Abstract
The antitumor efficacy of genetically engineered “living drugs,” including chimeric antigen receptor and T-cell receptor T cells, is influenced by their activation, proliferation, inhibition, and exhaustion. A sensitive and reproducible cytotoxicity assay that collectively reflects these functions is an essential requirement for translation of these cellular therapeutic agents. Here, we compare various in vitro cytotoxicity assays (including chromium release, bioluminescence, impedance, and flow cytometry) with respect to experimental setup, appropriate uses, advantages, disadvantages, and measures to overcome limitations. We also highlight the FDA directives for a potency assay for release of clinical cell therapy products. In addition, we discuss advanced assays of repeated antigen exposure and simultaneous testing of combinations of immune effector cells, immunomodulatory antibodies, and targets with variable antigen expression. This Review article should help to equip investigators with the necessary knowledge to select appropriate cytotoxicity assays to test the efficacy of immunotherapeutic agents alone or in combination.
Introduction
Immunotherapy harnesses a patient’s immune system to combat cancer, either by restoring the function of endogenous immunity or by generating new immune responses through the establishment of synthetic immunity. The former has been achieved through the use of immune checkpoint inhibitors—a class of immunomodulatory antibodies that target inhibitory receptors on T-cells, such as programmed cell death protein 1 (PD-1) and cytotoxic T-lymphocyte–associated protein 41—and bispecific T-cell engager (BiTE) antibodies, which mediate T-cell responses by binding to a target on the tumor cell and T cell simultaneously.2 With continuing advances in genetic engineering, biological therapies, such as adoptive T cells and oncolytic viruses, are being translated to clinical use to lyse tumor cells. The resulting release of cancer cell antigens in an altered tumor immune microenvironment attracts cells of both the innate and the adaptive immune system to the tumor,3 promoting neoantigen immune responses.4
Adoptive cell therapy relies on the isolation and expansion of the patient’s immune cells (such as T cells), which are genetically modified to express a cancer antigen–specific chimeric antigen receptor (CAR) or T-cell receptor (TCR). CAR T cells—a patient’s own T cells transduced to express a CAR—are called “living drugs” because of their ability to proliferate, expand, and persist following antigen stimulation. They have been approved for the treatment of hematological malignancies. CAR T cells, CAR natural killer (NK) cells, and CAR macrophages, all subsets of engineered immune cells, are currently in clinical trials for the treatment of solid tumors.5 Genetic-engineering tools, such as CRISPR,6 can be used to potentiate CARs by targeting specific pathways involved in the suppression of the immune effector function in the tumor microenvironment.7 More importantly, such tools can be used to insert CARs at the TCRα constant locus.8 Following the establishment of the feasibility, safety, efficacy, and persistence of CRISPR-engineered T cells in patients,9 the use of multiplex genome-engineering techniques is being extended to allogeneic donor T cells, thereby increasing their broad applicability.
Given the variety and complexity of multigene-engineered living drugs, there is an increasing need for reproducible and high-throughput screening assays to select the best therapeutic agents. A first step to screening agents is to measure the cancer cell–killing ability of an effector cell with a cytotoxicity assay. In this review article, we describe assays that quantify target cell lysis mediated by effector cells as a measure of cell-mediated cytotoxicity. We specifically focus on four of the most commonly used assays to investigate cell-mediated cytotoxicity: the chromium (51Cr)–release assay (51Cr assay), the luciferase-mediated bioluminescence imaging (BLI) assay, the impedance-based assay, and the flow cytometry assay (flow assay) (Figure 1 and Table 1). These assays differ in their requirements for target cell labeling, culture time, principal measures of cytotoxicity, number of measurements (temporal or endpoint), ability to measure differential cytotoxicity on heterogenous targets, throughput, and automatability. We further discuss their advantages and limitations and give specific examples of their applications for different classes of immunotherapies (Tables 2–5).
Figure 1. Interassay comparison of cell-mediated cytotoxicity assessment.
The figures show representative readouts of the respective assays to quantify cell-mediated cytotoxicity. a, 51Cr-release assay. 51Cr release of labeled target cells cocultured with target-specific effector cells (red curve) or control cells (blue curve) at different effector to target (E:T) ratios is determined relative to a maximum and a spontaneous 51Cr-release control. b, Bioluminescence assay. Bioluminescence intensity proportional to the number of viable luciferase-expressing target cells is indicative of effector cell–mediated cytotoxicity at different E:T ratios. c, Impedance-based assay. Detachment of adherent target cells is monitored as a decrease in normalized cell index upon addition of target-specific effector cells (red curve) or control cells (blue curve) at a specific E:T ratio and at a defined time after seeding of target cells (dotted line). Untreated target cells and target cells treated with 0.1% Triton X-100 (100% lysis control) are represented by the purple and green curves, respectively. d, Flow cytometry assay. The dot plot shows 7-amino-actinomycin D (7-AAD) and Annexin V staining of target cells undergoing effector cell–mediated apoptosis and cell death. Lower left quadrant: Live cells. Lower right quadrant: Early apoptotic cells. Upper right quadrant: Cells at terminal stage of apoptosis or cell death.
Table 1.
Properties of cell-mediated cytotoxicity assays
Assay format | Chromium release | Bioluminescence | Impedance | Flow cytometry |
---|---|---|---|---|
Principal measure of cytotoxicity | 51Cr release | Luciferase activity | Cell detachment | Live/dead staining, phenotype |
Radioactive materials needed | Yes | No | No | No |
Target cell labeling required | Yes | No | No | Yes |
Genetic modification of target cells required | No | Yes (reporter gene) | No | No |
Endpoint/kinetic | Endpoint | Endpoint | Temporal | Endpoint |
Real-time measurement | No | No | Yes | No |
Maximum time point measured | 18–24 hours | Days | Days | Days |
Ability to measure differential cytotoxicity on heterogenous targets | No | No | No | Yes |
Throughput and automatability | Low | High | High | High |
Table 2. Applications of cell-mediated cytotoxicity assays.
Listed are examples of published assays to analyze cytotoxic effects mediated by different effector cells, illustrating the various uses of the 51Cr-release assay.
51Cr-release assay | ||||||
---|---|---|---|---|---|---|
Application | Effector cells/drug | Target cells | Target cell number per well | E:T ratio(s) | Incubation time (h) | Reference |
T cell–mediated cytotoxicity | CD8 T-cell clone 3E6 | U251T | 2,000 or 5,000 | 50:1 to 0.3:1 | 4 | 24 |
Human PBMCs and murine splenocytes | K562, U266, UC101, P815, YAC1, EL-4, A20 | 100,00/mL | 100:1 to 5:1 | 2, 4 | 16 | |
Mesothelin-targeted CAR T cells with intrinsic checkpoint blockade | MSTO-211H expressing human mesothelin | 5,000 | 64:1 to 0.5:1 | 18 | 61 | |
TCR T cells specific for TARP | T2, Mel526 expressing TARP | 1,000 | 50:1 to 0.4:1 | 4 | 81 | |
T cells recognizing influenza matrix protein–derived peptide | OAW42 loaded with peptide | 20,000 | 5:1 to 0.005:1 | 6, 24 | 15 | |
HER2-specific T cells | SKBR3 | 100,000 | 40:1 to 1.25:1 | 5 | 34 | |
NK cell– mediated cytotoxicity | NK cells | K-562, Daudi | 10,000 | 20:1 to 2.5:1 | 4 | 82 |
NK cells | K562 | Unspecified | 100:1 to 3.125:1 | 4 | 44 | |
Oncolytic virus–mediated cytotoxicity | NK cells, parvovirus H-1PV | H-1PV–infected Panc-1, MiaPaCa-2, Capan-1, CF-Pac1, AsPC-1 | 5,000 | 20:1 to 2.5:1 | 4, 16 | 83 |
BiTE-mediated cytotoxicity | T cells, BiTE (HER2×CD3) | Panc89, Colo357 | Unspecified | 50:1 to 6.25:1 | 4 | 84 |
ADCC | Rituximab, T cells transduced with CD16 | Autologous B-lymphoblastoid cell line | Unspecified | 30:1 | 4 | 85 |
Cetuximab, PBMCs | A549, N417, LK87, Lu99 | 10,000 | 40:1 to 10:1 | 4 | 86 | |
Rituximab, macrophages | Raji | Unspecified | 50:1 to 12.5:1 | 8 | 87 | |
Rituximab or trastuzumab, T cells transduced with CD16 | Raji, SKOV-3 | 3,000 | 10:1 | 4 | 21 |
ADCC, antibody-dependent cellular cytotoxicity; BiTE, bispecific T-cell engager; CAR, chimeric antigen receptor; E:T, effector to target; PBMC, peripheral blood mononuclear cell; NK, natural killer; TARP, T-cell receptor γ chain alternate reading-frame protein; TCR, T-cell receptor.
Table 5. Applications of cell-mediated cytotoxicity assays.
Listed are examples of published assays to analyze cytotoxic effects mediated by different effector cells, illustrating the various uses of the flow cytometry assay.
Flow cytometry assay | ||||||
---|---|---|---|---|---|---|
Application | Effector cells/drug | Target cells | Staining | E:T ratio(s) | Incubation time (h) | Reference |
T cell– mediated cytotoxicity | NY-ESO–specific T cells | Jurkat/A2K | CFSE, CMTMR, propidium iodide | 1.5:1 to 0.1:1 | 4 | 97 |
CD8+ T cells | DoHH2, Karpas, Raji | CFSE, 7-AAD | 50:1 to 2.5:1 | 24 | 98 | |
γδ T cells | THP-1 | CFSE, propidium iodide | 20:1 to 1:1 | 4 | 39 | |
NK cell– mediated cytotoxicity | NK cells | K562 | CellTrace Violet Dye | 1:1 | 5 | 45 |
NK cells | K562 | Annexin V, propidium iodide, BCECF | 1:1 to 1:10 | 1 to 3 | 99 | |
NK cells | K562 | DiO, propidium iodide | 75:1 to 6.25:1 | 3 | 44 | |
NK cells | K562 | DIOC18, 7-AAD | 20:1 to 1:1 | 4 | 100 | |
Macrophage-mediated cytotoxicity | U937 | IGROV1 | CFSE, propidium iodide, CD89-phycoerythrin | 2:1 to 1:1 | 2.5 | 101 |
Oncolytic virus–mediated cytotoxicity | PBMCs, adenovirus expressing BiTE (EGFR×CD3) | A431, A549 | GFP, 7-AAD | MOI = 20, E:T = 6:1 | 5 days | 95 |
BiTE-mediated cytotoxicity | CD3+ T cells enriched from PBMCs, BiTE (CEA×CD3) | CHO, ASPC-1, BxPC3, HPAC, HPAF II, H727, LS174T, HT-29, MKN45, Pan0813, A549, PC3, BT474 | Vybrant DiO, propidium iodide | 10:1 to 1:2 | Up to 72 | 102 |
ADCC | PBMCs, NK92MI, trastuzumab | BT-474, MCF-7, Hs578T | CFSE, FVD/7-AAD/propidium iodide | 1:1 to 1:20 | Overnight | 103 |
7-AAD, 7-amino-actinomycin D; ADCC, antibody-dependent cellular cytotoxicity; BCECF, 2’,7’-bis(carboxyethyl)-5(6)-carboxyfluorescein; BiTE, bispecific T-cell engager; CFSE, carboxyfluorescein succinimidyl ester; CMTMR, 5- and 6-([(4-chloromethyl)benzoyl]-amino) tetramethylrhodamine; E:T, effector to target; FVD, fixed viability dye; GFP, green fluorescent protein; MOI, multiplicity of infection; PBMC, peripheral blood mononuclear cell; NK, natural killer.
51Cr assay
Historically, methods to measure cell-mediated cytotoxicity have been based on the quantification of target cells with loss of membrane integrity. The 51Cr assay, which works on this principle, was first described more than five decades ago by Brunner et al.10 and gained popularity as the gold standard to measure T cell– and NK cell–mediated cytotoxicity.11,12
In this assay, target cells are labeled with radioactive chromium (51Cr) and coincubated with effector cells at different effector-to-target (E:T) ratios. During effector-mediated cell killing, the target cells lose their membrane integrity and release 51Cr into the culture medium. Radioactivity (measured in the supernatant using a gamma scintillator counter) is proportional to the number of target cells killed during that period. A spontaneous release control (51Cr-labeled target cells alone in culture medium) and a maximum release control (51Cr-labeled target cells in culture medium treated with a detergent, such as Triton X-100) are included to allow determination of the percentage of specific lysis, which is calculated using the following formula: [(experimental release – spontaneous release)/(total release – spontaneous release)]*100.10,13
The standard 51Cr assay measures the 51Cr released 4 h after coincubation of effector and target cells, but longer incubation times might be necessary to allow observation of significant cell killing for more-resistant target cells and for solid-tumor cells in particular (Table 2). As an endpoint assay, the 51Cr assay is usually limited to a single time point, but measurements can be obtained at more than one time point if only part of the radioactive supernatant is used.14 However, spontaneous 51Cr release from the target cells leads to an increased background and a reduced signal-to-background ratio over time, which thereby prevents cytotoxicity measurement over extended periods.15 51Cr release in the assay is higher with detergent solubilization than with hypotonic lysis, leading to overestimation of 51Cr release and underestimation of cytotoxicity.16
In light of the potential effects of 51Cr on the health of the investigator, an alternative method that uses nonradioactive chromium was developed in 1995.17 However, because of its requirement for expensive equipment and specialized staff and its low-throughput capability, this method has never achieved widespread use. Despite its technical limitations, the 51Cr assay is considered the gold standard for measurement of cell-mediated cytotoxicity. For evaluation of the cytotoxicity of immune cells transduced with newer constructs, such as CARs or TCRs, our laboratory performs the 51Cr assay as a first step, with inclusion of well-established constructs for comparison. However, not all laboratories possess the mandatory radiation license and infrastructure for radioisotope storage, disposal, and monitoring. The following sections will provide investigators with nonradioactive alternatives to the 51Cr assay.
BLI luciferase assay
This BLI-based cytotoxicity assay measures the light (photons) emitted from target cells that are transduced with a luciferase reporter gene. The luciferase substrate is processed only by live cells that express the luciferase transgene. Consequently, the cytotoxicity of effector cells can be measured by the decrease in the BLI signal.16
Early luciferase-based cytotoxicity assays measured the activity of secreted firefly luciferase in the supernatant upon lysis of target cells. However, because of the short half-life (approximately 30 mins) of firefly luciferase in culture medium, these assays were restricted to assay conditions in which substantial lysis of the target cells occurs within a few hours.18 Furthermore, because of its molecular weight of 62 kDa,19 firefly luciferase might not efficiently leak into the supernatant upon effector cell–mediated pore formation in the target cell.20 Therefore, particularly for cytotoxicity studies that require longer incubation times, complete cell lysis might be necessary to allow for measurement of cell-associated luciferase activity,20 which would limit the readout to a single endpoint. However, luciferases derived from marine organisms are smaller in size, exhibit longer half-lives, and have increased brightness.21–23 For example, nanoluciferase, a 19-kDa reporter protein derived from deep-sea shrimp that has a half-life of 7.7 days in extracellular medium, is completely released during cell death, enabling the measurement of luciferase signal in supernatants over prolonged periods.21 In a similar assay, sensitivity was not compromised when a centrifugation step to separate supernatant and cell pellet was omitted.23 The ongoing development of such single-step homogenous assays has further driven the miniaturization and automation of BLI reporter assays.
Both the BLI assay and the 51Cr assay show an E:T ratio–dependent proportional increase in cytotoxicity, but the BLI assay yields a greater signal-to-background ratio16 and an up to 2-fold higher percentage of specific lysis than the 51Cr assay.16,24 Rossignol et al. found a comparable signal-to-background ratio between a nanoluciferase assay and the 51Cr assay, with similar cytotoxicity curves but with slight differences in EC50 values, likely because of different release mechanisms between nanoluciferase and 51Cr.21
Due to its ease of handling and simple quantification, the BLI assay has gained universal use (Table 3). However, a prerequisite for BLI-based reporter assays is the expression of luciferase in target cells. Stable transfection of cells is time-consuming and not feasible for every type of cell; therefore, the BLI assay is particularly useful for cases in which the same target cell line is used throughout the screening procedure. Of note, the BLI assay has also been successfully used with target cells that only transiently express the luciferase gene.23,24 Low transfection efficacies of plasmid-based approaches in primary cells can be overcome by electroporating target cells with luciferase gene–encoding RNAs produced in large amounts by in vitro transcription.25
Table 3. Applications of cell-mediated cytotoxicity assays.
Listed are examples of published assays to analyze cytotoxic effects mediated by different effector cells, illustrating the various uses of the bioluminescence assay.
Bioluminescence assay | ||||||
---|---|---|---|---|---|---|
Application | Effector cells/drug | Target cells | Target cells number per well | E:T ratio(s) | Incubation time (h) | Reference |
T cell– mediated cytotoxicity | CD19-specific CAR T cells | Raji | Unspecified | 10:1 | 4 | 23 |
Murine splenocytes | Neuro2a | Unspecified | 40:1 to 5:1 | 2 days | 88 | |
CD8 T-cell clone 3E6, 2D7, 2A7 | U251T, Daudi | 2,000 to 10,000 | 50:1 to 0.3:1 | 1–4 | 24 | |
Human PBMCs and murine splenocytes | K562, U266, UC101, P815, YAC1, EL-4, A20 | 300,000/mL | 100:1 to 5:1 | 2, 4 | 16 | |
Splenocytes transduced with HER2-specific TCR | 4T1 expressing HER2 | Unspecified | 20:1 to 5:1 | Up to 72 | 20 | |
NK cell–mediated cytotoxicity | NK92MI with CD19-specfic CAR | Raji | Unspecified | 0.5:1 | 4 | 23 |
NK cells | YAC-1, TS/A | 5,000 | 40:1 to 10:1 | 4 | 89 | |
Oncolytic virus–mediated cytotoxicity | NK92 with EGFR-specific CAR, herpes simplex virus 1 | MDA-MB-231 | 5,000 | Unspecified | Up to 4 days | 90 |
BiTE-mediated cytotoxicity | T cells, BiTE (EGFR×CD3) | SW480, OVCAR-8, A2058 | 2,500 | 10:1 | 48 | 91 |
T cells, blinatumomab | Raji | Unspecified | 20:1 | 4 | 23 | |
ADCC | IMAB 362, PBMCs | KATO-III, NUGC-4 | 20,000 to 25,000 | 40:1 | 24 | 25 |
Rituximab, PBMCs | Raji | Unspecified | 40:1 | 4 | 23 | |
Rituximab or trastuzumab, T cells transduced with CD16 | Raji, SKOV-3 | 3,000 | 10:1 | 4 | 21 |
ADCC, antibody-dependent cellular cytotoxicity; BiTE, bispecific T-cell engager; CAR, chimeric antigen receptor; E:T, effector to target; PBMC, peripheral blood mononuclear cell; NK, natural killer; TCR, T-cell receptor.
Impedance-based assay
A label-free alternative to the 51Cr assay and the BLI assay relies on microelectrodes embedded in the bottom of a microtiter well to measure the impedance of the flow of an electric current between electrodes upon adhesion of cells. Whereas impedance in the absence of cells is basically zero and is mainly dependent on the ionic environment of the medium, attachment of cells to the electrodes leads to a change in impedance that is dependent on the number, morphologic aspect (confluence), viability, and degree of adhesion strength for each cell type.26,27 Electrode impedance is reported as “cell index,” a dimensionless parameter that is proportional to the number of adherent cells and is used to monitor changes in impedance over time. Suspension and nonadherent cells typically cause no change in electric impedance or significantly less change than adherent cells. This principle informs the measurement of cytolysis of adherent target cells by nonadherent effector cells, such as T cells, NK cells, and other immune cell subsets.28 The workflow for this assay includes measuring the baseline impedance of medium alone, followed by seeding with the target cells. The seeding concentration of the target cells is optimized to reach a cell index in the upper-to-maximum range at the time that the effector cells are added, often 20 to 30 h after seeding, for convenience reasons.29 In contrast to the 51Cr assay and the luciferase assay, the impedance-based assay allows label-free, real-time monitoring of cytolysis (measured as target cell detachment) over an interval of time. Dynamic monitoring of treatment efficacy can be used to compare the cytotoxicity kinetics of different cells and varied constructs at multiple E:T ratios over a period of several days—a major advantage compared with other assays. Very low E:T ratios (in the range of 0.05:1 to 10:1) are feasible because of the nature of the assay, allowing it to be conducted over the course of several days,15,30 significantly reducing the number of effector cells needed.
As the measurement of impedance is dependent on the adherence of cells at the electrode-solution interface, precoating the wells with extracellular matrix proteins, such as fibronectin, laminin, and collagen, can improve binding of lightly adherent cells and has been shown to induce adhesion of leukemia and lymphoma cells.31 Another strategy to tether suspension cells to the electrodes is to coat the wells with antibodies against cell-specific surface markers (e.g., CD19 or CD40 for B-cell tumors).32 Specialized assay applications exist to monitor cell migration or indirect cytotoxicity through soluble factors by using cell-permeable or cell-impermeable membranes, respectively.33
Several groups have compared the impedance-based assay to the 51Cr assay, with inconsistent results. Peper et al. found a close correlation between the assays at 6 h and complete target cell lysis at 24 h for both assays, but the impedance-based assay exhibited a significantly higher percentage of specific lysis at E:T ratios <5:1 at 24 h.15 In contrast, Erskine et al. reported a significant difference between both assays at 5 h, with the impedance-based assay showing a higher level of lysis than the 51Cr assay.34 Whereas Erskine et al. used >6 times more target cells in the 51Cr assay, Peper et al. used the same number of target cells in both assays. Both groups concluded that the increased lysis in the impedance-based assay can be attributed to its higher sensitivity, compared with the 51Cr assay. Owing to its ability to measure effector cell function at low E:T ratios without the need for manipulation of the target cells, the impedance-based assay is increasingly used to study immunotherapeutic interventions (Table 4).
Table 4. Applications of cell-mediated cytotoxicity assays.
Listed are examples of published assays to analyze cytotoxic effects mediated by different effector cells, illustrating the various uses of the impedance-based assay.
Impedance-based assay | ||||||
---|---|---|---|---|---|---|
Application | Effector cells/drug | Target cells | Target cell number per well | E:T ratio(s) | Incubation time (h) after effector addition | Reference |
T cell– mediated cytotoxicity | HER2-specific CAR T cells | MC57 expressing HER2 | 20,000 | 10:1 to 1:1 | Up to 50 | 30 |
T cells recognizing influenza matrix protein–derived peptide | OAW42 | 20,000 | 10:1 to 0.005:1 | Up to 81 | 15 | |
HER2-specific T cells | SKBR3 | 7,500 | 40:1 to 1.25:1 | Up to 20 | 34 | |
NK cell– mediated cytotoxicity | NK92 | PC3, MCF7 | 10,000 to 40,000 | 10:1 to 0.625:1 | >40 | 32 |
NK92 | Raji tethered to plate via anti-CD40 antibody | 60,000 | 2:1 to 0.1:1 | >20 | 32 | |
Macrophage-mediated cytotoxicity | Differentiated U937, ANA-1 | MDA-MB-231, MDA-MB-435, MCF-7 | 2,500 to 20,000 | 40:1 | 48 | 92 |
Differentiated U937 | MDA-MB-231 | Unspecified | 1:5 | 72 | 93 | |
Oncolytic virus–mediated cytotoxicity | Adenovirus-parvovirus chimera | HeLa, A549, ME-180, Lox-IMVI, HCT-15, HCC-2998, pMeIL | 4,000 to 8,000 | MOI = 100 to 1 | Up to 1 week | 94 |
Preactivated T cells, adenovirus expressing BiTE (EGFR×CD3) | A549, HCT116 | 10,000 | MOI = 1, E:T = 5:1 | >90 | 95 | |
BiTE-mediated cytotoxicity | T cells, BiTE (HER2×CD3; HER2× Vγ9) | PancTu-I, Panc89, Colo357 | 5,000 | 50:1–5:1 | Up to 120 | 84 |
PBMCs, BiTE (Epcam×CD3) | PC3 | 10,000 | 10:1 to 0.625:1 | >80 | 32 | |
ADCC | NK-92, patient-derived mononuclear or polynuclear cells, trastuzumab | BT-474 | 5,000–20,000 | 1:1 | 72 | 96 |
ADCC, antibody-dependent cellular cytotoxicity; BiTE, bispecific T-cell engager; CAR, chimeric antigen receptor; E:T, effector to target; MOI, multiplicity of infection; PBMC, peripheral blood mononuclear cell; NK, natural killer.
Flow assay
Unlike the aforementioned assays, the flow assay allows for analysis and quantification of the cytotoxicity of cell subpopulations in heterogenous cell mixtures, providing a unique advantage for the investigation of the differential treatment susceptibility of distinct cell types within a heterogeneous target cell population.35 The flow assay distinguishes between target and effector cells by their properties in size and granularity (by forward and sideward scatter, respectively) and by specific staining of cells with antibodies coupled to a fluorescence dye. For the evaluation of cell death, DNA-intercalating fluorescent agents, such as propidium iodide or 7-aminoactinomycin D, are commonly used, which are preferably taken up by dead cells and undergo a spectral shift upon association with DNA.36 Stages of apoptosis can be assessed using cell staining with Annexin V, which specifically binds to phosphatidylserine that is externalized during apoptotic cell death.37,38 Cell-permeable dyes that react with intracellular free amines, such as carboxyfluorescein succinimidyl ester, which is frequently used in cell proliferation assays, can be used to further distinguish cell populations.35,39
Reports of either target or effector cell staining in conjunction with a cell death or apoptosis marker revealed a positive correlation between the 51Cr assay and the flow assay.35,37,40,41 Liu et al. developed a flow-based assay that measures cytotoxic T lymphocyte–induced caspase activation in target cells using a fluorogenic caspase substrate and reported that the flow assay was more sensitive than the 51Cr assay.42 A similar approach was developed by Packard and Komoriya to measure cell death on the basis of activation of intracellular proteases using cell-permeable fluorogenic substrate probes. This method was used to quantify the entry of granzyme B into target cells as an early event in cell-mediated cytotoxicity43
Flow assays require acquisition and analysis of individual sample data, which renders flow cytometry–based assays laborious unless an automatic workflow is implemented. Even though the intra-assay variability between the flow assay and the 51Cr assay was similar, Motzer and colleagues found no compelling reason to adopt an NK cell flow assay over the 51Cr assay, owing to a greater time requirement for the flow assay.44 However, a recently reported miniaturized and automated flow assay was found to be capable of reliably measuring NK cell–mediated cytotoxicity in a 1,536-well format with a throughput of 50,000 wells per day.45 Owing to the ongoing development of high-throughput multiparameter instruments and mass cytometry at single-cell resolution,46,47 cytotoxicity and cellular behavior can now be studied concurrently in a timely fashion. Martinez et al. reported a high-throughput multiparameter flow assay that combines CAR T cell–mediated killing with concurrent evaluation of CAR T-cell transduction and activation status.48 The capability of the flow assay to simultaneously analyze target cell killing and effector cell phenotype, as well as its ability to distinguish between heterogenous cell populations, further advances the possibility of studying the dynamic relationship between costimulatory and coinhibitory signaling in a multicell environment in a single run. However, one must be cautious in experimental design, as well as the interpretation of multiparametric results beyond cytotoxicity by checking for reproducibility and confirmation by other assays.
Alternative assays
Other nonradioactive methods that are an alternative to the 51Cr assay include assays that directly measure target cell lysis, such as calcein- and europium-release assays. In the former, target cells are loaded with the lipid-soluble acetoxymethyl ester of calcein, which is converted by intracellular esterases to the lipid-insoluble fluorochrome calcein.49 Measurement of calcein in the supernatant is affected by autofluorescence of media components and a significantly higher spontaneous release of calcein, compared with 51Cr, leading to a low signal-to-background ratio.50 Consequently, advanced methods using high-throughput flow cytometry or cell imaging focus on the quantification of fluorescent live cells.51,52 In europium-release assays, the lanthanide europium, loaded into target cells in a complex with diethylenetriaminepentaacetate, is quantified in the supernatant of lysed cells upon transformation into a fluorescent chelate and detection of time-resolved fluorescence.53 Difficulties with target cell labeling accompanied by high spontaneous release of europium have prevented widespread use of this assay.53
Another class of cytotoxicity assays is based on cytosolic enzymes, such as lactate dehydrogenase, which are released from damaged cells and whose activity can be quantified via colorimetric or fluorometric readouts during the enzymatic reaction.54 One glaring limitation of this approach is that dead effector cells will also contribute to the enzyme concentration and measurement of cytotoxicity. As a result, it is suggested that, when using these assays, effector cell viability should remain above 95% to ensure the accuracy of measurements; however, events such as activation-induced cell death of effector cells may still confound the interpretation of results.23
Another approach enumerates effector cell function by means of secreted proteins involved in specific pathways characteristic of cell-mediated cytotoxicity. The enzyme-linked immunospot assay operates under a similar principle as the classic enzyme-linked immunosorbent assay to measure the release of effector cytokines during effector and target coculture.55 Efforts to quantify effector cell responses have been centered around interferon-gamma (INF-γ)56 as a measure of effector cell activation, as well as granzyme B and perforin as key mediators of cell death via the granule-mediated pathway.57,58 Granzyme B and perforin may be more representative surrogate markers for cell-mediated cytotoxicity than INF-γ, as INF-γ is not always found to be secreted by cytotoxic lymphocytes.58 Degranulation of T and NK cells can be further assessed by CD107a flow cytometry.59 CD107a becomes accessible to antibody staining during effector cell degranulation, and its expression is correlated with target cell lysis.60 It is critical to emphasize that this class of assays assesses only effector cell function as an indirect measure of cell-mediated cytotoxicity, whereas assays such as the 51Cr assay directly measure target cell death.
Advanced in vitro antitumor efficacy assays
Whereas in vitro efficacy assays typically investigate treatment modalities under single-effector, single-target, or single-antigen conditions, additional layers of complexity can be introduced to mimic the conditions that CAR T cells and other cell therapies face in the tumor microenvironment in vivo. Inhibition of cytolytic and cytokine secretion functions resulting from chronic antigen exposure–induced T-cell exhaustion can be investigated during repeated antigen stimulation. In this “antigen stress test,” effector cells are harvested and repeatedly transferred to wells with freshly seeded target cells at a defined E:T ratio. Retention or loss of effector function upon repeated antigen stimulation can be quantified using the assays outlined above to compare, for example, the effects of different costimulatory domains on effector cell–mediated cytotoxicity under high antigen stress.4,14,61 With the modification of variables such as E:T ratio, target antigen exposure time, the measurement of intermittent cytotoxicity following specified rounds of antigen exposure, and the addition of rescue immunomodulatory antibodies, antigen stress tests can yield more-comprehensive information than a single-exposure cytotoxicity assay.
Unlike CD19, which is specifically used as a target antigen for tumors of the B-cell lineage, solid-tumor antigen targets are expressed heterogeneously.62 This variable expression and distribution of antigens constitutes a hurdle for cell therapy, as it potentially renders tumor cells with low antigen density less susceptible to treatment and could increase the risk of “on-target/off-tumor” toxicities toward normal tissue with very low expression of the target antigen.63 Assessing the differential cytotoxicity of CAR T cells towards targets with variable antigen expression provides an opportunity to investigate the antigen activation threshold, the correlation between antigen density and cytotoxicity, the additive cytotoxicity on low-antigen targets in the presence of high-antigen targets, and on-target/off-tumor toxicity. In a similar fashion, simultaneously targeting more than one antigen, either by using dual-antigen CAR T cells or by coadministering two or more CAR T-cell constructs (each with single-antigen specificity), presents a strategy to overcome tumor-associated antigen heterogeneity and tumor immune escape.63 Differential cytotoxicity can be assessed through selective labeling of the target cell in a heterogenous mixture of cells in the 51Cr assay, or by transduction of target cells with different fluorophores in the flow assay.
In addition to facing antigen heterogeneity, CAR T cells encounter varied immune cells and tumor cell–secreted inhibitory or excitatory cytokines within the tumor microenvironment. In particular, regulatory T cells and M2 macrophages, among others, can inhibit T-cell function.64 Moreover, soluble factors and cytokines, such as transforming growth factor beta (TGFβ), play a role in modulating treatment response.7 The inclusion of soluble and cellular components into the assay system allows investigation of CAR T-cell efficacy in the presence of immunomodulatory factors. Our laboratory has developed a malignant pleural effusion–derived ex vivo culture system to investigate the efficacy of immunotherapeutic agents, including CAR T cells derived from a patient’s own T cells, in a human, immunocompetent, tumorlike environment65 and to further test the ability of immune modulatory antibodies against PD-1 or TGFβ to rescue CAR T-cell cytotoxicity.
The advanced assays described above allow the investigation of the cytotoxicity of cell therapies under conditions resembling the environment these living drugs face in the tumor microenvironment in vivo. Table 6 provides an overview of the assays discussed in this section. More importantly, after gaining expertise in the conduct and interpretation of the assays described above, investigators can rationally combine the assays to simultaneously investigate transduced T-cell expansion, memory, and exhaustion phenotypic changes and effector cytokine secretion, in addition to cytotoxicity. For example, while conducting repeated antigen stress tests with multiple effector T cells against heterogenous antigen expression targets, in addition to cytotoxicity at predetermined time points, one could assess transduced and untransduced T-cell expansion by cell count or bead assay, phenotypic changes by flow cytometry, and effector cytokine secretion by analysis of supernatants with Luminex assay. A combined interpretation of the results from these assays provides insights into underlying biological changes in addition to cytotoxicity.61
Table 6.
Advanced in vitro antitumor efficacy assays
Assay | Investigation of focus | Description | Recommended assay(s) |
---|---|---|---|
Antigen stress test | Adaptive resistance, cytotoxicity following functional exhaustion | Repeated antigen stimulation by multiple rounds of target cell addition to investigate effector cell cytotoxicity under high antigen stress | 51Cr, BLI, impedance, or flow cytometry assay* |
Differential cytotoxicity on target cells with heterogeneous antigen expression | Cytotoxicity on target cells with antigen heterogeneity (low and high), on-target/off-tumor toxicity on normal cells with very low antigen expression | Cytotoxicity on target cells with variable antigen expression to investigate the correlation between antigen density and cytotoxicity and any unanticipated cytotoxicity of activated effector cells on normal cells with very low antigen expression | If target cells with different antigen expression levels are assessed individually (one cell type per well): 51Cr, BLI, impedance, or flow cytometry assay. If target cells are plated as mixture: flow cytometry assay. |
Multi-CAR cytotoxicity | Antigen heterogeneity, antigen escape | Cytotoxicity assessment of effector cells with multiple CAR constructs targeting different antigens simultaneously | 51Cr, BLI, impedance, or flow cytometry assay |
Cytotoxicity in the presence of soluble factors | Immunosuppression by soluble factors known to be present in the tumor environment | Cytotoxicity in the presence of various doses of single or multiple inhibitory or excitatory cytokines and/or soluble factors | 51Cr, BLI, impedance†, or flow cytometry assay |
Cytotoxicity in the presence of immune cells | Immunosuppression by immune cells known to be present in the tumor immune environment | Cytotoxicity in the presence of various ratios of single or multiple immunomodulatory immune cells | Flow cytometry assay to investigate cytotoxicity against heterogenous (target) cell populations |
It is imperative that the target cells are completely lysed before the effector cells are pooled from the coculture condition and exposed to freshly plated target cells. Transfer of viable target cells to the next round of antigen stimulation might change the E:T ratio, potentially confounding comparison across different effector constructs.
Addition of soluble factors to the impedance assay may impact tumor cell morphology and cell index. It is critical to include a control to account for soluble factor–induced changes in cell index in absence of effector cells. As an example, treatment of target cells with soluble factors such as TGFβ or TNFα can alter the size, adherence, and cell surface receptor expression on target as well as T cells, and it is important to include controls at different concentrations and durations of incubation.
CAR, chimeric antigen receptor.
Potency assays for clinical cell therapy products
The potency requirements for investigational cell therapy products vary with the phase of investigation, and although a potency assay is not required to initiate early-phase clinical studies, a qualified assay to determine the dose is recommended.66 A validated in vitro or in vivo assay that measures the biological activity is required by the end of phase 2.67 Due to the complexity of the mode of action of cellular products, no standardized potency assay exists, but the assay should represent the product’s relevant biological properties.68 Although characteristics such as migration, proliferation, persistence, and specific phenotypes are linked to positive clinical outcomes of adoptive cell therapies, only the cytotoxic activity of the effector cells lead to a reduction in tumor burden and is therefore considered the preferred mode of action for a release assay.69
Despite the vast diversity of cytotoxicity readouts available and presented in this article, the majority of CD19-targeting CAR T-cell therapies in clinical trials utilize the 51Cr assay and INF-γ release as potency assays for product release.70 Fewer studies utilize flow cytometry to determine the percentage of alive target cells after coculture with the final product.71,72 Potency assays for the two FDA-approved CD19-specific CAR T-cell products tisagenlecleucel and axicabtagene ciloleucel measure the percentage of transduced cells and INF-γ production in response to CD19-expressing tumor cells.73–75 However, an FDA briefing document for tisagenlecleucel states that INF-γ production varied greatly from lot to lot in the clinical trials, complicating the correlation of INF-γ production to product safety or efficacy.74 With emerging changes in cell manufacturing, such as the addition of IL-15 or IL-21 or selecting a phenotype, these products are to be validated in vivo in addition to the above mentioned in vitro assays prior to entering the clinic.
Assessment of cell therapies beyond cytotoxicity
Successful immunotherapy is not based only on the cytotoxic activity of engineered immune cells towards the tumor target but includes tumor infiltration and migration, antigen-dependent expansion, cytokine secretion, functional persistence, alleviation of exhaustion, and the recruitment of the innate and adaptive immune system. Therefore, to assess cell therapies in vitro and in vivo, investigators rely on additional assays.76
Antigen-dependent T-cell expansion can be measured by fold-increase in cell number or number of cell divisions. The latter is typically achieved by labeling the effector cells with a fluorescent dye that is equally split between the daughter cells, resulting in a diminished fluorescence with each cell division.77 Persistent exposure to antigen can lead to exhaustion, a state characterized by loss of effector cell function, that is accompanied by an increase in inhibitory receptors such as PD-1, whose expression can be determined by flow cytometry. Furthermore, the differentiation status and certain memory features of the effector cells are associated with antitumor efficacy and persistence. The percentage of memory T cells and terminally differentiated T cells can be measured by staining for CD45RA and CD62L, among other markers.78 Ultimately, functional persistence, an indicator of long-term tumor control, can be assessed by performing tumor rechallenge experiments in vivo.14 Although certain characteristics mentioned above can be established concurrently during repeated antigen stimulations, we encourage the investigator to use orthogonal assays to gather additional performance parameters, considering the specific cell therapy platform utilized and its unique mode of actions.
Discussion
The emerging role of genetically engineered immune cells, oncolytic viruses, and immunomodulatory agents for the treatment of cancer and autoimmune diseases relies upon sensitive and robust in vitro potency assays for the research, development, and manufacturing phases of these living drugs. These assays involve (1) labeling of the target cells (such as 51Cr, live or dead, or antibody staining for markers of interest in flow cytometry), (2) reporter enzyme activity (e.g., luciferase, GFP), (3) cell attachment or detachment (impedance-based assay), or (4) release of compounds from effector cells (e.g., IFN-γ, granzyme B). The underlying physical and biological mechanisms for quantifying drug potency and measuring cell-mediated cytotoxicity differ substantially across the platforms. Whereas, for example, INF-γ– and granzyme B–based assays focus on effector cell function, the 51Cr assay measures target cell death. Since effector cell function does not always relate to target cell death, we focused this review on assays that directly measure target cell lysis.
Owing to the inherent variability in function between different donor effector cells, there are several requirements for cytotoxicity assays to be versatile tools for the study of immunotherapeutic agents. They should ideally possess (1) high specificity and sensitivity, (2) the ability to reflect the mode of action, (3) linearity over a wide concentration or E:T range, (4) robustness, (5) reproducibility, (6) scalability, and (7) automatability. Although the 51Cr assay has historically been the gold standard measurement of cell-mediated cytotoxicity, powerful technologies for cytotoxicity assessment have now been developed that do not rely on radioactive labeling of target cells and provide comparable results, with increased sensitivity and higher sample throughput. As such, the traditional 51Cr assay is starting to lose its primacy. Its signal-to-background ratio and sensitivity have been reported to be inferior to those of the BLI assay,16 impedance-based assay,15 and flow assay.42 A certain limitation of the 51Cr assay is its restriction to measure time points within approximately 24 h, after which specific 51Cr release usually becomes indistinguishable from background release.15 In addition, the high E:T ratios required in the 51Cr assay are not representative of the ratios observed in patients with cancer, who often have a high tumor burden and relatively low numbers of effector cells infiltrating into the tumor.79,80 The BLI assay, impedance-based assay, and flow assay allow for the application of much lower E:T ratios that are physiologically more relevant.
With our laboratory’s focus on solid tumor CAR T-cell therapy translation, we perform an impedance assay that allows repeated antigen stimulation and assessment over at least 4 days following initial 51Cr assay and a focused flow assay based on the results of first two assays. Each assay platform has intrinsic strengths but also intrinsic weaknesses (Figure 1). The decision of which assay to use should be based on the individual properties of the experiment. One should consider the characteristics of the target and effector cells (e.g., primary cells, cell line or cell mixture, adhering or suspension cells, easy- or hard-to-transfect cells), as well as the number of samples to be measured and the level of automation required. The impedance-based assay is unique in that it does not require any cell labeling. The flow assay has the distinct advantage of allowing the analysis of differential cytotoxicity in a heterogenous cell population. Such characteristics set these assays apart from the 51Cr assay and the BLI assay when it comes to hard-to-transfect cells and multiparameter readouts, respectively. Whereas the 51Cr assay is laborious and not suitable for high-throughput applications, the impedance-based and BLI assays can be scaled up to high-density screening formats. Although it has historically been hindered by laborious sample preparation and slow data acquisition, the flow assay can be implemented in a high-throughput workflow dedicated to accelerating phenotype-based drug discovery.45
Cytotoxicity assays compatible with downstream applications and automated workflows, such as the BLI, impedance-based, and flow assays, have the capacity to accelerate the discovery of novel living drug modalities in a high-throughput manner. With increased knowledge of the agents being used to augment synthetic immunity, these assays can aid in the assessment of immune cell function comprehensively.
Acknowledgments
Funding support: PSA’s laboratory work is supported by grants from the National Institutes of Health (P30 CA008748, R01 CA236615-01, and R01 CA235667), the U.S. Department of Defense (BC132124, LC160212, CA170630, and CA180889), the Batishwa Fellowship, the Comedy vs Cancer Award, the Esophageal Cancer Education Fund, the Memorial Sloan Kettering Technology Development Fund, the Miner Fund for Mesothelioma Research, the Mr. William H. Goodwin and Alice Goodwin, the Commonwealth Foundation for Cancer Research, and the Experimental Therapeutics Center of Memorial Sloan Kettering Cancer Center.
Competing Interests: PSA has received research funding from ATARA Biotherapeutics and Acea Biosciences, has served on the Scientific Advisory Board or as consultant to ATARA Biotherapeutics, Bayer, Carisma Therapeutics, Imugene, and Takeda Therapeutics, and has patents, royalties, and intellectual property on mesothelin-targeted CARs and other T-cell therapies, method for detection of cancer cells using virus, and pending patent applications on T-cell therapies.
References
- 1.Darvin P, Toor SM, Sasidharan Nair V. & Elkord E. Immune checkpoint inhibitors: recent progress and potential biomarkers. Exp Mol Med 50, 1–11 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Slaney CY, Wang P, Darcy PK & Kershaw MH CARs versus BiTEs: a comparison between T cell-redirection strategies for cancer treatment. Cancer Discov 8, 924–934 (2018). [DOI] [PubMed] [Google Scholar]
- 3.Bommareddy PK, Shettigar M. & Kaufman HL Integrating oncolytic viruses in combination cancer immunotherapy. Nat Rev Immunol 18, 498–513 (2018). [DOI] [PubMed] [Google Scholar]
- 4.Grosser R, Cherkassky L, Chintala N. & Adusumilli PS Combination immunotherapy with CAR T Cells and checkpoint blockade for the treatment of solid tumors. Cancer Cell 36, 471–482 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Klichinsky M. et al. Human chimeric antigen receptor macrophages for cancer immunotherapy. Nat Biotechnol 38, 947–953 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Ran FA et al. Genome engineering using the CRISPR-Cas9 system. Nat Protoc 8, 2281–2308 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Kiesgen S, Chicaybam L, Chintala NK & Adusumilli PS Chimeric antigen receptor (CAR) T-cell therapy for thoracic malignancies. J Thorac Oncol 13, 16–26 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Eyquem J. et al. Targeting a CAR to the TRAC locus with CRISPR/Cas9 enhances tumour rejection. Nature 543, 113–117 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Stadtmauer EA et al. CRISPR-engineered T cells in patients with refractory cancer. Science (New York, N.Y.) 367, eaba7365 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Brunner KT, Mauel J, Cerottini JC & Chapuis B. Quantitative assay of the lytic action of immune lymphoid cells on 51-Cr-labelled allogeneic target cells in vitro; inhibition by isoantibody and by drugs. Immunology 14, 181–196 (1968). [PMC free article] [PubMed] [Google Scholar]
- 11.Smith EL et al. GPRC5D is a target for the immunotherapy of multiple myeloma with rationally designed CAR T cells. Sci Transl Med 11 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Daher M. & Rezvani K. Next generation natural killer cells for cancer immunotherapy: the promise of genetic engineering. Curr Opin Immunol 51, 146–153 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Whiteside TL Measurement of cytotoxic activity of NK/LAK cells. Curr Protoc Immunol Chapter 7, Unit 7 18 (2001). [DOI] [PubMed] [Google Scholar]
- 14.Adusumilli PS et al. Regional delivery of mesothelin-targeted CAR T cell therapy generates potent and long-lasting CD4-dependent tumor immunity. Sci Transl Med 6, 261ra151 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Peper JK et al. An impedance-based cytotoxicity assay for real-time and label-free assessment of T-cell-mediated killing of adherent cells. J Immunol Methods 405, 192–198 (2014). [DOI] [PubMed] [Google Scholar]
- 16.Karimi MA et al. Measuring cytotoxicity by bioluminescence imaging outperforms the standard chromium-51 release assay. PLoS One 9, e89357 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Borella P, Bargellini A, Salvioli S, Medici CI & Cossarizza A. The use of non-radioactive chromium as an alternative to 51Cr in NK assay. J Immunol Methods 186, 101–110 (1995). [DOI] [PubMed] [Google Scholar]
- 18.Schafer H, Schafer A, Kiderlen AF, Masihi KN & Burger R. A highly sensitive cytotoxicity assay based on the release of reporter enzymes, from stably transfected cell lines. J Immunol Methods 204, 89–98 (1997). [DOI] [PubMed] [Google Scholar]
- 19.Thorne N, Inglese J. & Auld DS Illuminating insights into firefly luciferase and other bioluminescent reporters used in chemical biology. Chem Biol 17, 646–657 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Fu X. et al. A simple and sensitive method for measuring tumor-specific T cell cytotoxicity. PLoS One 5, e11867 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Rossignol A, Bonnaudet V, Clemenceau B, Vie H. & Bretaudeau L. A high-performance, non-radioactive potency assay for measuring cytotoxicity: A full substitute of the chromium-release assay targeting the regulatory-compliance objective. MAbs 9, 521–535 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Hayek S. et al. Identification of primary natural killer cell modulators by chemical library screening with a luciferase-based functional assay. SLAS Discov 24, 25–37 (2019). [DOI] [PubMed] [Google Scholar]
- 23.Matta H. et al. Development and characterization of a novel luciferase based cytotoxicity assay. Sci Rep 8, 199 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Brown CE et al. Biophotonic cytotoxicity assay for high-throughput screening of cytolytic killing. J Immunol Methods 297, 39–52 (2005). [DOI] [PubMed] [Google Scholar]
- 25.Omokoko TA et al. Luciferase mRNA transfection of antigen presenting cells permits sensitive nonradioactive measurement of cellular and humoral cytotoxicity. J Immunol Res 2016, 9540975 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Abassi YA et al. Label-free, real-time monitoring of IgE-mediated mast cell activation on microelectronic cell sensor arrays. J Immunol Methods 292, 195–205 (2004). [DOI] [PubMed] [Google Scholar]
- 27.Zhu J, Wang X, Xu X. & Abassi YA Dynamic and label-free monitoring of natural killer cell cytotoxic activity using electronic cell sensor arrays. J Immunol Methods 309, 25–33 (2006). [DOI] [PubMed] [Google Scholar]
- 28.Xi B. et al. A real-time potency assay for chimeric antigen receptor T cells targeting solid and hematological cancer cells. J Vis Exp 153 (2019). [DOI] [PubMed] [Google Scholar]
- 29.Kho D. et al. Application of xCELLigence RTCA biosensor technology for revealing the profile and window of drug responsiveness in real time. Biosensors (Basel) 5, 199–222 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Davenport AJ et al. CAR-T cells inflict sequential killing of multiple tumor target cells. Cancer Immunol Res 3, 483–494 (2015). [DOI] [PubMed] [Google Scholar]
- 31.Martinez-Serra J. et al. xCELLigence system for real-time label-free monitoring of growth and viability of cell lines from hematological malignancies. Onco Targets Ther 7, 985–994 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Cerignoli F. et al. In vitro immunotherapy potency assays using real-time cell analysis. PLoS One 13, e0193498 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Hillger JM, Lieuw WL, Heitman LH & AP IJ Label-free technology and patient cells: from early drug development to precision medicine. Drug Discov Today 22, 1808–1815 (2017). [DOI] [PubMed] [Google Scholar]
- 34.Erskine CL, Henle AM & Knutson KL Determining optimal cytotoxic activity of human Her2neu specific CD8 T cells by comparing the Cr51 release assay to the xCELLigence system. J Vis Exp 66, e3683 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Jedema I, van der Werff NM, Barge RM, Willemze R. & Falkenburg JH New CFSE-based assay to determine susceptibility to lysis by cytotoxic T cells of leukemic precursor cells within a heterogeneous target cell population. Blood 103, 2677–2682 (2004). [DOI] [PubMed] [Google Scholar]
- 36.Riccardi C. & Nicoletti I. Analysis of apoptosis by propidium iodide staining and flow cytometry. Nat Protoc 1, 1458–1461 (2006). [DOI] [PubMed] [Google Scholar]
- 37.Aubry JP et al. Annexin V used for measuring apoptosis in the early events of cellular cytotoxicity. Cytometry 37, 197–204 (1999). [DOI] [PubMed] [Google Scholar]
- 38.Logue SE, Elgendy M. & Martin SJ Expression, purification and use of recombinant annexin V for the detection of apoptotic cells. Nat Protoc 4, 1383–1395 (2009). [DOI] [PubMed] [Google Scholar]
- 39.Jin Q. et al. Rapid flow cytometry-based assay for the evaluation of gammadelta T cell-mediated cytotoxicity. Mol Med Rep 17, 3555–3562 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Ozdemir O, Ravindranath Y. & Savasan S. Cell-mediated cytotoxicity evaluation using monoclonal antibody staining for target or effector cells with annexinV/propidium iodide colabeling by fluorosphere-adjusted counts on three-color flow cytometry. Cytometry A 56, 53–60 (2003). [DOI] [PubMed] [Google Scholar]
- 41.Goldberg JE, Sherwood SW & Clayberger C. A novel method for measuring CTL and NK cell-mediated cytotoxicity using annexin V and two-color flow cytometry. J Immunol Methods 224, 1–9 (1999). [DOI] [PubMed] [Google Scholar]
- 42.Liu L. et al. Visualization and quantification of T cell-mediated cytotoxicity using cell-permeable fluorogenic caspase substrates. Nat Med 8, 185–189 (2002). [DOI] [PubMed] [Google Scholar]
- 43.Packard BZ & Komoriya A. Intracellular protease activation in apoptosis and cell-mediated cytotoxicity characterized by cell-permeable fluorogenic protease substrates. Cell Res 18, 238–247 (2008). [DOI] [PubMed] [Google Scholar]
- 44.Motzer SA, Tsuji J, Hertig V, Johnston SK & Scanlan J. Natural killer cell cytotoxicity: a methods analysis of 51chromium release versus flow cytometry. Biol Res Nurs 5, 142–152 (2003). [DOI] [PubMed] [Google Scholar]
- 45.Joslin J. et al. A fully automated high-throughput flow cytometry screening system enabling phenotypic drug discovery. SLAS Discov 23, 697–707 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Spitzer MH & Nolan GP Mass cytometry: single cells, many features. Cell 165, 780–791 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Brummelman J. et al. Development, application and computational analysis of high-dimensional fluorescent antibody panels for single-cell flow cytometry. Nat Protoc 14, 1946–1969 (2019). [DOI] [PubMed] [Google Scholar]
- 48.Martinez EM et al. High-throughput flow cytometric method for the simultaneous measurement of CAR-T cell characterization and cytotoxicity against solid tumor cell lines. SLAS Discov 23, 603–612 (2018). [DOI] [PubMed] [Google Scholar]
- 49.Lichtenfels R, Biddison WE, Schulz H, Vogt AB & Martin R. CARE-LASS (calcein-release-assay), an improved fluorescence-based test system to measure cytotoxic T lymphocyte activity. J Immunol Methods 172, 227–239 (1994). [DOI] [PubMed] [Google Scholar]
- 50.Neri S, Mariani E, Meneghetti A, Cattini L. & Facchini A. Calcein-acetyoxymethyl cytotoxicity assay: standardization of a method allowing additional analyses on recovered effector cells and supernatants. Clin Diagn Lab Immunol 8, 1131–1135 (2001). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Gillissen MA et al. The modified FACS calcein AM retention assay: A high throughput flow cytometer based method to measure cytotoxicity. J Immunol Methods 434, 16–23 (2016). [DOI] [PubMed] [Google Scholar]
- 52.Fassy J, Tsalkitzi K, Salavagione E, Hamouda-Tekaya N. & Braud VM A real-time digital bio-imaging system to quantify cellular cytotoxicity as an alternative to the standard chromium-51 release assay. Immunology 150, 489–494 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.von Zons P. et al. Comparison of europium and chromium release assays: cytotoxicity in healthy individuals and patients with cervical carcinoma. Clin Diagn Lab Immunol 4, 202–207 (1997). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Decker T. & Lohmann-Matthes ML A quick and simple method for the quantitation of lactate dehydrogenase release in measurements of cellular cytotoxicity and tumor necrosis factor (TNF) activity. J Immunol Methods 115, 61–69 (1988). [DOI] [PubMed] [Google Scholar]
- 55.Janetzki S. et al. Guidelines for the automated evaluation of Elispot assays. Nat Protoc 10, 1098–1115 (2015). [DOI] [PubMed] [Google Scholar]
- 56.Streeck H, Frahm N. & Walker BD The role of IFN-gamma Elispot assay in HIV vaccine research. Nat Protoc 4, 461–469 (2009). [DOI] [PubMed] [Google Scholar]
- 57.Shafer-Weaver K. et al. The Granzyme B ELISPOT assay: an alternative to the 51Cr-release assay for monitoring cell-mediated cytotoxicity. J Transl Med 1, 14 (2003). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Malyguine AM, Strobl S, Dunham K, Shurin MR & Sayers TJ ELISPOT assay for monitoring cytotoxic T lymphocytes (CTL) activity in cancer vaccine clinical trials. Cells 1, 111–126 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Aktas E, Kucuksezer UC, Bilgic S, Erten G. & Deniz G. Relationship between CD107a expression and cytotoxic activity. Cell Immunol 254, 149–154 (2009). [DOI] [PubMed] [Google Scholar]
- 60.Alter G, Malenfant JM & Altfeld M. CD107a as a functional marker for the identification of natural killer cell activity. J Immunol Methods 294, 15–22 (2004). [DOI] [PubMed] [Google Scholar]
- 61.Cherkassky L. et al. Human CAR T cells with cell-intrinsic PD-1 checkpoint blockade resist tumor-mediated inhibition. J Clin Invest 126, 3130–3144 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Morello A, Sadelain M. & Adusumilli PS Mesothelin-targeted CARs: driving T cells to solid tumors. Cancer Discov 6, 133–146 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Chen N, Li X, Chintala NK, Tano ZE & Adusumilli PS Driving CARs on the uneven road of antigen heterogeneity in solid tumors. Curr Opin Immunol 51, 103–110 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Guha P, Reha J. & Katz SC Immunosuppression in liver tumors: opening the portal to effective immunotherapy. Cancer Gene Ther 24, 114–120 (2017). [DOI] [PubMed] [Google Scholar]
- 65.Tano Z. et al. MA06. 06 An ex-vivo patient-derived, immunocompetent (PDI) culture system to evaluate immunotherapeutic agents’ anti-tumor efficacy. J Thorac Oncol 13, S376 (2018). [Google Scholar]
- 66.Food and Drug Administration. Chemistry, Manufacturing, and Control (CMC) Information for Human Gene Therapy Investigational New Drug Applications (INDs): Guidance for Industry. 2020. Available at: https://www.fda.gov/media/113760/download. Accessed September 18, 2020.
- 67.Food and Drug Administration. Guidance for FDA Reviewers and Sponsors: Content and Review of Chemistry, Manufacturing, and Control (CMC) Information for Human Somatic Cell Therapy Investigational New Drug Applications (INDs). 2008. https://www.fda.gov/regulatory-information/search-fda-guidance-documents/content-and-review-chemistry-manufacturing-and-control-cmc-information-human-somatic-cell-therapy. Accessed September 18, 2020.
- 68.Food and Drug Administration. Guidance for Industry: Potency Tests for Cellular and Gene Therapy Products. 2011. Available at: https://www.fda.gov/media/79856/download. Accessed September 18, 2020.
- 69.de Wolf C, van de Bovenkamp M. & Hoefnagel M. Regulatory perspective on in vitro potency assays for human T cells used in anti-tumor immunotherapy. Cytotherapy 20, 601–622 (2018). [DOI] [PubMed] [Google Scholar]
- 70.Kassim S. Toward an integrated model of product characterization for CAR-T cell therapy drug development efforts. Cell Gene Ther Insight 3, 227–237 (2017). [Google Scholar]
- 71.Castella M. et al. Point-of-care CAR T-Cell production (ARI-0001) using a closed semi-automatic bioreactor: experience from an academic phase I clinical trial. Front Immunol 11, 482 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Jackson Z. et al. Automated manufacture of autologous CD19 CAR-T cells for treatment of non-Hodgkin lymphoma. Front Immunol 11, 1941 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Food and Drug Administration. Summary Basis for Regulatory Action. BLA 125643. YESCARTA. 2017. Available at: https://www.fda.gov/media/108788/download. Accessed September 18, 2020.
- 74.Food and Drug Administration. FDA Briefing Document: Oncologic Drugs Advisory Committee Meeting. BLA 125646: Tisagenlecleucel, Novartis Pharmaceuticals Corporation. Available at: https://www.fda.gov/media/106081/download. Accessed September 18, 2020. [Google Scholar]
- 75.Tyagarajan S, Spencer T. & Smith J. Optimizing CAR-T cell manufacturing processes during pivotal clinical trials. Mol Ther Methods Clin Dev 16, 136–144 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Rafiq S. et al. Optimized T-cell receptor-mimic chimeric antigen receptor T cells directed toward the intracellular Wilms Tumor 1 antigen. Leukemia 31, 1788–1797 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Quah BJ & Parish CR The use of carboxyfluorescein diacetate succinimidyl ester (CFSE) to monitor lymphocyte proliferation. J Vis Exp 44, 2259 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Feucht J. et al. Calibration of CAR activation potential directs alternative T cell fates and therapeutic potency. Nature Med 25, 82–88 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Eerola AK, Soini Y. & Paakko P. A high number of tumor-infiltrating lymphocytes are associated with a small tumor size, low tumor stage, and a favorable prognosis in operated small cell lung carcinoma. Clin Cancer Res 6, 1875–1881 (2000). [PubMed] [Google Scholar]
- 80.Steele KE et al. Measuring multiple parameters of CD8+ tumor-infiltrating lymphocytes in human cancers by image analysis. J Immunother Cancer 6, 20 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Hillerdal V, Nilsson B, Carlsson B, Eriksson F. & Essand M. T cells engineered with a T cell receptor against the prostate antigen TARP specifically kill HLA-A2+ prostate and breast cancer cells. Proc Natl Acad Sci U S A 109, 15877–15881 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Tseng CT & Klimpel GR Binding of the hepatitis C virus envelope protein E2 to CD81 inhibits natural killer cell functions. J Exp Med 195, 43–49 (2002). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Bhat R, Dempe S, Dinsart C. & Rommelaere J. Enhancement of NK cell antitumor responses using an oncolytic parvovirus. Int J Cancer 128, 908–919 (2011). [DOI] [PubMed] [Google Scholar]
- 84.Oberg HH et al. Novel bispecific antibodies increase gammadelta T-cell cytotoxicity against pancreatic cancer cells. Cancer Res 74, 1349–1360 (2014). [DOI] [PubMed] [Google Scholar]
- 85.Clemenceau B. et al. Antibody-dependent cellular cytotoxicity (ADCC) is mediated by genetically modified antigen-specific human T lymphocytes. Blood 107, 4669–4677 (2006). [DOI] [PubMed] [Google Scholar]
- 86.Kurai J. et al. Antibody-dependent cellular cytotoxicity mediated by cetuximab against lung cancer cell lines. Clin Cancer Res 13, 1552–1561 (2007). [DOI] [PubMed] [Google Scholar]
- 87.Joshi T. et al. The PtdIns 3-kinase/Akt pathway regulates macrophage-mediated ADCC against B cell lymphoma. PLoS One 4, e4208 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Rigo V. et al. Combined immunotherapy with anti-PDL-1/PD-1 and anti-CD4 antibodies cures syngeneic disseminated neuroblastoma. Sci Rep 7, 14049 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Liu C. et al. Expansion of spleen myeloid suppressor cells represses NK cell cytotoxicity in tumor-bearing host. Blood 109, 4336–4342 (2007). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Chen X. et al. A combinational therapy of EGFR-CAR NK cells and oncolytic herpes simplex virus 1 for breast cancer brain metastases. Oncotarget 7, 27764–27777 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Nazarian AA et al. Characterization of bispecific T-cell Engager (BiTE) antibodies with a high-capacity T-cell dependent cellular cytotoxicity (TDCC) assay. J Biomol Screen 20, 519–527 (2015). [DOI] [PubMed] [Google Scholar]
- 92.Martin-Manso G. et al. Thrombospondin 1 promotes tumor macrophage recruitment and enhances tumor cell cytotoxicity of differentiated U937 cells. Cancer Res 68, 7090–7099 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Cook KL et al. Hydroxychloroquine inhibits autophagy to potentiate antiestrogen responsiveness in ER+ breast cancer. Clin Cancer Res 20, 3222–3232 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.El-Andaloussi N. et al. Generation of an adenovirus-parvovirus chimera with enhanced oncolytic potential. J Virol 86, 10418–10431 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Fajardo CA et al. Oncolytic adenoviral delivery of an EGFR-targeting T-cell engager improves antitumor efficacy. Cancer Res 77, 2052–2063 (2017). [DOI] [PubMed] [Google Scholar]
- 96.Kute TE et al. Breast tumor cells isolated from in vitro resistance to trastuzumab remain sensitive to trastuzumab anti-tumor effects in vivo and to ADCC killing. Cancer Immunol Immunother 58, 1887–1896 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Hermans IF et al. The VITAL assay: a versatile fluorometric technique for assessing CTL- and NKT-mediated cytotoxicity against multiple targets in vitro and in vivo. J Immunol Methods 285, 25–40 (2004). [DOI] [PubMed] [Google Scholar]
- 98.Yang ZZ, Novak AJ, Ziesmer SC, Witzig TE & Ansell SM Attenuation of CD8(+) T-cell function by CD4(+)CD25(+) regulatory T cells in B-cell non-Hodgkin’s lymphoma. Cancer Res 66, 10145–10152 (2006). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Kasatori N, Ishikawa F, Ueyama M. & Urayama T. A differential assay of NK-cell-mediated cytotoxicity in K562 cells revealing three sequential membrane impairment steps using three-color flow-cytometry. J Immunol Methods 307, 41–53 (2005). [DOI] [PubMed] [Google Scholar]
- 100.Hoppner M, Luhm J, Schlenke P, Koritke P. & Frohn C. A flow-cytometry based cytotoxicity assay using stained effector cells in combination with native target cells. J Immunol Methods 267, 157–163 (2002). [DOI] [PubMed] [Google Scholar]
- 101.Bracher M, Gould HJ, Sutton BJ, Dombrowicz D. & Karagiannis SN Three-colour flow cytometric method to measure antibody-dependent tumour cell killing by cytotoxicity and phagocytosis. J Immunol Methods 323, 160–171 (2007). [DOI] [PubMed] [Google Scholar]
- 102.Oberst MD et al. CEA/CD3 bispecific antibody MEDI-565/AMG 211 activation of T cells and subsequent killing of human tumors is independent of mutations commonly found in colorectal adenocarcinomas. MAbs 6, 1571–1584 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Yamashita M. et al. A novel method for evaluating antibody-dependent cell-mediated cytotoxicity by flowcytometry using cryopreserved human peripheral blood mononuclear cells. Sci Rep 6, 19772 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]