US20120309016A1 - 3d adcc nk facs assay - Google Patents

3d adcc nk facs assay Download PDF

Info

Publication number
US20120309016A1
US20120309016A1 US13/577,309 US201113577309A US2012309016A1 US 20120309016 A1 US20120309016 A1 US 20120309016A1 US 201113577309 A US201113577309 A US 201113577309A US 2012309016 A1 US2012309016 A1 US 2012309016A1
Authority
US
United States
Prior art keywords
cells
cell
antibody
natural killer
well plate
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US13/577,309
Inventor
Andrea Challand
Christian Klein
Manfred Kubbies
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Hoffmann La Roche Inc
Original Assignee
Hoffmann La Roche Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Hoffmann La Roche Inc filed Critical Hoffmann La Roche Inc
Publication of US20120309016A1 publication Critical patent/US20120309016A1/en
Assigned to F. HOFFMANN-LA ROCHE AG reassignment F. HOFFMANN-LA ROCHE AG ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: KLEIN, CHRISTIAN, CHALLAND, ANDREA, KUBBIES, MANFRED
Assigned to HOFFMANN-LA ROCHE INC. reassignment HOFFMANN-LA ROCHE INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: F. HOFFMANN-LA ROCHE AG
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/02Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving viable microorganisms
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • G01N33/5014Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics for testing toxicity
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0634Cells from the blood or the immune system
    • C12N5/0636T lymphocytes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0693Tumour cells; Cancer cells
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/483Physical analysis of biological material
    • G01N33/487Physical analysis of biological material of liquid biological material
    • G01N33/49Blood
    • G01N33/4915Blood using flow cells
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • G01N33/5014Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics for testing toxicity
    • G01N33/5017Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics for testing toxicity for testing neoplastic activity
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • G01N33/5044Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics involving specific cell types
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • G01N33/5044Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics involving specific cell types
    • G01N33/5047Cells of the immune system
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor

Definitions

  • Monolayer cultures of established tumor cell lines are frequently used in basic tumor biology research and anti-tumor drug development.
  • a two-dimensional, flat culture model insufficiently reflects the three-dimensional (3D) tumor architecture. Therefore, specific aspects related to the in vivo development of solute diffusion gradients can only be studied in a three-dimensional culture system like for instance the multicellular tumor spheroid or aggregate model.
  • Tumor spheroids or aggregates mimic avascular tumor regions, characterized by limited nutrient supply due to diffusion barriers through multicellular layers.
  • a first aspect as reported herein is a method for in vitro detection of the effector function of an antibody comprising the step of incubating a three-dimensional spheroid or aggregate comprising tumor cells and natural killer cells with the immunoglobulin.
  • the natural killer cells are human natural killer cells and have a purity of 90% or more.
  • the natural killer cells and tumor target cells are mixed at a ratio of from 10:1 to 1:10.
  • the ratio is of from 1:3 to 1:10.
  • the ratio is of from 1:2 to 1:4.
  • the centrifuging is for 10 min. at 100 to 1,000 rpm.
  • the centrifuging is at about 1,000 rpm.
  • the second fluorescent dye is propidium iodide.
  • the incubating is for about 20 hours to about 28 hours.
  • the tumor cells are lymphoma cells.
  • the lymphoma cell is selected from the group comprising Raji-cells, SU-DHL4 cells, and Z138 cells.
  • the antibody is added at a final concentration in the well of from 100 ⁇ g/ml to 0.001 ⁇ g/ml. In a further embodiment the antibody is added at a final concentration in the well of from 20 ⁇ g/ml to 0.1 ⁇ g/ml. In one embodiment the antibody is added at a final concentration in the well of from 8 ⁇ g/ml to 12 ⁇ g/ml.
  • a further aspect as reported herein is the use of a three-dimensional spheroid or aggregate comprising tumor cells and natural killer cells for the high-throughput analysis of the combination of a multitude of antibodies and a multitude of tumor cells.
  • kits comprising:
  • the multi well plate is a 96-well multi well plate.
  • a cell analytical technology based on the use of a three-dimensional spheroid or aggregate co-culture assay, wherein the spheroids or aggregates comprise tumor cells and natural killer cells.
  • This assay is useful in one embodiment for the in vitro functional analysis of immunoglobulins in single and high-throughput format.
  • a single three-dimensional spheroid or aggregate is placed in each well of a 96-well round bottom multi well plate that has been coated with polyHEMA (poly (hydroxyethyl methacrylic) acid).
  • the NK cells are normal diploid human natural killer (NK) cells.
  • the NK cells have been selected by applying a negative selection technique, i.e.
  • the cells are not touched during the selection step (see e.g. Horgan, K. et al., Curr. Prot. Immunol. (2009), Chapter 7, Unit 7.4. Immunomagnetic purification of T cell subpopulations, and Neurauter, A.A., et al., Adv. Biochem. Eng. Biotechnol. 106 (2007) 41-73). It has been found that it is possible with these NK cells to quantitate correct percentages of viable and dead cells.
  • the method for the generation of three dimensional spheroids from aggregates comprises the addition of reconstituted basement matrix derived from the Engelbreth-Holm-Swarm murine tumor (rBM, MatrigelTM), a proteinaceous gel containing extracellular matrix components such as collagens, laminin, fibronectin, entactin (nidogen), and proteoglycans, to the cultivation medium.
  • the three-dimensional architecture allows the co-cultivation of tumor cells with fibroblast, immune and endothelial cells, enabling the investigation of tumor/stroma interaction effects in vitro (Friedrich, J., et al., Int. J. Radiat. Biol. 83 (2007) 849-871).
  • spheroids or aggregates in a high-throughput fashion in research requires a standardized protocol that rapidly generates spheroids of homogenous size with similar diffusion gradients and cell physiology in a multi-well plate format that is easily accessible for subsequent biochemical or cell analysis. Moreover such a protocol should be applicable to a large variety of tumor cell lines.
  • one aspect as reported herein is an assay for detection of effector function of an antibody comprising
  • a sensitive antibody-dependent-cellular-cytotoxicity assay can be provided.
  • the detection of effector function of an antibody is a detection or determination of antibody-dependent-cellular-cytotoxicity of an antibody.
  • a three-dimensional assay set-up of tumor and immuno effector function cells is also advantageous.
  • the methods as reported herein are in vitro methods.
  • the mixing of the tumor cells and the natural killer cells results in the formation of a three-dimensional spheroid.
  • FIGS. 1 and 2 the distribution of viable and dead cells analyzed by FACS is shown.
  • FIG. 1 a shows the distribution of viable and dead Raji-cells labeled with CMFDA determined by FACS analysis. Viable Raji-cells are located in the lower right sector of the FACS diagram.
  • FIGS. 1 b and 1 c show the distribution of viable and dead cells of a co-culture of Raji-cells and natural killer cells in the absence of an added antibody. Viable natural killer cells are located in the lower left sector of the FACS diagram, viable Raji-cells are located in the lower right sector of the FACS diagram, dead natural killer cells are located in the upper left sector of the FACS diagram and dead Raji-cells are located in the upper right sector of the FACS diagram. From FIGS.
  • FIG. 2 a shows the FACS analysis of viable and dead Raji-cells after incubation with an antibody.
  • the fraction of dead cells increases due to the direct cell death inducing function of the antibody.
  • the number of dead Raji cells increases even more due to the ADCC effector function (upper right clusters: FIG. 2 b Raji/NK ratio 1:1 and FIG. 2 c Raji/NK ratio 1:10). It can be seen that by the addition of natural killer cells the sensitivity of the assay can be increased.
  • the multi well plates are centrifuged in one embodiment for 10 min. at 1,000 g. During centrifugation all cells within each well are pelleted at the bottom of the well. This ensures equal cell numbers for initiation of the formation of a single spheroid or aggregate in each well.
  • the assay comprises the formation of the lymphoma-NK three-dimensional spheroid or aggregates prior to the addition of the antibody to be tested.
  • the assay as reported herein can be performed with any tumor (target) cell.
  • the tumor cell is a lymphoma cell.
  • the lymphoma cell is selected from Raji-cell, SU-DHL4 cell, and Z138 cell.
  • the formation of the three-dimensional spheroid is performed in the presence of liquid reconstituted basement membrane (rBM).
  • rBM liquid reconstituted basement membrane
  • a concentration of 2.5% rBM (v/v) is used.
  • all cells were incorporated in one distinct spheroid with a round geometry. Formation and compaction was completed after 24 hours of culture time. Therefore, in one embodiment the incubation is for 20 hours to 28 hours.
  • Lower concentrations of rBM did not ensure the incorporation of all cells into the spheroid, and higher concentrations impaired the round geometry of the spheroids. After the 10 min. centrifugation step, all cells within a well are incorporated into one flat pellet.
  • the tumor cell is a Raji-cell and rBM is absent in all steps of the method.
  • spheroid size was analyzed after a 24 hour culture period. All spheroids are regular in shape, display a uniform round geometry, and exhibit a narrow size variation.
  • the assay as reported herein is performed with different lymphoma tumor (target) cell lines and with different antibody concentrations. It can be seen that the assay as reported herein can be performed with different lymphoma cell lines at the same efficiency. It can further be seen that the assay can be performed at an antibody concentration of from 10 ⁇ g/ml to 0.1 ⁇ g/ml. Therefore, in one embodiment the assay as reported herein comprises adding the antibody at a concentration of from 0.1 ⁇ g/ml to 15 ⁇ g/ml, in a further embodiment of from 8 ⁇ g/ml to 12 ⁇ g/ml.
  • FIG. 5 the sensitivity of the assay as reported herein depending on the ratio of lymphoma cells to natural killer cells is shown. It can be seen that a ratio of from 1:1 to 1:10 of lymphoma (target) cells to natural killer cells indicates the ADCC effector function of NK cells. Therefore, in one embodiment the ratio of lymphoma cell to natural killer cell is of from 1:1 to 1:10, in another embodiment of from 1:3 to 1:10, in a further embodiment of from 1:2 to 1:4.
  • single three-dimensional spheroids or aggregates with a narrow size distribution and homogenous spherical geometry can be generated in a single well or in multiple wells of a multi-well plate in parallel within a 24 hour culture period. It has been shown that this can be a standardized culture format with easy access for compound handling and spheroid harvest for subsequent analysis.
  • the almost uniform size and geometry of the spheroids or aggregates guarantees the development of similar diffusion gradients in each spheroid. Therefore, one aspect as reported herein is an automated or high-throughput assay comprising the assay as outlined above.
  • the spheroid generation protocol includes the addition of a murine basement membrane extract (rBM), a mixture of extracellular matrix proteins that induces a compaction of the aggregate to a spheroid.
  • rBM murine basement membrane extract
  • FIG. 6 an exemplary scheme for the generation of three-dimensional spheroids is shown.
  • the natural killer cells are human natural killer cells.
  • the natural killer cells and tumor cells are mixed at a ratio of from 10:1 to 1:10. In a further embodiment the ratio is of from 1:2 to 1:4.
  • the incubating is for about 20 hours to about 28 hours.
  • the centrifuging is at 1,000 rpm for 10 min.
  • the tumor cell is a lymphoma cell.
  • the lymphoma cell is a Raji-cell, or a SU-DHL4 cell, or a Z138 cell.
  • the antibody is added at a concentration of from 15 ⁇ g/ml to 0.1 ⁇ g/ml. In another embodiment the antibody is added at a concentration of from 8 ⁇ g/ml to 12 ⁇ g/ml.
  • Another aspect as reported herein is the use of a three-dimensional spheroid or aggregate comprising tumor cells and natural killer cells for the determination of effector function of a combination of a multitude of antibodies with a multitude of tumor cells.
  • FIG. 1 FACS analysis of viable and dead Raji-cells and natural killer (NK) cells in the absence of an antibody. Quadrants: lower left: viable NK cells, upper left: dead NK cells, lower right: viable Raji-cells, upper right: dead Raji-cells. Panel A: Raji-cells only, panel B: Raji-cells and NK cells at a ratio of 1:1, panel C: Raji-cells and NK-cells 1:10.
  • FIG. 2 FACS analysis of viable and dead Raji-cells and natural killer cells in the presence of an added anti-CD20 antibody (10 ⁇ g/ml). Quadrants: lower left: viable NK cells, upper left: dead NK cells, lower right: viable Raji-cells, upper right: dead Raji-cells. Panel A: Raji-cells only, panel B: Raji-cells and NK cells at a ratio of 1:1, panel C: Raji-cells and NK cells at a ratio of 1:10.
  • FIG. 3 Optimization of lymphoma/spheroid-aggregate co-culture by variation of lymphoma cell, NK cell and antibody application schedule.
  • Lymphoma cells Raji-cells.
  • FIG. 4 Percentages of viable lymphoma cells in the presence of NK cells as a function of the anti-CD20 antibody concentration.
  • Panel A Raji-cells
  • panel B SU-DHL4 cells
  • panel C Z138 cells.
  • E:T ratio NK cell to lymphoma cell ratio
  • FIG. 5 Percentages of viable lymphoma cells in the presence of the anti-CD20 antibody concentration as a function of the effector (NK):target (lymphoma) ratio.
  • Panel A Raji-cells
  • panel B SU-DHL4 cells
  • panel C Z138 cells.
  • Anti-CD20 antibody concentration 10 ⁇ g/ml.
  • FIG. 6 Schematic exemplary method.
  • FIG. 7 Microscopic images of Raji-cells only and Raji cell co-cultivated with purified NK cells.
  • Raji-cells, SU-DHL4 cells and Z138 cell lines were obtained from ATCC (Manassas, Va., USA), from DSMZ (Braunschweig, Germany) and from Prof. M. Dyer (University of Leicester, UK), respectively.
  • Raji-cells and SU-DHL4 cells were cultivated in RPMI 1640 medium (PAN Biotech, Cat. no. PO4-18500) and Z138 in DMEM medium (PAN Biotech, Cat. no. PO4-02500) supplemented with 10% FCS (Gibco, Cat. no. 10500-064) and Pen/Strep (Roche, Cat. no. 11 074 440 001) at 37° C. in a humidified incubator. Exponential growing cells with cell viability of 90% or more were used for the NK cell co-cultivation experiments.
  • PBMC peripheral blood mononuclear cells
  • PBMC peripheral blood mononuclear cells
  • the purity of the NK cell preparation was determined by staining of an aliquot of the MACS eluate. In short about 2 ⁇ 10 5 cells were resuspended in 100 ⁇ l RPMI 1640/10% FCS and stained with 10 ⁇ l each of anti-CD56-PE and anti-CD3-FITC antibodies (Becton Dickinson, Cat. no. 555516 and 555339, respectively) for 15 min at 4° C. Thereafter, 2 ml of RPMI 1640/10% FCS were added to the cells which were centrifuged for 5 min. at 400 g.
  • the pellet was resuspended in 0.5 ml RPMI 1640/10% FCS and the percentage of the CD56 positive but CD3 negative cell fraction within the lymphocyte scatter gate was analyzed using a FACS Scan or FACS Canto II instrument (Becton Dickinson).
  • PBMC peripheral blood mononuclear cells
  • the CMFDA lyophilizate (Invitrogen Cat. no. C7025) was resuspended in DMSO to obtain a 10 mM stock solution. 1 ⁇ 10 6 lymphoma cells were incubated for 30 min. at 37° C. in 1 ml complete medium supplemented with 1 ⁇ M CMFDA. Thereafter, cells were pelleted, washed once in complete medium and resuspended finally in complete medium at 1 ⁇ 10 6 cells/ml.
  • the lymphoma cell number was determined using a CASY instrument (Schärfe-Systems, Reutlingen) and the cell suspension was diluted in ice cold medium to 2.5 ⁇ 10 4 cells/ml (for 5,000 cells per spheroid/aggregate) and 5 ⁇ 10 4 cells/ml (for 10,000 cells per spheroid/aggregate).
  • a volume of 200 ⁇ l of the cell suspension was added to each well of a 96-well plate with round (Corning Inc., New York, USA) or conical (Nunc, Roskilde, The Netherlands) bottom.
  • the plates were pre-coated with 50 ⁇ l 0.5% polyHEMA (Polysciences, Eppelheim, Germany) in 95% ethanol (v/v) and air dried at 37° C. for three days.
  • the spheroid formation was initiated by centrifugation of the plates at 1,000 g for 10 min. using an Eppendorf 5810 centrifuge (Eppendorf AG, Hamburg, Germany) with swinging buckets.
  • the plates were incubated under standard cell culture conditions at 37 ° C. and 7% CO 2 in humidified incubators.
  • Monolayer cells were detached with Accutase (PAA Laboratories GmbH, Innsbruck, Austria) to generate a single cell suspension.
  • the cell number was determined using a CASY instrument (Schärfe-Systems, Reutlingen) and the cell suspension was diluted in ice cold medium to 2.5 ⁇ 10 4 cells/ml (for 5,000 cells per spheroid/aggregate) and 5 ⁇ 10 4 cells/ml (for 10,000 cells per spheroid/aggregate).
  • the rBM was thawed on ice overnight and added at a final concentration of 2.5% (v/v) with ice cold pipette tips to the cell suspension.
  • a volume of 200 ⁇ l of the cell suspension was added to each well of a 96-well plate with round (Corning Inc., New York, USA) or conical (Nunc, Roskilde, The Netherlands) bottom.
  • the plates were pre-coated with 50 ⁇ l 0.5% polyHEMA (Polysciences, Eppelheim, Germany) in 95% ethanol and air dried at 37° C. for three days.
  • the spheroid formation was initiated by centrifugation of the plates at 1,000 g for 10 min. using an Eppendorf 5810 centrifuge (Eppendorf AG, Hamburg, Germany) with swinging buckets.
  • the plates were incubated under standard cell culture conditions at 37° C. and 7% CO 2 in humidified incubators.
  • lymphoma cells CMFDA labeled
  • NK cells NK cells mixed at ratios as indicated in 6 well plates.
  • E:T NK cell to lymphoma cell
  • NK cells NK cell to lymphoma cell
  • 200 ⁇ l of the cell suspension was added to a single well of a polyHEMA coated 96 well V-plate (Nunc, Cat. no. 249662).
  • PolyHEMA coating 50 ⁇ l 0.5% polyHEMA in 95% ethanol per well; drying for 72 h at 37° C. (Polysciences, Cat. No. 18894).
  • the plates were centrifuged for 10 min. at 1,000 g.
  • the antibodies were added thereafter at concentrations as indicated above, and cell aggregates/spheroids were incubated at 37° C., 7% CO 2 in a humidified incubator.
  • Microscopic images of Raji-cells only and Raji cell co-cultivated with purified NK cells are shown in FIG. 7 , as well as images of co-cultivated Raji and monocyte cells to illustrate 3D co-cultivation of tumor cells with other immune cells then NK cells.
  • Spheroids/aggregates were generated using 10,000 cells and incubated with the antibody as outlined in Examples 2 and 3.
  • the identification of viable lymphoma tumor cells was as follows: Individual aggregates from individual wells representing identical experimental conditions were pooled, dissociated by pipetting and centrifuged at 300 g for 10 min. Individual spheroids were pooled, washed once with phosphate buffered saline (PBS), resuspended in Accutase solution, and incubated at 37° C. Every five minutes, the spheroids/aggregates were resuspended by pipetting and dissociation was complete within 5 to 15 min.
  • PBS phosphate buffered saline
  • Viable lymphoma cells were identified as shown in FIG. 1 b.
  • the upper right quadrant of PI and CMFDA positive cells represent the dead lymphoma cells, and the lower right quadrant of PI negative but CMFDA positive cells represent the viable lymphoma tumor target cell fraction.
  • the lower left quadrant are viable NK cells, whereas dead NK cells are located in the upper left quadrant.
  • an apoptosis assay can be performed.
  • Spheroids/aggregates were generated using 10,000 cells and incubated with the antibody as outlined in Examples 2 and 3.
  • the spheroids/aggregates were transferred into a 96-well conical-bottom plate, washed once with phosphate buffered saline (PBS), resuspended in Accutase solution, and incubated at 37° C. Every five minutes, the spheroids/aggregates were resuspended by pipetting and dissociation was complete within 5 to 15 min.
  • PBS phosphate buffered saline
  • the single cell suspensions from eight spheroids/aggregates were pooled and cells were stained with annexin-V-fluos and propidium iodide in the presence of supplemented 2 mM CaCl 2 , (annexin-V-fluos staining kit, Roche Diagnostics GmbH, Mannheim, Germany).
  • the fluorescence of 10,000 cells was acquired using a flow cytometer (FACS scan instrument, Becton Dickinson, San Jose, Ca., USA). Quadrant statistics was applied on the dot plots, with the number of viable cells located in the lower-left quadrant.
  • the number of total cells from the spheroids/aggregates were counted using a Fuchs-Rosenthal cell counting chamber and multiplied with the percentage of viable or dead cells of the same spheroids/aggregates as determined from the annexin-V-fluos/PI staining

Abstract

Herein is reported a cell analytical technology based on a three-dimensional spheroid/aggregate co-culture assay, wherein the spheroid or aggregate is formed of tumor and natural killer cells. This method is useful for the in vitro functional analysis of antibodies in single and high-throughput format.

Description

  • Herein is reported a novel antibody-dependent-cytotoxicity-FACS-assay based on a three-dimensional spheroid or an aggregate formed of lymphoma cells and natural killer cells. This assay is useful for the in vitro functional analysis of therapeutic immunoglobulins in single as well as high-throughput format.
  • BACKGROUND OF THE INVENTION
  • Monolayer cultures of established tumor cell lines are frequently used in basic tumor biology research and anti-tumor drug development. However, a two-dimensional, flat culture model insufficiently reflects the three-dimensional (3D) tumor architecture. Therefore, specific aspects related to the in vivo development of solute diffusion gradients can only be studied in a three-dimensional culture system like for instance the multicellular tumor spheroid or aggregate model. Tumor spheroids or aggregates mimic avascular tumor regions, characterized by limited nutrient supply due to diffusion barriers through multicellular layers.
  • However, the widespread use of 3D cultures in research is limited by inconvenient generation and handling. Therefore, a simple and rapid method was developed to generate single spheroids or aggregates in suspension culture in a high-throughput fashion. Single spheroids or aggregates with equal sizes and homogenous spherical geometry can be generated in single wells of a 96-well plate within a 24 hour culture period. It is a standardized culture format with easy access for compound handling and spheroid harvest for subsequent analysis. The uniform size and geometry guarantees the development of almost identical diffusion gradients in each spheroid or aggregate (Ivascu, A. and Kubbies, M., J. Biomol. Screening 11 (2006) 922-932). The known spheroid generation protocol includes the addition of a murine basement membrane extract (rBM), a mixture of extracellular matrix proteins that induces a compaction of the aggregate to a spheroid.
  • Inami, K., et al. report antitumor activity of anti-C-ERC/mesothelin monoclonal antibody in vivo (Cancer Sci. 101 (2010) 969-974).
  • SUMMARY OF THE INVENTION
  • It has been found that with the combination of tumor cells and natural killer cells in a three-dimensional spheroid or aggregate the evaluation of immunoglobulins can on the one hand be made more in vivo like and on the other hand is now suited for high-throughput analysis.
  • A first aspect as reported herein is a method for in vitro detection of the effector function of an antibody comprising the step of incubating a three-dimensional spheroid or aggregate comprising tumor cells and natural killer cells with the immunoglobulin.
  • In one embodiment the method comprises the following steps:
      • a) labeling tumor target cells with a first fluorescent dye,
      • b) mixing natural killer cells and tumor target cells,
      • c) adding about 104 cells per 200 μl to a well of a multi well plate,
      • d) centrifuging the multi-well plate and thereby initiating the forming of a three-dimensional cell spheroid,
      • e) adding the immunoglobulin to the wells of the multi well plate,
      • f) incubating the multi-well plate for about 20 hours to about 72 hours,
      • g) labeling dead cells in the wells with a second fluorescent dye, and
      • i) analyzing the cells in the wells of the multi well plate by fluorescence activated cell sorting (FACS) and thereby detecting the effector function of the antibody.
  • In one embodiment the natural killer cells are human natural killer cells and have a purity of 90% or more. In a further embodiment the natural killer cells and tumor target cells are mixed at a ratio of from 10:1 to 1:10. In a further embodiment the ratio is of from 1:3 to 1:10. In another embodiment the ratio is of from 1:2 to 1:4. In one embodiment the centrifuging is for 10 min. at 100 to 1,000 rpm. In a further embodiment the centrifuging is at about 1,000 rpm. In one embodiment the second fluorescent dye is propidium iodide. In one embodiment the incubating is for about 20 hours to about 28 hours.
  • In one embodiment the tumor cells are lymphoma cells. In another embodiment the lymphoma cell is selected from the group comprising Raji-cells, SU-DHL4 cells, and Z138 cells. In another embodiment the antibody is added at a final concentration in the well of from 100 μg/ml to 0.001 μg/ml. In a further embodiment the antibody is added at a final concentration in the well of from 20 μg/ml to 0.1 μg/ml. In one embodiment the antibody is added at a final concentration in the well of from 8 μg/ml to 12 μg/ml.
  • A further aspect as reported herein is the use of a three-dimensional spheroid or aggregate comprising tumor cells and natural killer cells for the high-throughput analysis of the combination of a multitude of antibodies and a multitude of tumor cells.
  • Another aspect as reported herein is a method for determining in vitro an antibody with effector function comprising
      • a) providing at least one antibody,
      • b) labeling tumor cells with a first fluorescent dye,
      • c) mixing natural killer cells and tumor target cells,
      • d) adding about 104 cells per 200 μl to the wells of a multi well plate,
      • e) centrifuging the multi-well plate and thereby initiating the forming of a three-dimensional cell spheroid,
      • f) adding each of the provided antibodies to an individual well of the multi well plate,
      • g) incubating the multi-well plate for about 20 hours to about 72 hours
      • h) labeling dead cells in each of the incubated wells with a second fluorescent dye,
      • i) analyzing each well of the multi well plate by fluorescence activated cell sorting, and
      • j) determining the antibody with the highest ratio or a ratio of more than 1 of dead cells to viable cells as antibody with effector function.
  • Also an aspect as reported herein is a kit comprising:
      • a) a tumor cell labeled with a fluorescent dye,
      • b) isolated natural killer cells,
      • c) a 96-well multi well plate, and
      • d) propidium iodide.
  • In one embodiment the multi well plate is a 96-well multi well plate.
  • DETAILED DESCRIPTION OF THE INVENTION
  • Herein is reported a cell analytical technology based on the use of a three-dimensional spheroid or aggregate co-culture assay, wherein the spheroids or aggregates comprise tumor cells and natural killer cells. This assay is useful in one embodiment for the in vitro functional analysis of immunoglobulins in single and high-throughput format. In one embodiment a single three-dimensional spheroid or aggregate is placed in each well of a 96-well round bottom multi well plate that has been coated with polyHEMA (poly (hydroxyethyl methacrylic) acid). In a further embodiment the NK cells are normal diploid human natural killer (NK) cells. In one embodiment the NK cells have been selected by applying a negative selection technique, i.e. the cells are not touched during the selection step (see e.g. Horgan, K. et al., Curr. Prot. Immunol. (2009), Chapter 7, Unit 7.4. Immunomagnetic purification of T cell subpopulations, and Neurauter, A.A., et al., Adv. Biochem. Eng. Biotechnol. 106 (2007) 41-73). It has been found that it is possible with these NK cells to quantitate correct percentages of viable and dead cells.
  • Most of the in vitro experiments in the field of tumor biology are performed with monolayer cultures since these are easy and convenient to handle. However, although they provide a valuable model to study distinct functions, monolayer cultures insufficiently reflect the tumor pathobiology due to the lack of stroma components, extracellular matrix and fundamental geometric differences between two-dimensional (2D) cultures and three-dimensional (3D) solid tumors. The three-dimensional organization of cells provides a complex network of cell-cell and cell-matrix interactions relevant e.g. for distribution and function of hormones, growth factors and nutrients influencing cellular differentiation, proliferation and survival.
  • In one embodiment the method for the generation of three dimensional spheroids from aggregates comprises the addition of reconstituted basement matrix derived from the Engelbreth-Holm-Swarm murine tumor (rBM, Matrigel™), a proteinaceous gel containing extracellular matrix components such as collagens, laminin, fibronectin, entactin (nidogen), and proteoglycans, to the cultivation medium. The three-dimensional architecture allows the co-cultivation of tumor cells with fibroblast, immune and endothelial cells, enabling the investigation of tumor/stroma interaction effects in vitro (Friedrich, J., et al., Int. J. Radiat. Biol. 83 (2007) 849-871).
  • In the spinner (Sutherland, R. M. and Durand, R. E., Recent Results Cancer Res. 95 (1984) 24-49) and the gyratory rotation technique (Moscona, A., Exp. Cell Res. 22 (1961) 455-475) trypsinized cells are placed in a culture vessel with a magnetic stirrer inhibiting cell attachment to the substrate and favoring cell-cell adhesion. In a more recently developed technique, spheroids are grown in a hanging drop of an inverted microplate (Kelm, J. M., et al., Biotechnol. Bioeng. 83 (2003) 173-180). However, all these methods are limited by either long cultivation time, formation of unequal-size spheroids, or difficult mechanical accessibility. In addition, in suspension cultures, many tumor cell lines grow poorly in three dimensional compact spheroids (Mueller-Klieser, W., Crit. Rev. Oncol. Hematol. 36 (2000) 123-139).
  • The use of spheroids or aggregates in a high-throughput fashion in research requires a standardized protocol that rapidly generates spheroids of homogenous size with similar diffusion gradients and cell physiology in a multi-well plate format that is easily accessible for subsequent biochemical or cell analysis. Moreover such a protocol should be applicable to a large variety of tumor cell lines.
  • It has been found that this need can be fulfilled with the methods as reported herein. Therefore, one aspect as reported herein is an assay for detection of effector function of an antibody comprising
      • a) labeling lymphoma (target) cells with the green fluorophore CMFDA (5-chloromethylfluorescein diacetate),
      • b) isolating human normal natural killer (NK) cells from human blood, in one embodiment with a purity of more than 90%,
      • c) mixing NK and lymphoma target cells at a ratio of from 1:10 to 10:1,
      • d) adding about 104 cells per 200 μl to some or all of the wells of a multi well plate,
      • e) centrifuging the multi-well plate,
      • f) adding the immunoglobulin of interest to the wells of the multi well plate,
      • g) incubating the multi-well plate for up to 72 h, in one embodiment for 20 h to 72 h,
      • h) adding propidium iodide to the wells, and
      • i) analyzing the cells in the wells of the multi well plate by FACS.
  • It has been found that by the combination of lymphoma cells and natural killer cells a sensitive antibody-dependent-cellular-cytotoxicity assay can be provided. In one embodiment the detection of effector function of an antibody is a detection or determination of antibody-dependent-cellular-cytotoxicity of an antibody. Additionally a three-dimensional assay set-up of tumor and immuno effector function cells is also advantageous. In one embodiment the methods as reported herein are in vitro methods. In another embodiment the mixing of the tumor cells and the natural killer cells results in the formation of a three-dimensional spheroid.
  • In FIGS. 1 and 2 the distribution of viable and dead cells analyzed by FACS is shown. FIG. 1 a shows the distribution of viable and dead Raji-cells labeled with CMFDA determined by FACS analysis. Viable Raji-cells are located in the lower right sector of the FACS diagram. FIGS. 1 b and 1 c show the distribution of viable and dead cells of a co-culture of Raji-cells and natural killer cells in the absence of an added antibody. Viable natural killer cells are located in the lower left sector of the FACS diagram, viable Raji-cells are located in the lower right sector of the FACS diagram, dead natural killer cells are located in the upper left sector of the FACS diagram and dead Raji-cells are located in the upper right sector of the FACS diagram. From FIGS. 1 b (ratio of Raji to NK cells of 1:1) and 1 c (ratio of Raji to NK cells of 1:10) it can be seen that in the absence of antibody and independent of the ratio of Raji-cells to natural killer cells the percentage of the respective viable and dead lymphoma cells is not changed significantly.
  • FIG. 2 a shows the FACS analysis of viable and dead Raji-cells after incubation with an antibody. In comparison with the FACS diagram of FIG. 1 a it can be seen that by incubating the Raji-cells with the antibody only, the fraction of dead cells increases due to the direct cell death inducing function of the antibody. In the presence of NK cells the number of dead Raji cells increases even more due to the ADCC effector function (upper right clusters: FIG. 2 b Raji/NK ratio 1:1 and FIG. 2 c Raji/NK ratio 1:10). It can be seen that by the addition of natural killer cells the sensitivity of the assay can be increased.
  • In the assay the multi well plates are centrifuged in one embodiment for 10 min. at 1,000 g. During centrifugation all cells within each well are pelleted at the bottom of the well. This ensures equal cell numbers for initiation of the formation of a single spheroid or aggregate in each well.
  • In FIG. 3 the effect of the sequence of the addition of the individual components, i.e. Raji-cells, natural killer cells and antibody, on the assay is shown. It can be seen that the addition of all three components in parallel gave rise to slightly, but not significantly, higher cell death rates. However, to mimic more closely the in vivo situation, in one embodiment the assay comprises the formation of the lymphoma-NK three-dimensional spheroid or aggregates prior to the addition of the antibody to be tested.
  • The assay as reported herein can be performed with any tumor (target) cell. In one embodiment the tumor cell is a lymphoma cell. In a further embodiment the lymphoma cell is selected from Raji-cell, SU-DHL4 cell, and Z138 cell.
  • In one embodiment when using adherently growing carcinoma or sarcoma tumor cells as tumor cells the formation of the three-dimensional spheroid is performed in the presence of liquid reconstituted basement membrane (rBM). In one embodiment a concentration of 2.5% rBM (v/v) is used. In this embodiment all cells were incorporated in one distinct spheroid with a round geometry. Formation and compaction was completed after 24 hours of culture time. Therefore, in one embodiment the incubation is for 20 hours to 28 hours. Lower concentrations of rBM did not ensure the incorporation of all cells into the spheroid, and higher concentrations impaired the round geometry of the spheroids. After the 10 min. centrifugation step, all cells within a well are incorporated into one flat pellet. Three hours later, some degree of compaction becomes evident in the presence and absence of rBM. Without rBM, no further tightening of the aggregates can be observed after 6 hours and 24 hours. In one embodiment the tumor cell is a Raji-cell and rBM is absent in all steps of the method.
  • In one embodiment five thousand cells were centrifuged in RPMI 1640 with 10% FCS (fetal calf serum) and 2.5% rBM (v/v). The spheroid size was analyzed after a 24 hour culture period. All spheroids are regular in shape, display a uniform round geometry, and exhibit a narrow size variation.
  • In FIG. 4 the assay as reported herein is performed with different lymphoma tumor (target) cell lines and with different antibody concentrations. It can be seen that the assay as reported herein can be performed with different lymphoma cell lines at the same efficiency. It can further be seen that the assay can be performed at an antibody concentration of from 10 μg/ml to 0.1 μg/ml. Therefore, in one embodiment the assay as reported herein comprises adding the antibody at a concentration of from 0.1 μg/ml to 15 μg/ml, in a further embodiment of from 8 μg/ml to 12 μg/ml.
  • In FIG. 5 the sensitivity of the assay as reported herein depending on the ratio of lymphoma cells to natural killer cells is shown. It can be seen that a ratio of from 1:1 to 1:10 of lymphoma (target) cells to natural killer cells indicates the ADCC effector function of NK cells. Therefore, in one embodiment the ratio of lymphoma cell to natural killer cell is of from 1:1 to 1:10, in another embodiment of from 1:3 to 1:10, in a further embodiment of from 1:2 to 1:4.
  • With the assay as reported herein single three-dimensional spheroids or aggregates with a narrow size distribution and homogenous spherical geometry can be generated in a single well or in multiple wells of a multi-well plate in parallel within a 24 hour culture period. It has been shown that this can be a standardized culture format with easy access for compound handling and spheroid harvest for subsequent analysis. The almost uniform size and geometry of the spheroids or aggregates guarantees the development of similar diffusion gradients in each spheroid. Therefore, one aspect as reported herein is an automated or high-throughput assay comprising the assay as outlined above. The spheroid generation protocol includes the addition of a murine basement membrane extract (rBM), a mixture of extracellular matrix proteins that induces a compaction of the aggregate to a spheroid.
  • In FIG. 6 an exemplary scheme for the generation of three-dimensional spheroids is shown.
  • Herein is reported a method for the in vitro detection of effector function of an antibody comprising the incubating of a three-dimensional spheroid or aggregate comprising tumor cells and natural killer cells with the antibody.
  • In one embodiment the method comprising the following steps:
      • mixing natural killer cells and tumor cells,
      • adding about 104 cells per 200 μl to the wells of a multi well plate,
      • centrifuging the multi-well plate and thereby inducing the formation of a three-dimensional spheroid or aggregate,
      • adding the immunoglobulin to the wells of the multi well plate,
      • incubating the multi-well plate for about 20 hours to about 72 hours, and
      • analyzing the cells in the wells of the multi well plate by fluorescence activated cell sorting and thereby detecting the effector function of the antibody.
  • In a further embodiment the method comprises in addition the following step as first step:
      • labeling tumor cells with a first fluorescent dye.
  • In another embodiment the method comprises the further steps:
      • labeling dead cells with a second fluorescent dye, and
      • analyzing the cells in the wells of the multi well plate by fluorescence activated cell sorting and thereby detecting the effector function of the antibody.
  • In one embodiment the natural killer cells are human natural killer cells. In also an embodiment the natural killer cells and tumor cells are mixed at a ratio of from 10:1 to 1:10. In a further embodiment the ratio is of from 1:2 to 1:4.
  • In one embodiment the incubating is for about 20 hours to about 28 hours.
  • In one embodiment the centrifuging is at 1,000 rpm for 10 min.
  • In one embodiment the tumor cell is a lymphoma cell. In a further embodiment the lymphoma cell is a Raji-cell, or a SU-DHL4 cell, or a Z138 cell.
  • In one embodiment the antibody is added at a concentration of from 15 μg/ml to 0.1 μg/ml. In another embodiment the antibody is added at a concentration of from 8 μg/ml to 12 μg/ml.
  • Another aspect as reported herein is the use of a three-dimensional spheroid or aggregate comprising tumor cells and natural killer cells for the determination of effector function of a combination of a multitude of antibodies with a multitude of tumor cells.
  • A further aspect as reported herein is a method for determining in vitro an antibody with effector function comprising the steps:
      • mixing natural killer cells and the tumor cells,
      • adding about 104 cells per 200 μl to the wells of a multi well plate,
      • centrifuging the multi-well plate and thereby inducing the formation of a three-dimensional spheroid or aggregate,
      • adding the antibody to an individual well of the multi well plate,
      • incubating the multi-well plate for about 20 hours to about 72 hours, and
      • determining the antibody with a ratio of dead to viable cells as antibody with effector function.
  • In one embodiment the method comprises in addition the following first steps:
      • providing at least one antibody, and
      • labeling tumor cells with a first fluorescent dye.
  • In another embodiment the method comprises the steps:
      • adding each of the provided antibodies to an individual well of the multi well plate, whereby to each well at most one antibody is added,
      • labeling dead cells in each of the incubated wells with a second fluorescent dye,
      • analyzing the cells in each well of the multi well plate by fluorescence activated cell sorting, and
      • determining the antibody with the highest ratio of dead to viable cells as antibody with effector function.
  • The assay and method as reported herein is exemplified with an anti-CD20 antibody as reported in WO 2005/044859 (incorporated by reference herein). This antibody has been chosen only for exemplifying the current invention and should not be interpreted as restriction. The scope of the invention is set forth in the claims.
  • The following examples and figures are provided to aid the understanding of the present invention, the true scope of which is set forth in the appended claims. It is understood that modifications can be made in the procedures set forth without departing from the spirit of the invention.
  • DESCRIPTION OF THE FIGURES
  • FIG. 1 FACS analysis of viable and dead Raji-cells and natural killer (NK) cells in the absence of an antibody. Quadrants: lower left: viable NK cells, upper left: dead NK cells, lower right: viable Raji-cells, upper right: dead Raji-cells. Panel A: Raji-cells only, panel B: Raji-cells and NK cells at a ratio of 1:1, panel C: Raji-cells and NK-cells 1:10.
  • FIG. 2 FACS analysis of viable and dead Raji-cells and natural killer cells in the presence of an added anti-CD20 antibody (10 μg/ml). Quadrants: lower left: viable NK cells, upper left: dead NK cells, lower right: viable Raji-cells, upper right: dead Raji-cells. Panel A: Raji-cells only, panel B: Raji-cells and NK cells at a ratio of 1:1, panel C: Raji-cells and NK cells at a ratio of 1:10.
  • FIG. 3 Optimization of lymphoma/spheroid-aggregate co-culture by variation of lymphoma cell, NK cell and antibody application schedule. Lymphoma cells: Raji-cells.
  • FIG. 4 Percentages of viable lymphoma cells in the presence of NK cells as a function of the anti-CD20 antibody concentration. Panel A: Raji-cells, panel B: SU-DHL4 cells, panel C: Z138 cells. NK cell to lymphoma cell ratio (E:T ratio) of 3:1.
  • FIG. 5 Percentages of viable lymphoma cells in the presence of the anti-CD20 antibody concentration as a function of the effector (NK):target (lymphoma) ratio. Panel A: Raji-cells, panel B: SU-DHL4 cells, panel C: Z138 cells. Anti-CD20 antibody concentration: 10 μg/ml.
  • FIG. 6 Schematic exemplary method.
  • FIG. 7 Microscopic images of Raji-cells only and Raji cell co-cultivated with purified NK cells.
  • EXAMPLE 1
  • Material and Methods
  • Cell lines:
  • Raji-cells, SU-DHL4 cells and Z138 cell lines were obtained from ATCC (Manassas, Va., USA), from DSMZ (Braunschweig, Germany) and from Prof. M. Dyer (University of Leicester, UK), respectively. Raji-cells and SU-DHL4 cells were cultivated in RPMI 1640 medium (PAN Biotech, Cat. no. PO4-18500) and Z138 in DMEM medium (PAN Biotech, Cat. no. PO4-02500) supplemented with 10% FCS (Gibco, Cat. no. 10500-064) and Pen/Strep (Roche, Cat. no. 11 074 440 001) at 37° C. in a humidified incubator. Exponential growing cells with cell viability of 90% or more were used for the NK cell co-cultivation experiments.
  • Purification of NK cells:
  • Whole blood was withdrawn from normal healthy donors into vaccutainer tubes (Becton Dickinson, Cat. no. 368484). PBMC were obtained by Ficoll preparation (PAN Biotech Cat. no. PO4-60125). To leave the NK cells untouched, the NK cells were purified using a NK cell, negative selection kit (Miltenyi, Cat. no. 130-092-657). In short, the Ficoll isolated PBMCs were resuspended in MACS-buffer (PBS/0.5% BSA/2 mM EDTA) at 1×107 cells/40 μl. 10 μl of an NK-Cell-Biotin-Antibody cocktail was added to the cells and incubated for 10 min. at 4° C., followed by the addition of 30 μl A MACS buffer. Thereafter, 20 μl A of the NK-Cell-Microbead cocktail was added to the cells and incubated for 15 min. at 4° C. 2 ml of MACS-buffer was added and the cells were centrifuged for 10 min. at 300 g. The pellet was resuspended in 500 μl A MACS-buffer and loaded onto the separation column which was equilibrated with 500 μl A MACS-buffer before. The column was washed subsequently three times with 500 μl MACS-buffer and the cell number was determined in the total eluate using the CASY Cell Counter (Schärfe System).
  • The purity of the NK cell preparation was determined by staining of an aliquot of the MACS eluate. In short about 2×105 cells were resuspended in 100 μl RPMI 1640/10% FCS and stained with 10 μl each of anti-CD56-PE and anti-CD3-FITC antibodies (Becton Dickinson, Cat. no. 555516 and 555339, respectively) for 15 min at 4° C. Thereafter, 2 ml of RPMI 1640/10% FCS were added to the cells which were centrifuged for 5 min. at 400 g. The pellet was resuspended in 0.5 ml RPMI 1640/10% FCS and the percentage of the CD56 positive but CD3 negative cell fraction within the lymphocyte scatter gate was analyzed using a FACS Scan or FACS Canto II instrument (Becton Dickinson).
  • Purification of monocytes:
  • Whole blood was withdrawn from normal healthy donors into vaccutainer tubes (Becton Dickinson, Cat. no. 368484). PBMC were obtained by Ficoll preparation
  • (PAN Biotech Cat. no. PO4-60125). To leave the monocytes untouched, the monocytes were purified using a negative selection, monocyte enrichment kit (Stem Cell Technologies, Cat No.: 19059).
  • CMFDA staining of lymphoma cells:
  • The CMFDA lyophilizate (Invitrogen Cat. no. C7025) was resuspended in DMSO to obtain a 10 mM stock solution. 1×106 lymphoma cells were incubated for 30 min. at 37° C. in 1 ml complete medium supplemented with 1 μM CMFDA. Thereafter, cells were pelleted, washed once in complete medium and resuspended finally in complete medium at 1×106 cells/ml.
  • EXAMPLE 2
  • Generation of 3D spheroids/aggregates from lymphoma cell lines
  • The lymphoma cell number was determined using a CASY instrument (Schärfe-Systems, Reutlingen) and the cell suspension was diluted in ice cold medium to 2.5×104 cells/ml (for 5,000 cells per spheroid/aggregate) and 5×104 cells/ml (for 10,000 cells per spheroid/aggregate). A volume of 200 μl of the cell suspension was added to each well of a 96-well plate with round (Corning Inc., New York, USA) or conical (Nunc, Roskilde, The Netherlands) bottom. To prevent cell attachment the plates were pre-coated with 50 μl 0.5% polyHEMA (Polysciences, Eppelheim, Germany) in 95% ethanol (v/v) and air dried at 37° C. for three days. The spheroid formation was initiated by centrifugation of the plates at 1,000 g for 10 min. using an Eppendorf 5810 centrifuge (Eppendorf AG, Hamburg, Germany) with swinging buckets. The plates were incubated under standard cell culture conditions at 37 ° C. and 7% CO2 in humidified incubators.
  • EXAMPLE 3
  • Generation of 3D spheroids/aggregates from solid tumor cell lines
  • Monolayer cells were detached with Accutase (PAA Laboratories GmbH, Innsbruck, Austria) to generate a single cell suspension. The cell number was determined using a CASY instrument (Schärfe-Systems, Reutlingen) and the cell suspension was diluted in ice cold medium to 2.5×104 cells/ml (for 5,000 cells per spheroid/aggregate) and 5×104 cells/ml (for 10,000 cells per spheroid/aggregate). The rBM was thawed on ice overnight and added at a final concentration of 2.5% (v/v) with ice cold pipette tips to the cell suspension. A volume of 200 μl of the cell suspension was added to each well of a 96-well plate with round (Corning Inc., New York, USA) or conical (Nunc, Roskilde, The Netherlands) bottom. To prevent cell attachment the plates were pre-coated with 50 μl 0.5% polyHEMA (Polysciences, Eppelheim, Germany) in 95% ethanol and air dried at 37° C. for three days. The spheroid formation was initiated by centrifugation of the plates at 1,000 g for 10 min. using an Eppendorf 5810 centrifuge (Eppendorf AG, Hamburg, Germany) with swinging buckets. The plates were incubated under standard cell culture conditions at 37° C. and 7% CO2 in humidified incubators.
  • EXAMPLE 4
  • Spheroid/aggregate lymphoma/NK co-cultivation and incubation with antibody
  • The sequence of cell co-cultivation and antibody addition can be varied. In an exemplary co-cultivation experiment, lymphoma cells (CMFDA labeled) and NK cells were mixed at ratios as indicated in 6 well plates. For example, an E:T (NK cell to lymphoma cell) ratio of 3:1 corresponds to a cell mixture of 3+1 (e. g. 75% NK cells and 25% lymphoma cells). 200 μl of the cell suspension was added to a single well of a polyHEMA coated 96 well V-plate (Nunc, Cat. no. 249662). PolyHEMA coating: 50 μl 0.5% polyHEMA in 95% ethanol per well; drying for 72 h at 37° C. (Polysciences, Cat. No. 18894). The plates were centrifuged for 10 min. at 1,000 g. The antibodies were added thereafter at concentrations as indicated above, and cell aggregates/spheroids were incubated at 37° C., 7% CO2 in a humidified incubator. Microscopic images of Raji-cells only and Raji cell co-cultivated with purified NK cells are shown in FIG. 7, as well as images of co-cultivated Raji and monocyte cells to illustrate 3D co-cultivation of tumor cells with other immune cells then NK cells.
  • EXAMPLE 5
  • Viable cell and cell death analysis
  • Spheroids/aggregates were generated using 10,000 cells and incubated with the antibody as outlined in Examples 2 and 3. The identification of viable lymphoma tumor cells was as follows: Individual aggregates from individual wells representing identical experimental conditions were pooled, dissociated by pipetting and centrifuged at 300 g for 10 min. Individual spheroids were pooled, washed once with phosphate buffered saline (PBS), resuspended in Accutase solution, and incubated at 37° C. Every five minutes, the spheroids/aggregates were resuspended by pipetting and dissociation was complete within 5 to 15 min. Cells were washed using complete medium, centrifuged and cell pellets were resuspended in complete medium and propidium iodide was added at a concentration of 1 μg/ml (Sigma, Cat. no. P4170). Fluorescence analysis was performed by FACS analysis (Becton Dickinson, Canto II instrument).
  • Viable lymphoma cells were identified as shown in FIG. 1 b. The upper right quadrant of PI and CMFDA positive cells represent the dead lymphoma cells, and the lower right quadrant of PI negative but CMFDA positive cells represent the viable lymphoma tumor target cell fraction. In the lower left quadrant are viable NK cells, whereas dead NK cells are located in the upper left quadrant.
  • In an alternative setting an apoptosis assay can be performed. Spheroids/aggregates were generated using 10,000 cells and incubated with the antibody as outlined in Examples 2 and 3. For apoptosis analysis, the spheroids/aggregates were transferred into a 96-well conical-bottom plate, washed once with phosphate buffered saline (PBS), resuspended in Accutase solution, and incubated at 37° C. Every five minutes, the spheroids/aggregates were resuspended by pipetting and dissociation was complete within 5 to 15 min. The single cell suspensions from eight spheroids/aggregates were pooled and cells were stained with annexin-V-fluos and propidium iodide in the presence of supplemented 2 mM CaCl2, (annexin-V-fluos staining kit, Roche Diagnostics GmbH, Mannheim, Germany). The fluorescence of 10,000 cells was acquired using a flow cytometer (FACS scan instrument, Becton Dickinson, San Jose, Ca., USA). Quadrant statistics was applied on the dot plots, with the number of viable cells located in the lower-left quadrant.
  • To obtain the absolute number of dead and viable cells, the number of total cells from the spheroids/aggregates were counted using a Fuchs-Rosenthal cell counting chamber and multiplied with the percentage of viable or dead cells of the same spheroids/aggregates as determined from the annexin-V-fluos/PI staining

Claims (14)

1. A method for the in vitro detection of effector function of an antibody comprising incubating a three-dimensional spheroid comprising tumor cells and natural killer cells or a three-dimensional aggregate comprising tumor cells and natural killer cells with the antibody.
2. The method according to claim 1 comprising the following steps:
a) mixing natural killer cells and tumor cells,
b) adding about 104 cells per 200 μl to the wells of a multi-well plate,
c) centrifuging the multi-well plate to induce the formation of a three-dimensional spheroid or a three-dimensional aggregate,
d) adding the antibody to the wells of the multi-well plate,
e) incubating the multi-well plate for about 20 hours to about 72 hours,
f) analyzing the cells in the wells of the multi-well plate by fluorescence activated cell sorting, and
g) detecting the effector function of the antibody.
3. The method according to claim 2, wherein the natural killer cells are human natural killer cells.
4. The method according to claim 2, wherein the natural killer cells and tumor cells are mixed at a ratio of from 10:1 to 1:10.
5. The method according to claim 4, wherein the ratio is from 1:2 to 1:4.
6. The method according to claim 1 or claim 2, wherein the incubating is for about 20 hours to about 28 hours.
7. The method according to claim 2, wherein the centrifuging is at 1,000 rpm for 10 min.
8. The method according to claim 1, wherein the tumor cell is a lymphoma cell.
9. The method according to claim 8, wherein the lymphoma cell is selected from a Raji-cell, a SU-DHL4 cell, and a Z138 cell.
10. The method according to claim 2, wherein the antibody is added at a concentration of from 15 μg/ml to 0.1 μg/ml.
11. The method according to claim 10, wherein the antibody is added at a concentration of from 8 μg/ml to 12 μg/ml.
12. A method comprising use of a three-dimensional spheroid comprising a multitude of tumor cells and natural killer cells or a three-dimensional aggregate comprising a multitude of tumor cells and natural killer cells for the determination of effector function of a combination of a multitude of antibodies with the multitude of tumor cells.
13. A method for determining in vitro an antibody with effector function from a multitude of provided antibodies comprising
a) mixing natural killer cells and tumor cells,
b) adding about 104 cells per 200 μl to the wells of a multi-well plate,
c) centrifuging the multi-well plate to induce the formation of a three-dimensional spheroid or a three-dimensional aggregate,
d) adding each of the provided antibodies to an individual well of the multi-well plate,
e) incubating the multi-well plate for about 20 hours to about 72 hours, and
f) determining the antibody with a ratio of more than 1 of dead cells to viable cells as the antibody with effector function.
14. A kit comprising:
a) a tumor cell labeled with a fluorescent dye,
b) isolated natural killer cells,
c) a 96-well multi-well plate, and
d) propidium iodide.
US13/577,309 2010-02-11 2011-02-04 3d adcc nk facs assay Abandoned US20120309016A1 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
EP10153277.8 2010-02-11
EP10153277 2010-02-11
PCT/EP2011/051633 WO2011098402A1 (en) 2010-02-11 2011-02-04 3d adcc nk facs assay

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2011/051633 A-371-Of-International WO2011098402A1 (en) 2010-02-11 2011-02-04 3d adcc nk facs assay

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US15/148,938 Continuation US10330669B2 (en) 2010-02-11 2016-05-06 3D ADCC NK FACS assay

Publications (1)

Publication Number Publication Date
US20120309016A1 true US20120309016A1 (en) 2012-12-06

Family

ID=42169475

Family Applications (2)

Application Number Title Priority Date Filing Date
US13/577,309 Abandoned US20120309016A1 (en) 2010-02-11 2011-02-04 3d adcc nk facs assay
US15/148,938 Active 2031-09-27 US10330669B2 (en) 2010-02-11 2016-05-06 3D ADCC NK FACS assay

Family Applications After (1)

Application Number Title Priority Date Filing Date
US15/148,938 Active 2031-09-27 US10330669B2 (en) 2010-02-11 2016-05-06 3D ADCC NK FACS assay

Country Status (12)

Country Link
US (2) US20120309016A1 (en)
EP (1) EP2534480B1 (en)
JP (1) JP5719384B2 (en)
KR (1) KR101497196B1 (en)
CN (1) CN102741695B (en)
BR (1) BR112012016230A2 (en)
CA (1) CA2787157A1 (en)
ES (1) ES2459122T3 (en)
HK (1) HK1174392A1 (en)
MX (1) MX336473B (en)
RU (1) RU2577702C2 (en)
WO (1) WO2011098402A1 (en)

Families Citing this family (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10655107B2 (en) 2011-09-20 2020-05-19 Corning Incorporated Adherent cell culture method
US10883083B2 (en) 2013-08-02 2021-01-05 The Trustees Of Columbia University In The City Of New York Tissue-engineered three-dimensional model for tumor analysis
CN103712967B (en) * 2013-12-27 2014-12-03 首都医科大学附属北京友谊医院 Method for detecting activity of NK (Natural Killer) cell
WO2015173425A1 (en) 2014-05-16 2015-11-19 Koninklijke Nederlandse Akademie Van Wetenschappen Improved culture method for organoids
GB201421092D0 (en) 2014-11-27 2015-01-14 Koninklijke Nederlandse Akademie Van Wetenschappen Culture medium
GB201603569D0 (en) 2016-03-01 2016-04-13 Koninklijke Nederlandse Akademie Van Wetenschappen Improved differentiation method
KR101935989B1 (en) 2017-01-18 2019-01-07 서강대학교산학협력단 Method for manufacturing multicellular spheroid using hydrogel microwell
JP2021511790A (en) * 2018-02-02 2021-05-13 ウェイク・フォレスト・ユニバーシティ・ヘルス・サイエンシーズWake Forest University Health Sciences Immunotherapy organoids and how to prepare and use them
WO2023103785A1 (en) * 2021-12-06 2023-06-15 Wuxi Biologics (Shanghai) Co., Ltd. 3d bioassays to measure antibody-dependent cell-mediated cytotoxicity

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5420016A (en) * 1992-03-24 1995-05-30 Serim Research Corporation Test device and kit for detecting helicobacter pylori
US20040219224A1 (en) * 2001-06-21 2004-11-04 Kirill Yakovlevsky Spherical protein particles and methods for making and using them

Family Cites Families (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0302435A1 (en) * 1987-07-31 1989-02-08 Neorx Corporation Method for augmenting antibody-dependent cellular cytotoxicity
BRPI0412890B8 (en) * 2003-07-24 2021-05-25 Innate Pharma method of selecting an anti-kir2dl1 antibody or antigen-binding antibody fragment
EA036531B1 (en) 2003-11-05 2020-11-19 Роше Гликарт Аг Type ii anti-cd20 humanized antibody (variants), pharmaceutical composition comprising these antibody variants, and use thereof
RU2409816C2 (en) * 2006-01-04 2011-01-20 Новартис Аг Antibody-dependent cellular cytotoxicity assay
JP5282040B2 (en) * 2007-09-28 2013-09-04 オリンパス株式会社 Method for assaying antibody-dependent cytotoxicity using Fc receptor gene-transferred NK cells

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5420016A (en) * 1992-03-24 1995-05-30 Serim Research Corporation Test device and kit for detecting helicobacter pylori
US20040219224A1 (en) * 2001-06-21 2004-11-04 Kirill Yakovlevsky Spherical protein particles and methods for making and using them

Non-Patent Citations (5)

* Cited by examiner, † Cited by third party
Title
Flieger et al., A novel non-radioactive cellular cytotoxicity test based on the differential assessment of living and killed target and effector cells, Journal of Immunological Methods 180 (1995) pages 1-13. *
Ivascu et al., Rapid Generation of Single-Tumor Spheroids for High-Throughput Cell Function and Toxicity Analysis, J Biomol Screen (2006) pages 922-932. *
Liu et al., Apoptosis of Raji cells by an anti-CD20 antibody H147 and its fragments, Leukemia Research 28 (2004) pages 209-211. *
Masuda et al., Enhanced binding affinity for FcRIIIa of fucose-negative antibody is sufficient to induce maximal antibody-dependent cellular cytotoxicity, Molecular Immunology 44 (2007) pages 3122-3131. *
Ortaldo et al., Analysis of effector cells in human antibody-dependent cellular cytotoxicity with murine monoclonal antibodies, Journal of Immunology (1987) pages 3566-3572. *

Also Published As

Publication number Publication date
CA2787157A1 (en) 2011-08-18
ES2459122T3 (en) 2014-05-08
CN102741695A (en) 2012-10-17
HK1174392A1 (en) 2013-06-07
WO2011098402A1 (en) 2011-08-18
US10330669B2 (en) 2019-06-25
RU2577702C2 (en) 2016-03-20
CN102741695B (en) 2015-08-26
EP2534480A1 (en) 2012-12-19
MX336473B (en) 2016-01-20
JP2013519360A (en) 2013-05-30
JP5719384B2 (en) 2015-05-20
RU2012138554A (en) 2014-03-20
US20160252493A1 (en) 2016-09-01
KR20120125369A (en) 2012-11-14
BR112012016230A2 (en) 2019-09-24
MX2012008950A (en) 2014-02-27
EP2534480B1 (en) 2014-03-19
KR101497196B1 (en) 2015-02-27

Similar Documents

Publication Publication Date Title
US10330669B2 (en) 3D ADCC NK FACS assay
Lea Caco-2 cell line
Guldevall et al. Microchip screening platform for single cell assessment of NK cell cytotoxicity
Bai et al. based 3D scaffold for multiplexed single cell secretomic analysis
Zhou et al. Microfluidic device for primary tumor spheroid isolation
AU2002365177A1 (en) Microarrays for cell phenotyping and manipulation
Lindström et al. Towards high‐throughput single cell/clone cultivation and analysis
Papp et al. Life on a microarray: assessing live cell functions in a microarray format
CN107904278B (en) Method for detecting influence of drug on cell proliferation
JPWO2020045368A1 (en) Evaluation method for anti-infective drugs, vaccines, etc. using immortalized monocyte cells and inducible cells
CN110716053B (en) Novel method for detecting population reactive antibodies
Desalvo et al. Time-resolved microwell cell-pairing array reveals multiple T cell activation profiles
Cedillo-Alcantar et al. On-Chip analysis of protein secretion from single cells using microbead biosensors
Jones et al. Characterization of cell seeding and specific capture of B cells in microbubble well arrays
EP3203237B1 (en) Urine-derived epithelial cell lines for diagnosis and therapy of an anti-bk-virus or anti-graft immune response
Perez et al. Multiparameter analysis of intracellular phosphoepitopes in immunophenotyped cell populations by flow cytometry
CN112921004B (en) Cell strain for detecting CD3 receptor agonist, construction method thereof and CD3 receptor agonist detection method
Baecher‐Allan et al. The purification and functional analysis of human CD4+ CD25high regulatory T cells
Chan Novel plate-based detection method for T cell activation/proliferation, migration, and cytotoxicity assay using image cytometry
Carannante et al. Generation of tumor spheroids in microwells to study NK cell cytotoxicity, infiltration and phenotype
Schwietzer et al. A tractable microscopy-and flow cytometry-based method to measure natural killer cell-mediated killing and infiltration of tumor spheroids
Migita et al. Reproducible fashion of the HSP70B'promoter‐induced cytotoxic response on a live cell‐based biosensor by cell cycle synchronization
Beelen et al. An in vitro model to monitor natural killer cell effector functions against breast cancer cells derived from human tumor tissue
Padhan et al. Human follicular CD4 T cell function is defined by specific molecular, positional and TCR dynamic signatures
Da Silva et al. Mouse CD8+ T cell migration in vitro and CXCR3 internalization assays

Legal Events

Date Code Title Description
AS Assignment

Owner name: F. HOFFMANN-LA ROCHE AG, SWITZERLAND

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:CHALLAND, ANDREA;KLEIN, CHRISTIAN;KUBBIES, MANFRED;SIGNING DATES FROM 20120625 TO 20120703;REEL/FRAME:031125/0983

Owner name: HOFFMANN-LA ROCHE INC., NEW JERSEY

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:F. HOFFMANN-LA ROCHE AG;REEL/FRAME:031126/0090

Effective date: 20120726

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION