrifugation as recommended by the manufacturer. For extraction of CD3-positive T cells from PBMC, non-CD3 cells were depleted using a Pan T cell isolation kit (Miltenyi Biotec, catalog no. 130-096-535). Isolated T cells were resuspended in RPMI 1640 (Gibco, catalog no. R8758) supplemented with 10% heat-inactivated fetal bovine serum, 2 mm l-glutamine (Gibco, catalog no. 25030-081), 10 mm HEPES (VWR Life Science, catalog no. J848), 0.5 mm sodium pyruvate (Sigma, catalog no. S8636), 1× nonessential amino acids (Gibco, catalog no. 11140-035), 100 units/ml penicillin, and 100 μg/ml streptomycin (Gibco, catalog no. 15140-122) (T-cell medium).
CAR transduction and expansion of CD3 T cells
On day 0, T cells were resuspended in T-cell medium at 106 cells/ml and dispensed at 100 μl/well into a 96-well plate. 2 μl/well CD3/CD28 Dynabeads (Thermo Fisher Scientific, catalog no. 111.61D) were washed following the manufacturer guidelines, resuspended in 50× volume T-cell medium containing 200 units/ml IL-2 (Miltenyi Biotec, catalog no. 130-097-746) and 20 ng/ml IL-7 (Miltenyi Biotec, catalog no. 130-095-362) and added to the 96-well plate at 100 μl/well. On day 1, 120 μl of medium was removed and replaced with lentiviruses encoding candidate CARs (MOI = 10). On day 5, the cells were transferred to a 24-well plate and cultured in cytokine supplemented T-cell medium until the cells were ∼90–95% confluent; thereafter cultures were scaled up to a T25 flask.
Assessment of CAR-T cell activation and cytotoxicity during culture with target cells
Expanded CAR-T cells derived from two healthy donors were independently harvested, counted, and incubated with either CHO-X or CHO–EpCAM cells (target/effector ratio, 1:5). After 48 h, the cell samples were harvested and analyzed by flow cytometry to measure T-cell activation using an anti-CD25 antibody (Clone BC96, Biolegend, catalog no. 302606). Supernatants were also harvested, and total cytotoxicity was assessed by measuring release of lactate dehydrogenase using a CytoTox 96 nonradioactive cytotoxicity assay (Promega, catalog no. G1780)
Analysis of time-dependent cytotoxicity using xCELLigence
Viability of CHO-X and CHO–EpCAM cells incubated with transduced CAR-T cells (ratio of 1:5) was monitored in real time using xCELLigence RTCA DP technology (Acea Biosciences). The cells were incubated for 24 h (37 °C, 5% CO2) before CAR-T cells were added, and cell impedance was measured every 15 min for a total of 120 h.
CAR transduction and expansion of Jurkat cells
Jurkat cells were incubated in culture medium (RPMI 1640, 10% FBS) at 2 × 10
5- Parola C.
- Neumeier D.
- Friedensohn S.
- Csepregi L.
- Di Tacchio M.
- Mason D.M.
- Reddy S.T.
Antibody discovery and engineering by enhanced CRISPR–Cas9 integration of variable gene cassette libraries in mammalian cells.
cells/well with lentiviruses encoding candidate CARs at MOI = 20 (120 μl/well) in a round-bottomed 96-well plate (Corning). On the following day, 100 μl of culture medium was added to each well, and on day 3 the cells were transferred to a 48-well plate (final volume, 300 μl/well). On day 4, the cells were expanded by seeding at a density of 1 × 10
5- Parola C.
- Neumeier D.
- Friedensohn S.
- Csepregi L.
- Di Tacchio M.
- Mason D.M.
- Reddy S.T.
Antibody discovery and engineering by enhanced CRISPR–Cas9 integration of variable gene cassette libraries in mammalian cells.
cells/ml and dispensed into multiple wells of a 48-well plate. The cells were subsequently expanded in culture medium.
Assessment of CAR Jurkat cell activation via CD69 expression
CAR Jurkat cell lines, CHO-X, CHO–EpCAM and MCF-7 cells were washed once in culture medium and adjusted to a density of 1 × 10
6- Parthiban K.
- Perera R.L.
- Sattar M.
- Huang Y.
- Mayle S.
- Masters E.
- Griffiths D.
- Surade S.
- Leah R.
- Dyson M.R.
- McCafferty J.
A comprehensive search of functional sequence space using large mammalian display libraries created by gene editing.
cells/ml. CAR Jurkat cells were co-incubated with each target cell line at a ratio of 1:1 (200 μl/well, 96 round-bottomed plate) for 4 h at 37 °C. The cells were washed twice with MACS buffer and stained with anti-human CD69 FITC (BioLegend, catalog no. 310904) for 25 min at 4 °C. The cells were then washed three times with MACS buffer, followed by analysis of CD69 expression on an Attune
TM NxT flow cytometer (ThermoFisher Scientific) with accompanying software.
Article info
Publication history
Published online: October 30, 2020
Received in revised form:
October 27,
2020
Received:
June 30,
2020
Edited by Peter Cresswell
Footnotes
Author contributions—N. R., L. S., and R. C. conceptualization; N. R. and N. L.-A. data curation; N. R. and N. L.-A. formal analysis; N. R., T. P., T. D. J., L. S., and R. C. supervision; N. R., N. L.-A., S. A. G., H. K., Z. K., S. C., V. R. L., and R. P.-M. investigation; N. R., N. L.-A., S. A. G., R. P.-M., and AR methodology; N. R. and T. D. J. writing-original draft; N. R., T. P., T. D. J., and R. C. project administration; N. R., N. L.-A., T. D. J., L. S., and R. C. writing-review and editing; A. R. software; L. S. and R. C. resources; R. C. funding acquisition.
Funding and additional information—This work was supported by Innovate UK Grant 102859 and by OXGENE.
Conflict of interest—The authors are employees and shareholders of OXGENE, a biotech CRO company involved in the discovery and development of antibody-based therapeutics.
Abbreviations—The abbreviations used are: scFv
single-chain fragment variable
CHOChinese hamster ovary
EpCAMepithelial cell adhesion molecule
CAR-T cellchimeric antigen receptor T cell
HAhemagglutinin
MACSmagnetic activated cell sorting
PEphycoerythrin
MOImultiplicity of infection
PBMCperipheral blood mononuclear cell
NGSnext-generation sequencing
SPRsurface plasmon resonance.
Copyright
© 2020 Robertson et al.