Elotuzumab (Elo) is an IgG1 monoclonal antibody targeting SLAMF7 (CS1, CRACC, and CD319), which is highly expressed on multiple myeloma (MM) cells, natural killer (NK) cells, and subsets of other leukocytes. By engaging with FcγRIIIA (CD16), Elo promotes potent NK cell–mediated antibody-dependent cellular cytotoxicity (ADCC) and macrophage-mediated antibody-dependent cellular phagocytosis (ADCP) toward SLAMF7+ MM tumor cells. Relapsed/refractory MM patients treated with the combination of Elo, lenalidomide, and dexamethasone have improved progression-free survival. We previously showed that Elo enhances NK cell activity via a costimulation mechanism, independent of CD16 binding. Here, we further studied the effect of Elo on cytotoxicity of CD16-negative NK-92 cells. Elo, but not other SLAMF7 antibodies, uniquely enhanced cytotoxicity mediated by CD16-negative NK-92 cells toward SLAMF7+ target cells. Furthermore, this CD16-independent enhancement of cytotoxicity required expression of SLAMF7 containing the full cytoplasmic domain in the NK cells, implicating costimulatory signaling. The CD16-independent costimulation by Elo was associated with increased expression of NKG2D, ICAM-1, and activated LFA-1 on NK cells, and enhanced cytotoxicity was partially reduced by NKG2D blocking antibodies. In addition, an Fc mutant form of Elo that cannot bind CD16 promoted cytotoxicity of SLAMF7+ target cells by NK cells from most healthy donors, especially if previously cultured in IL2. We conclude that in addition to promoting NK cell–mediated ADCC (CD16-dependent) responses, Elo promoted SLAMF7–SLAMF7 interactions in a CD16-independent manner to enhance NK cytotoxicity toward MM cells.

Multiple myeloma (MM) is a bone marrow plasma cell neoplasm and second most common hematologic malignancy (1). Immunomodulatory drugs (thalidomide, lenalidomide, and pomalidomide), proteasome inhibitors (bortezomib, carfilzomib, and ixazomib), and autologous stem cell transplantation have significantly improved patient survival (2–8). However, MM remains largely incurable, as most patients relapse and become refractory to existing treatments (9).

Elotuzumab (Elo) is an IgG1 monoclonal antibody targeting signaling lymphocytic activation molecule family member 7 (SLAMF7), which is expressed on tumor cells in 95% of MM cases (10–12). SLAMF7 is also found on immune cells, including natural killer (NK) cells, monocytes, and macrophages (13–16). Elo generates effective responses toward MM in preclinical studies in vitro and in vivo (10, 11, 17) and improves progression-free survival (PFS) of relapsed/refractory (RR)MM patients when administered as an immunotherapeutic in combination with lenalidomide/dexamethasone (17, 18). Elo plus pomalidomide/dexamethasone also significantly improves PFS compared with pomalidomide/dexamethasone alone (19). Antitumor effects result from several innate immune cell activation mechanisms: (i) NK cell–mediated antibody-dependent cellular cytotoxicity (ADCC) through FcγRIIIA (CD16), (ii) FcγR-dependent macrophage-mediated antibody-dependent cellular phagocytosis (ADCP), and (iii) CD16-independent costimulation of NK cells through direct interaction with SLAMF7 (10, 11, 14, 16, 20–22).

The efficacy of ADCC-inducing antibodies, such as rituximab, in hematologic malignancies is enhanced in patients homozygous for the high-affinity polymorphic variant of CD16 [valine at position 176 (or position 158 if leader sequence is subtracted)] compared with patients with one or two alleles encoding the low-affinity variant with phenylalanine (F) at position 176 (23–25). Accordingly, in a randomized phase II clinical trial of Elo plus bortezomib and dexamethasone, 176V/V homozygous patients have higher PFS compared with 176F/F patients (26).

Like most members of SLAM family receptors, SLAMF7 serves as a self-ligand (27), but it has unique costimulatory function in NK cells (28). SLAMF7 contains an intracellular immunoreceptor tyrosine-based switch motif (ITSM), which can recruit the cytosolic EAT-2 adaptor protein (29). NK cells express EAT-2, which mediates intracellular costimulatory signaling by SLAMF7, but plasma and MM cells do not express EAT-2 and thereby lack SLAMF7 signaling (29–31). Tyrosine phosphorylated EAT-2 recruits PLC-γ1, resulting in calcium mobilization, ERK activation, and enhanced functional responses by NK cells (29, 32). SLAMF7 can also physically interact with Mac-1 to trigger activation signaling in macrophages (13). Alternative mRNA splicing generates SLAMF7-long (L) and SLAMF7-short (S) isoforms (33). SLAMF7-S lacks the ITSM, interaction with EAT-2, and activation signaling.

Previous work showed that Elo promotes cytotoxicity by NK cells independent from ADCC (22) by causing CD16-independent costimulation of NK cells through SLAMF7 (16). Here, we demonstrated that CD16-independent enhancement of cytotoxicity by Elo required SLAMF7 expression on both NK and target cells and required expression of SLAMF7-L in the NK cells. Elo had unique capacity among several SLAMF7 antibodies to enhance cytotoxicity by promoting SLAMF7–SLAMF7 interactions between NK and MM cells. In addition, an Fc mutant form of Elo lacking CD16-binding properties promoted cytotoxicity of MM target cells by primary NK cells from most healthy donors, especially when the NK cells were cultured with IL2.

Cells and cell lines

NK-92 cells were obtained from ATCC in 2005 and cultured in complete α-MEM medium as described (34), supplemented with 100 U/mL of human recombinant IL2 (teceleukin, Hoffman-La Roche Inc.). Cells were passed with fresh medium and IL2 every 3 to 4 days. The cDNAs encoding either high-affinity (176V; GenBank: BC017865.1) or low-affinity (176F; GenBank: NM_000569.6) variants of FcγRIIIA and human SLAMF7-L (GenBank: NM_021181.4) or SLAMF7–S (GenBank: NM_001282592.1) were subcloned into pBMN-NoGFP retroviral vector (35, 36). NK-92 parental cells retrovirally transduced to express either CD16 variant were previously described (34, 37) and always obtained from master stocks. NK-92 SLAMF7 knockout cells were generated using a doxycycline-inducible CRISPR/Cas9 system as previously described (38). Specific guide RNA sequences [sg1 5′-AAAGAGCTGGTCGGTTCCGT-3′, sg2 5′-GAGACACAGGAGGACCATGG-3′ (ref. 39; Integrated DNA Technologies)] were cloned into pLRG to generate lentivirus in combination with pCMV-dR8.74PAX2 and pMD2.G [from Dr. Neil Johnson, Fox Chase Cancer Center (FCCC)] in 293T cells. NK-92 cells were infected twice with doxycycline-inducible blasticidin-resistant Cas9 lentivirus, viable blasticidin-resistant clones were single-cell sorted, and clones with the highest induced Cas9 expression were selected. These were infected with lentivirus containing sgRNA and GFP, GFP+ cells were sorted and cultured with doxycycline for 48 hours, and SLAMF7-deficient cells were sorted. NK-92 SLAMF7 KO cells were then transduced with retrovirus encoding CD16 176V, SLAMF7-L, or SLAMF7-S and sorted for SLAMF7 expression (34).

MM cell lines were cultured in complete RPMI-1640 medium (supplemented with 10% FBS, penicillin/streptomycin, L-glutamine, sodium pyruvate, 10 mmol/L HEPES, and 50 μmol/L 2-mercaptoethanol). MM.1R and RPMI8226 cells were obtained from ATCC at the beginning of the project and reauthenticated in 2018. RPMI8226 were retrovirally transduced to express higher levels of SLAMF7-L (RPMI8226+SLAMF7), and zinc finger nucleases (product number CKOZFND19392-1KT; Sigma-Aldrich) were introduced by nucleofection to disrupt SLAMF7 expression in MM.1R cells, followed by sorting for SLAMF7 expression, as previously described (16). SKOV3 cells were obtained from the FCCC Cell Culture Facility, authenticated in 2015, cultured in complete RPMI-1640 medium, and retrovirally transduced with SLAMF7-L cDNA in pBMN-IRES-NoGFP vector to obtain SLAMF7+ cells after sorting.

An hSLAMF7-TCR-ζ reporter cell line was engineered by transfecting 3A9 mouse T cells with a construct encoding fusion of human (h)SLAMF7 extracellular domain and mouse CD247 (TCR-ζ) transmembrane and intracellular domains in UCOE vector (detailed in Supplementary Fig. S1). Cells were cultured in complete RPMI-1640 supplemented with 10% FBS and 800 μg/mL of hygromycin B. SLAMF7 engagement was assayed by measuring mouse IL2 secretion with ELISA according to the manufacturer (BD OptEIA, #55148).

Cell lines were authenticated using genetic short tandem repeat profiling analysis by IDEXX Bioresearch. Cell lines were thawed fresh every 2 months and found to be free of Mycoplasma at least yearly in assays by the FCCC Cell Culture Facility. Primary NK cells were enriched from healthy donor PBMCs using the EasySep NK cell–negative selection kit (STEMCELL Technologies) and confirmed to be >95% purity (CD45+CD56+CD3; see Supplementary Fig. S2). Healthy donor volunteers provided written informed consent using HIPAA-compliant procedures approved by the Institutional Review Board of FCCC and experiments were performed in accordance with the Declaration of Helsinki principles.

Reagents

SLAMF7 antibodies: Elo, Elo with Fc domain mutations that prevent recognition by CD16 (Elo Fc mut; ref. 16), Elo Fab fragments (produced as recombinant proteins), Elo F(ab')2 fragments (produced by pepsin cleavage), and PDL241 (40) were provided by Bristol-Myers Squibb (BMS). The SLAMF7 antibody ChLuc90, which consists of chimerized human IgG1 Fc fused to mouse CDR variable regions, was provided by AbbVie Inc. (11). Elo Fc mut antibody (provided by BMS) was expressed by the CHO-S cell line cotransfected with vectors pICOFSCneoK (containing a cDNA encoding Elo variable regions) and pODpurIgG1.1f (containing a cDNA encoding IgG1 heavy-chain constant region with L234A-L235E-G237A-A330S-P331S mutations, which abolish interaction with CD16, as previously described, ref. 16). Comparative binding of Elo and Elo Fc mut to myeloma cells is shown in Supplementary Fig. S3. The m24 antibody targeting activated LFA-1 and NKG2D blocking antibody, 1D11, were from BioLegend. The DNAM-1 blocking antibody was from R&D Systems (AF666; polyclonal goat IgG).

Flow cytometry

SLAMF7 expression was assayed by staining with biotin conjugated Elo antibody (EZ-Link NHS-Biotin, Thermo Fisher #20217) and APC–streptavidin (BioLegend, #405207), SLAMF7 antibody conjugated with APC or DL650 (BioLegend, clone 162.1; ref. 41), or isotype control (BioLegend, #400330). Surface expression of 176V and 176F CD16 was assessed with GRM1 antibody (Southern Biotech, #9570-31), although CD16 176V was sometimes assayed with 3G8-APC (BioLegend, #302012). SLAMF7 antibody competition experiments were performed using the CHO cell line transfected to express high levels of SLAMF7-L. Cells (100,000 cells/condition) were incubated with various concentrations of SLAMF7 antibodies (Elo, PDL241, and 162.1) on ice, washed 2× with staining buffer (0.1% Na azide and 1% FBS in HBSS) and incubated with conjugated SLAMF7 antibodies (162.1-PE, BioLegend; Elo-AF647, PDL241-AF647) on ice, washed ×2 with staining buffer and 100 ng/mL of propidium iodide (Invitrogen) in the final wash, and then subjected to analysis on a BD LSR II or Aria II flow cytometer. Data were analyzed using FlowJo software (BD).

Cytotoxicity assay

NK cell cytotoxicity was assessed using the CytoTox 96 nonradioactive cytotoxicity assay (Promega, #G1780), which is a colorimetric assay that measures lactate dehydrogenase released when target cells are lysed, according to the manufacturer's protocol.

xCELLigence assay

The xCELLigence Real-Time Cell Analyzer (RTCA; ACEA Biosciences) detects electrical impedance of adherent cells over time and can be used as a sensitive measure of kinetic cytotoxicity responses by NK cells. The upward trajectory of plots over time is characteristic of growing adherent cells, and downward plots upon addition of NK cells are characteristic of adherent target cell death (Supplementary Fig. S4). SKOV3 cells (5,000/100 μL of complete RPMI-1640) were added into each well of E16 xCELLigence plates (ACEA Biosciences) and cultured overnight at 37°C with 5% CO2. The next day, complete medium was added to control wells, plates were inserted into an xCELLigence RTCA DP unit (ACEA Biosciences) at 37°C with 5% CO2 and baseline impedance was measured. NK-92 cells or purified donor NK cells (100 μL of 0.5 × 106/mL) and SLAMF7 antibodies (Elo, Elo Fc mut, Elo F(ab')2, Elo Fab, PDL241, ChLuc90, 162.1; 10 μg/mL) were added to desired wells. Data were recorded at 10-minute intervals for up to 24 hours. Every condition containing NK cells was performed in duplicate wells within each experiment and a total of 3 to 6 experiments were performed for each condition. Total lysis (100%) was determined by adding lysis buffer (20 mmol/L Tris HCl pH = 8, 2 mmol/L EDTA pH = 8, 150 mmol/L NaCl, 0.2% SDS, 10% Triton X-100) to a well of target cells alone and the percent cytotoxicity for each experimental condition was normalized to this 100% value at the 10-hour time point (Supplementary Fig. S4). All kinetic plots represent the lines generated from means of all values from every experiment at individual time points. Statistical analysis was performed using Student t tests in GraphPad Prism on mean values from the duplicate wells for the percent cytotoxicity at 10 hours within each experiment performed for each condition, as detailed in figure legends, with significance defined as P < 0.05.

ADCC of SLAMF7+ MM cells by elotuzumab is influenced by CD16 polymorphism

We tested the effect of Elo on the cytotoxicity of MM cell lines with various SLAMF7 expression levels by NK-92 cell lines expressing high (CD16-176V) or low (CD16-176F) affinity FcγRIIIA (Fig. 1A). Two matching pairs of MM target cell lines were genetically modified as previously described (16): RPMI8226 cells expressing low endogenous levels of SLAMF7 were retrovirally transduced to express high SLAMF7 levels (RPMI8226 + SLAMF7), and MM.1R cells, which constitutively express high levels of SLAMF7, were treated with zinc finger nucleases to knock out SLAMF7 expression (MM.1R SLAMF7 KO cells; Fig. 1B). Adding low concentrations of Elo (0.001–0.1μg/mL) to CD16-bearing NK-92 cell lines in the presence of MM.1R cells promoted higher cytotoxicity by CD16-176V NK-92 cells, as compared with CD16-176F NK-92 cells. Using higher concentrations of Elo (1–300 μg/mL) stimulated similar maximal response by NK cells expressing the high- or low-affinity polymorphism (Fig. 1C). A similar trend was observed when NK cells were cocultured with RPMI8226 + SLAMF7 cells, although the difference was not statistically significant (Fig. 1D). Elo did not appreciably stimulate cytotoxicity toward MM cells lacking (MM.1R KO) or expressing low levels of SLAMF7 (parental RPMI8226). These results demonstrate that low concentrations of Elo promote more potent NK cell–mediated ADCC toward SLAMF7+ MM cells if the NK cells express high-affinity (176V) CD16, although the response plateaus to a level of cytotoxicity equivalent for CD16-176F at concentrations above 1 μg/mL of Elo.

Figure 1.

Elotuzumab promoted maximal ADCC if NK cells expressed high-affinity CD16 and required SLAMF7 expression on MM cells. A, Comparison of CD16 expression (GRM1 antibody, Southern Biotech) on parental NK-92 cells (CD16; gray) and NK-92 cells transduced to express 176V (solid line) or 176F (dashed) polymorphic variants of CD16, and mean GMFI ± SD from five experiments (bottom). B, SLAMF7 expression before and after the genetic modification of two human MM cell lines (Elo + anti-IgG1 secondary stain). MM.1R cells were transfected with SLAMF7 zinc finger nucleases (ZFN) to genetically deplete SLAMF7 (MM.1R SLAMF7KO; left), and RPMI8226 cells were stably transduced with SLAMF7 cDNA (RPMI8226+SLAMF7; right), and mean GMFI ± SD from five experiments (bottom). Cells were stained with biotinylated Elo plus secondary streptavidin–APC. C and D, Effect of SLAMF7 expression on NK cell–mediated ADCC in the presence of various concentrations of Elo (0.001–300 μg/mL). Values are mean ± S D. Asterisks indicate statistical significance comparing cytotoxicity by NK-92 cells expressing 176V or 176F variants of CD16 using Welch unequal variance t test. *, P < 0.05; n = 5 independent experiments.

Figure 1.

Elotuzumab promoted maximal ADCC if NK cells expressed high-affinity CD16 and required SLAMF7 expression on MM cells. A, Comparison of CD16 expression (GRM1 antibody, Southern Biotech) on parental NK-92 cells (CD16; gray) and NK-92 cells transduced to express 176V (solid line) or 176F (dashed) polymorphic variants of CD16, and mean GMFI ± SD from five experiments (bottom). B, SLAMF7 expression before and after the genetic modification of two human MM cell lines (Elo + anti-IgG1 secondary stain). MM.1R cells were transfected with SLAMF7 zinc finger nucleases (ZFN) to genetically deplete SLAMF7 (MM.1R SLAMF7KO; left), and RPMI8226 cells were stably transduced with SLAMF7 cDNA (RPMI8226+SLAMF7; right), and mean GMFI ± SD from five experiments (bottom). Cells were stained with biotinylated Elo plus secondary streptavidin–APC. C and D, Effect of SLAMF7 expression on NK cell–mediated ADCC in the presence of various concentrations of Elo (0.001–300 μg/mL). Values are mean ± S D. Asterisks indicate statistical significance comparing cytotoxicity by NK-92 cells expressing 176V or 176F variants of CD16 using Welch unequal variance t test. *, P < 0.05; n = 5 independent experiments.

Close modal

Elotuzumab enhances natural cytotoxicity of NK cells in a CD16-independent manner

To follow-up studies showing CD16-independent enhancement of NK cell–mediated natural cytotoxicity by Elo (10, 11, 14, 16, 20–22), we utilized NK-92 cells lacking endogenous CD16 but expressing SLAMF7. In the presence or absence of Elo, knocking out SLAMF7 on MM.1R decreased cytotoxicity by NK-92 cells and overexpressing SLAMF7 on RPMI8226 boosted natural cytotoxicity by NK-92 cells (Fig. 2A), suggesting that SLAMF7–SLAMF7 interactions potentiate cytotoxicity. Interestingly, adding Elo to NK-92 further enhanced cytotoxicity of RPMI8226 + SLAMF7 in a dose-responsive manner (Fig. 2A, bottom). Although it is unclear why this effect was not observed for MM.1R targets, the enhancement toward RPMI8226 + SLAMF7 targets by Elo suggests that this effect may occur toward only a subset of SLAMF7+ MM target cells and further indicates that Elo promotes SLAMF7–SLAMF7 interactions between NK and myeloma cells.

Figure 2.

Elotuzumab enhances natural cytotoxicity by NK cells in a CD16-independent manner. A, Effect of Elo (0.001–300 μg/mL) on natural cytotoxicity of parental NK-92 cells expressing SLAMF7 and lacking CD16 (SLAMF7+, CD16), toward MM.1R (top) and RPMI8226 (bottom) cells. Gray squares are genetically modified MM.1R SLAMF7KO (top) or RPMI83226 + SLAMF7 (bottom) targets. Values are mean ±SD. Overhead bars mark statistical significance between untreated and different Elo concentration groups using paired Student t test. **, P < 0.01; *, P < 0.05; n = 5 independent experiments. B, SLAMF7 expression on parental SKOV3 cells (solid line) and SKOV3 cells that were stably transduced with SLAMF7 cDNA (dashed line) and stained with the 162.1 SLAMF7 antibody (BioLegend), and mean GMFI ± SD from five experiments (bottom). C, Kinetics of parental SKOV3 (top graph) or SKOV3(+SLAMF7; bottom graph) target cell death by parental NK-92 cells (SLAMF7+, CD16) analyzed in xCELLigence assays. n = 4–6. D, Mean, SD, and statistical analysis for percentage target cell death (compared with detergent lysis conditions as 100%) at 10 hours of xCELLigence assays from four to six independent experiments. Overhead bars mark statistical comparison between indicated groups using paired Student t test; **, P < 0.01; *, P < 0.05. E, Schematic of Elo effect on the cytotoxicity of NK cells against SLAMF7+ and SLAMF7 targets.

Figure 2.

Elotuzumab enhances natural cytotoxicity by NK cells in a CD16-independent manner. A, Effect of Elo (0.001–300 μg/mL) on natural cytotoxicity of parental NK-92 cells expressing SLAMF7 and lacking CD16 (SLAMF7+, CD16), toward MM.1R (top) and RPMI8226 (bottom) cells. Gray squares are genetically modified MM.1R SLAMF7KO (top) or RPMI83226 + SLAMF7 (bottom) targets. Values are mean ±SD. Overhead bars mark statistical significance between untreated and different Elo concentration groups using paired Student t test. **, P < 0.01; *, P < 0.05; n = 5 independent experiments. B, SLAMF7 expression on parental SKOV3 cells (solid line) and SKOV3 cells that were stably transduced with SLAMF7 cDNA (dashed line) and stained with the 162.1 SLAMF7 antibody (BioLegend), and mean GMFI ± SD from five experiments (bottom). C, Kinetics of parental SKOV3 (top graph) or SKOV3(+SLAMF7; bottom graph) target cell death by parental NK-92 cells (SLAMF7+, CD16) analyzed in xCELLigence assays. n = 4–6. D, Mean, SD, and statistical analysis for percentage target cell death (compared with detergent lysis conditions as 100%) at 10 hours of xCELLigence assays from four to six independent experiments. Overhead bars mark statistical comparison between indicated groups using paired Student t test; **, P < 0.01; *, P < 0.05. E, Schematic of Elo effect on the cytotoxicity of NK cells against SLAMF7+ and SLAMF7 targets.

Close modal

Sensitive kinetic measurement of adherent target cell cytotoxicity can be performed using xCELLigence RTCA assays (see Materials and Methods). Unfortunately, available MM cell lines are minimally adherent and could not provide a sensitive enough signal in the xCELLigence system, even when wells were coated with adhesion ligands, such as fibronectin. Therefore, we utilized the adherent human ovarian cancer cell line SKOV3, which undergoes minimal natural cytotoxicity by NK-92. We further generated SKOV3 cells that stably expressed SLAMF7 [SKOV3(+SLAMF7); Fig. 2B]. Adding Elo at 0.1 to 1 μg/mL with CD16-expressing NK-92 cells significantly enhanced cytotoxicity of SKOV3(+SLAMF7) target cells to a similar level of cytotoxicity upon addition of trastuzumab, an ADCC-inducing antibody that binds the SKOV3 surface marker, Her2 (Supplementary Fig. S4A and S4B). Adding Elo to a coculture of parental NK-92 cells lacking CD16 and parental SKOV3 cells lacking SLAMF7 expression did not affect target cell death (Fig. 2C, top; 2D, left). Similarly, trastuzumab did not promote cytotoxicity by parental NK-92 cells, due to their lack of CD16 (Fig. 2C, top). In contrast, Elo significantly enhanced killing of SKOV3(+SLAMF7) cells by parental NK-92 cells, but trastuzumab had no effect (Fig. 2C, bottom; 2D, right). To confirm the Fc-independent enhancement of cytotoxicity, the addition of a mutant form of Elo (Elo Fc mut), which has five amino acids altered in the Fc region to abrogate binding to CD16 (see ref. 16), also increased cytotoxicity of SKOV3(+SLAMF7) cells by CD16-deficient NK-92 cells (Supplementary Fig. S5, bottom), and this effect required SLAMF7 expression on the SKOV3 cells (Supplementary Fig. S5, top). Therefore, in the absence of CD16 interactions, Elo still enhances NK cell cytotoxicity apparently by promoting SLAMF7–SLAMF7 interactions between NK and target cells (Fig. 2E), analogous to our results with RPMI8226 + SLAMF7 target cells (Fig. 2A, bottom).

Elotuzumab efficacy required dimeric F(ab')2 composition and SLAMF7-expressing NK cells

To further investigate the observed effect of Elo to promote NK cell cytotoxicity toward SKOV3(+SLAMF7) cells, we generated SLAMF7-knockout NK-92 cells (SLAMF7 KO, CD16-) using a doxycycline-inducible CRISPR/Cas9 system (see Materials and Methods). The loss of SLAMF7 expression was confirmed by flow cytometry (Fig. 3A). Elo had no effect on the cytotoxicity of SLAMF7 KO NK-92 cells toward SKOV3(+SLAMF7) target cells (Fig. 3B and C, top). In contrast, adding Elo to NK cells that expressed SLAMF7 [NK-92(SLAMF7+,CD16)] significantly enhanced the killing of SKOV3(+SLAMF7) cells (Fig. 3B and C, top), consistent with our previous results toward RPMI8226 + SLAMF7 cells (Fig. 2A, bottom). Elo Fc mut had a similar effect on enhancing NK cell cytotoxicity (Fig. 3B and C, bottom). To further rule out a role for the Fc domain of Elo in the response, we generated bivalent F(ab')2 fragments, which had similar capacity to stimulate cytotoxicity as intact Elo (Fig. 3D). Alternatively, monovalent Fab fragments of Elo were incapable of stimulation, demonstrating a requirement for bivalent association with SLAMF7 to stimulate cytotoxicity (Fig. 3D). These results showed that enhanced NK-mediated killing of SLAMF7+ tumor cells by Elo required SLAMF7 expression on NK cells and the bivalent structure of Elo, but was independent of the Fc domain (summarized in Fig. 3E).

Figure 3.

Elotuzumab is unable to enhance natural cytotoxicity of NK cells lacking expression of both SLAMF7 and CD16. A, SLAMF7 expression (162.1 SLAMF7 antibody, BioLegend) on NK-92 control (GFP knockout, SLAMF7+, CD16; solid line) and NK-92 SLAMF7 KO (CD16; dashed line) cells generated using CRISPR/Cas9, and mean GMFI ± SD from five experiments (bottom). B, Cytotoxicity toward SKOV3(+SLAMF7) target cells by NK-92 (SLAMF7+, CD16) and NK-92 (SLAMF7 KO, CD16) cells studied over time in xCELLigence assays with or without Elo (10 μg/mL; top) or Elo Fc mutant (Elo Fc mut; 10 μg/mL; bottom). C, Mean ± SD target cell death at 10 hours from xCELLigence assays for Elo (top) and Elo Fc mut (bottom) toward SKOV3(+SLAMF7) target cells from four to five independent experiments. Overhead bars mark statistical comparison between indicated groups using paired Student t test. **, P < 0.01; *, P < 0.05. D, Cytotoxicity toward SKOV3(+SLAMF7) target cells by NK-92 (SLAMF7+, CD16) cells studied over time in xCELLigence assays (top) and mean ±SD target cell death at 10 hours (bottom) for Elo versus Elo F(ab')2 versus Fab (3 and 10 μg/mL). Overhead bars mark statistics as in C. n = 4 independent experiments. E, Schematic showing requirement for SLAMF7 expression on NK cells for CD16-independent NK cell–mediated cytotoxicity.

Figure 3.

Elotuzumab is unable to enhance natural cytotoxicity of NK cells lacking expression of both SLAMF7 and CD16. A, SLAMF7 expression (162.1 SLAMF7 antibody, BioLegend) on NK-92 control (GFP knockout, SLAMF7+, CD16; solid line) and NK-92 SLAMF7 KO (CD16; dashed line) cells generated using CRISPR/Cas9, and mean GMFI ± SD from five experiments (bottom). B, Cytotoxicity toward SKOV3(+SLAMF7) target cells by NK-92 (SLAMF7+, CD16) and NK-92 (SLAMF7 KO, CD16) cells studied over time in xCELLigence assays with or without Elo (10 μg/mL; top) or Elo Fc mutant (Elo Fc mut; 10 μg/mL; bottom). C, Mean ± SD target cell death at 10 hours from xCELLigence assays for Elo (top) and Elo Fc mut (bottom) toward SKOV3(+SLAMF7) target cells from four to five independent experiments. Overhead bars mark statistical comparison between indicated groups using paired Student t test. **, P < 0.01; *, P < 0.05. D, Cytotoxicity toward SKOV3(+SLAMF7) target cells by NK-92 (SLAMF7+, CD16) cells studied over time in xCELLigence assays (top) and mean ±SD target cell death at 10 hours (bottom) for Elo versus Elo F(ab')2 versus Fab (3 and 10 μg/mL). Overhead bars mark statistics as in C. n = 4 independent experiments. E, Schematic showing requirement for SLAMF7 expression on NK cells for CD16-independent NK cell–mediated cytotoxicity.

Close modal

Elotuzumab uniquely promotes SLAMF7–SLAMF7 interactions

To further study whether Elo promotes SLAMF7–SLAMF7 interactions, a mouse reporter T-cell line expressing a chimeric receptor consisting of human SLAMF7 extracellular domain and mouse CD247 (TCR-ζ) intracellular domain was generated. The hSLAMF7-TCR-ζ reporter cells produced robust levels of mouse IL2 (mIL2) when exposed to plate-bound Elo or three other plate-bound SLAMF7 antibodies (ChLuc90, 162.1, and PDL241; Fig. 4A). We found that Elo, PDL241, and 162.1 bind to distinct epitopes on SLAMF7, because they do not compete (Supplementary Fig. S6). To test for the capacity of these antibodies to mediate SLAMF7–SLAMF7 interactions, the reporter cell line was added to culture wells containing plate-bound recombinant human SLAMF7 in the presence of varying concentrations of each antibody. Elo concentrations of 0.1 to 3 μg/mL were highly effective in inducing mIL2 secretion by the hSLAMF7-TCR-ζ reporter cells, whereas the three other SLAMF7 antibodies (ChLuc90, 162.1, and PDL241) induced very low levels of mIL2 at concentrations of 1 to 3 μg/mL (Fig. 4B). In contrast, incubating hSLAMF7-TCR-ζ reporter cells with Elo did not affect mIL2 secretion in plates coated with a negative control recombinant protein (mesothelin) or if soluble recombinant hSLAMF7 was added (Fig. 4C). These data demonstrate that Elo has unique capacity among these antibodies to orient SLAMF7-TCR-ζ in an appropriate oligomerized form to induce cell signaling through the chimeric reporter construct, apparently by promoting SLAMF7–SLAMF7 interactions with the plate-bound SLAMF7 molecules.

Figure 4.

Elotuzumab uniquely promotes SLAMF7–SLAMF7 interactions. A, SLAMF7-TCRζ reporter cell line was cultured with four different plate-bound SLAMF7 antibodies (Elo, ChuLuc90, PDL241, and 162.1; coated overnight at 10 μg/mL) and mouse IL2 (mIL2) was assayed in culture supernatant. B, Indicated SLAMF7 antibodies or isotype controls (0.01–3 μg/mL) were added to 3A9 SLAMF7-TCRζ reporter cell line in plates coated with recombinant human SLAMF7 (hSLAMF7) overnight, and mIL2 secretion was assayed. C, Soluble Elo (left) or hIgG1 isotype control (right) were added to SLAMF7-TCRζ reporter cell line on plate-bound (PB) recombinant hSLAMF7 or PB mesothelin control protein, or hSLAMF7 was added as soluble protein (+hSLAMF7; right). Plotted as mean ± SEM.

Figure 4.

Elotuzumab uniquely promotes SLAMF7–SLAMF7 interactions. A, SLAMF7-TCRζ reporter cell line was cultured with four different plate-bound SLAMF7 antibodies (Elo, ChuLuc90, PDL241, and 162.1; coated overnight at 10 μg/mL) and mouse IL2 (mIL2) was assayed in culture supernatant. B, Indicated SLAMF7 antibodies or isotype controls (0.01–3 μg/mL) were added to 3A9 SLAMF7-TCRζ reporter cell line in plates coated with recombinant human SLAMF7 (hSLAMF7) overnight, and mIL2 secretion was assayed. C, Soluble Elo (left) or hIgG1 isotype control (right) were added to SLAMF7-TCRζ reporter cell line on plate-bound (PB) recombinant hSLAMF7 or PB mesothelin control protein, or hSLAMF7 was added as soluble protein (+hSLAMF7; right). Plotted as mean ± SEM.

Close modal

The four SLAMF7 antibodies (Elo, ChLuc90, 162.1, and PDL241) were also compared in xCELLigence assays for their capacity to costimulate NK-92 cells through interaction with SLAMF7 and enhance cytotoxicity of SLAMF7+ SKOV3 target cells. Again, only Elo promoted CD16-independent NK cell–mediated cytotoxicity (Fig. 5A; Supplementary Fig. S7A). In fact, the other SLAMF7 antibodies slightly decreased cell death of SKOV3(+SLAMF7) by NK-92 cells (although not to a statistically significant degree). In contrast, the Elo-induced enhancement of cytotoxicity was not evident with SLAMF7 KO NK-92 cells (Fig. 5B; Supplementary Fig. S7B). Taken together, our results indicated that Elo has unique capacity among SLAMF7 antibodies to promote SLAMF7–SLAMF7 interactions between NK cells and target cells, which resulted in enhanced cytotoxicity responses.

Figure 5.

Elotuzumab promotes costimulation-mediated cytotoxicity. Impact of various SLAMF7 antibodies (10 μg/mL each) on cytotoxicity by NK-92(SLAMF7+,CD16) cells (A) or SLAMF7-deficient NK-92 (SLAMF7 KO, CD16) cells (B) toward SKOV3(+SLAMF7) target cells in xCELLigence assays. Mean, SD; and statistics for target cell death are shown at 10 hours from three to four independent xCELLigence assays. Overhead bars mark statistical significance between indicated groups using paired Student t test. **, P < 0.01; *, P < 0.05. ns, not significant. Elo, elotuzumab; CL90, ChLuc90; 162, 162.1; P241, PDL241. C and D, Test of the impact of blocking NKG2D (C) and DNAM1 (D) antibodies on Elo-induced cytotoxicity by NK-92(SLAMF7+, CD16) cells toward SKOV3(+SLAMF7) target cells in xCELLigence assays. Mean, SD; statistics for target cell death at 10 hours from five independent assays using blocking antibodies at 10 μg/mL each. Overhead bars mark statistics as in A and B. Kinetic plots for all of these xCELLigence assays can be found in Supplementary Fig. S5C and S5D.

Figure 5.

Elotuzumab promotes costimulation-mediated cytotoxicity. Impact of various SLAMF7 antibodies (10 μg/mL each) on cytotoxicity by NK-92(SLAMF7+,CD16) cells (A) or SLAMF7-deficient NK-92 (SLAMF7 KO, CD16) cells (B) toward SKOV3(+SLAMF7) target cells in xCELLigence assays. Mean, SD; and statistics for target cell death are shown at 10 hours from three to four independent xCELLigence assays. Overhead bars mark statistical significance between indicated groups using paired Student t test. **, P < 0.01; *, P < 0.05. ns, not significant. Elo, elotuzumab; CL90, ChLuc90; 162, 162.1; P241, PDL241. C and D, Test of the impact of blocking NKG2D (C) and DNAM1 (D) antibodies on Elo-induced cytotoxicity by NK-92(SLAMF7+, CD16) cells toward SKOV3(+SLAMF7) target cells in xCELLigence assays. Mean, SD; statistics for target cell death at 10 hours from five independent assays using blocking antibodies at 10 μg/mL each. Overhead bars mark statistics as in A and B. Kinetic plots for all of these xCELLigence assays can be found in Supplementary Fig. S5C and S5D.

Close modal

Adhesion molecules and NKG2D contributed to elotuzumab-induced NK cell cytotoxicity

To better understand the mechanism by which Elo enhances cytotoxicity in a CD16-independent manner, we tested for upregulation of a variety of activation markers and receptors on NK-92 cells when cultured for 24 hours with SLAMF7+ SKOV3 cells ± Elo. The expression of CD69, NKG2D, CD25, ICAM-1, and the activated form of LFA-1 were significantly increased under these conditions (Supplementary Fig. S8A), demonstrating NK cell activation. Most of these increases required or were more pronounced in the presence of SLAMF7+ SKOV3 target cells. The increased expression of ICAM-1 and activated LFA-1 suggests improved target cell adhesion upon Elo treatment, although we could not detect enhanced conjugation by Elo of NK-92 cells cultured with either MM1.R or RPMI8226 + SLAMF7 target cells (Supplementary Fig. S8B). Nonetheless, upregulation of adhesion molecules by Elo may be playing a modest role in the effect. The increase in NKG2D levels by Elo was also intriguing, particularly when we found that SKOV3 target cells expressed ligands for both NKG2D (ULBPs) and DNAM-1 (Nectin 2 and PVR; Supplementary Fig. S8C). Therefore, we tested whether blocking either NKG2D or DNAM-1 with antibodies could disrupt Elo-induced cytotoxicity. Whereas NKG2D blockade partially reduced the cytotoxicity, DNAM-1 blocking antibodies were ineffective (Fig. 5C and D; Supplementary Fig. S7C and S7D). Taken together, the CD16-independent enhancement of cytotoxicity by Elo appears to be mediated through multiple mechanisms, including the activation of LFA-1, upregulation of ICAM-1 and NKG2D, and potentially upregulation of CD69 and CD137, which can act as activating receptors on NK cells (42, 43).

Elotuzumab enhances NK cell cytotoxicity only if SLAMF7 contains the full cytoplasmic domain

At least two differentially spliced isoforms of SLAMF7 are found in humans, which differ based on long (-L) or short (-S) cytoplasmic domains that contain or lack the ITSM, respectively. SLAMF7-S does not transduce activation signals (33, 44). We reconstituted NK-92 (SLAMF7KO, CD16) cells with these SLAMF7-L or SLAMF7-S isoforms and tested the ability of Elo to stimulate cytotoxicity toward SKOV3 + SLAMF7 cells using the xCELLigence assay. Although SLAMF7-S was expressed at slightly lower levels than SLAMF7-L (Fig. 6A), Elo significantly increased cytotoxicity by NK-92(+SLAMF7-L, CD16) cells, whereas Elo was entirely incapable of potentiating cytotoxicity by NK-92(+SLAMF7-S, CD16) or NK-92(SLAMF7KO,CD16) cells (Fig. 6B and C). The results suggest that the potentiated cytotoxicity required costimulatory signaling within the NK cells, because the cytoplasmic ITSM signaling motif that recruits EAT-2 was only present in the SLAMF7-L isoform.

Figure 6.

Elotuzumab enhances NK cell cytotoxicity only if SLAMF7 contains the full cytoplasmic domain. A, Representative SLAMF7 expression (162.1 antibody, BioLegend) on NK-92 SLAMF7 KO cells (gray) and reconstituted with SLAMF7-L (dashed) or SLAMF7-S (solid line) isoforms, and mean GMFI ± SD from five experiments (bottom). B, Impact of Elo (10 μg/mL) on cytotoxicity by NK-92(SLAMF7 KO, CD16) cells reconstituted with either SLAMF7-L or SLAMF7-S toward SKOV3(+SLAMF7) target cells studied in xCELLigence assays. C, Mean, SD; statistics for target cell death at 10 hours from five independent assays. Overhead bars mark statistical comparisons between indicated groups using paired Student t test. *, P < 0.05.

Figure 6.

Elotuzumab enhances NK cell cytotoxicity only if SLAMF7 contains the full cytoplasmic domain. A, Representative SLAMF7 expression (162.1 antibody, BioLegend) on NK-92 SLAMF7 KO cells (gray) and reconstituted with SLAMF7-L (dashed) or SLAMF7-S (solid line) isoforms, and mean GMFI ± SD from five experiments (bottom). B, Impact of Elo (10 μg/mL) on cytotoxicity by NK-92(SLAMF7 KO, CD16) cells reconstituted with either SLAMF7-L or SLAMF7-S toward SKOV3(+SLAMF7) target cells studied in xCELLigence assays. C, Mean, SD; statistics for target cell death at 10 hours from five independent assays. Overhead bars mark statistical comparisons between indicated groups using paired Student t test. *, P < 0.05.

Close modal

Costimulation of donor NK cell cytotoxicity by Elo Fc mut depends on NK cell activation

To validate our results from NK-92 cells, we used purified healthy donor NK cells to test the CD16-independent effect of Elo Fc mut on promoting natural killing of parental SKOV3 and SKOV3(+SLAMF7) target cells in xCELLigence assays. Neither Elo nor Elo Fc mut was able to promote healthy donor NK cell–mediated killing of parental SLAMF7-deficient SKOV3 cells (Supplementary Fig. S9). In contrast, Elo promoted complete lysis of SKOV3(+SLAMF7) cells within 8 to 10 hours by purified donor NK cells from healthy donor #1 (HD#1; Fig. 7A). On the other hand, Elo Fc mut had no effect on cytotoxicity of SKOV3(+SLAMF7) cells by NK cells from HD#1 (Fig. 7A). After screening seven healthy donors, we observed that Elo Fc mut promoted an increase in cytotoxicity by NK cells from only three of the donors, assessed as percent cytotoxicity at 10 hours in xCELLigence assays (Fig. 7B, left), with HD#8 being a representative example (Fig. 7C, left). Therefore, Elo Fc mut was able to potentiate cytotoxicity of fresh primary human NK cells in a subset of donors (3 of 7).

Figure 7.

Costimulation of healthy donor NK cell cytotoxicity by Elo Fc mut depends on NK cell activation status. A, Elo or Elo Fc mut (10 μg/mL) was added to SKOV3(+SLAMF7) target cells in the presence or absence of freshly purified NK cells from healthy donor (HD) #1 in xCELLigence assays. B, Compilation of cytotoxicity data for n = 7 healthy donors. Percentage target cell death at the 10-hour time point from xCELLigence assays in the absence (−) or presence (+) of Elo Fc mut by purified NK cells toward SKOV3(+SLAMF7) target cells on the day of the purification (left) or after cultured in the presence of recombinant human IL2 (100 U/mL; +) for 48 hours (right). Each patient is designated by a unique icon, and results with or without antibody addition are connected with lines. Overhead bars mark statistical comparison between indicated groups using Wilcoxon matched-pairs signed rank test. C and D, Representative time course data in which purified NK cells from HD#3 and HD#8 were assayed fresh (left) or after cultured in the presence of recombinant human IL2 (100 U/mL) for 48 hours (right).

Figure 7.

Costimulation of healthy donor NK cell cytotoxicity by Elo Fc mut depends on NK cell activation status. A, Elo or Elo Fc mut (10 μg/mL) was added to SKOV3(+SLAMF7) target cells in the presence or absence of freshly purified NK cells from healthy donor (HD) #1 in xCELLigence assays. B, Compilation of cytotoxicity data for n = 7 healthy donors. Percentage target cell death at the 10-hour time point from xCELLigence assays in the absence (−) or presence (+) of Elo Fc mut by purified NK cells toward SKOV3(+SLAMF7) target cells on the day of the purification (left) or after cultured in the presence of recombinant human IL2 (100 U/mL; +) for 48 hours (right). Each patient is designated by a unique icon, and results with or without antibody addition are connected with lines. Overhead bars mark statistical comparison between indicated groups using Wilcoxon matched-pairs signed rank test. C and D, Representative time course data in which purified NK cells from HD#3 and HD#8 were assayed fresh (left) or after cultured in the presence of recombinant human IL2 (100 U/mL) for 48 hours (right).

Close modal

The finding that Elo is therapeutically ineffective in treating RRMM as a single agent (18), yet shows significant efficacy in combination with lenalidomide and dexamethasone (17) suggests that coordinated immune stimulation is important to achieve optimal antitumor effect by Elo. Lenalidomide is an immunomodulatory drug that stimulates NK and T-cell activation, in part by increasing IL2 production by T cells (21, 45). Furthermore, our NK-92 cells are routinely cultured in IL2. Therefore, we hypothesized that culturing NK cells in the presence of IL2 might enhance the capacity of Elo Fc mut to stimulate cytotoxicity. When purified NK cells from HD#3 and HD#8 were assayed after culture for 48 hours in the presence of 100 U/mL recombinant human IL2, Elo Fc mut more potently increased cytotoxicity toward SKOV3(+SLAMF7) cells, as compared with freshly isolated NK cells from these donors (Fig. 7C and D, left vs. right). As summarized in Fig. 7B, whereas Elo Fc mut potentiated cytotoxicity by freshly prepared primary NK cells in only 3 of 7 healthy donors, cytotoxicity was boosted in 5 of 7 healthy donors after culture of the NK cells in IL2. Hence, the CD16-independent costimulation of cytotoxicity by Elo engagement with SLAMF7 can be demonstrated in primary human NK cells, most prominently when NK cells are activated by IL2.

We identified an additional mechanism by which Elo promotes NK cell cytotoxicity toward MM cells independent of CD16-mediated ADCC. SLAMF7 functions as a costimulatory receptor on NK cells that mediates intracellular signaling via recruitment of cytosolic EAT-2 adaptor protein (12, 29). Our previous work demonstrates that Elo can costimulate calcium mobilization responses triggered by the aggregation of NKp46 ± NKG2D activating receptors (16). Here, we demonstrated that Elo can also promote SLAMF7–SLAMF7 interactions between NK and MM cells to enhance NK cell–mediated cytotoxicity through costimulation in addition to facilitating ADCC.

At lower concentrations, Elo (1 ng/mL–1 μg/mL) promoted higher cytotoxicity by NK cells expressing the high-affinity CD16 176V polymorphism versus NK cells expressing the low-affinity 176F polymorphism. In contrast, at higher concentrations of Elo (up to 300 μg/mL; similar to the observed plasma concentrations in Elo-treated patients, which are routinely above 70 μg/mL; ref. 46), no effect of CD16 polymorphism on NK cell ADCC was observed. Despite the lack of differential response at high Elo concentrations, CD16 176V/V homozygous patients respond better to Elo plus bortezomide/dexamethasone compared with 176V/F or 176 F/F patients (26). These results are consistent with better ADCC by NK cells and improved clinical efficacy in response to rituximab in follicular lymphoma patients possessing the CD16 176V/V genotype (23, 47). It is currently unclear if the high-affinity CD16 polymorphism is also beneficial in patients treated with Elo in combination with lenalidomide/dexamethasone.

We further demonstrated that Elo can significantly enhance the natural cytotoxicity of SLAMF7-expressing target cells by CD16-deficient NK-92 cells, and this effect required SLAMF7 expression on both the NK and target cells. This result is consistent with the work of Collins and colleagues, showing that Elo induces CD16-independent target cell killing by NK-92 that requires SLAMF7 expression on the target cells (22). Our work extended their results by showing a requirement for expression of signaling-competent SLAMF7 in the NK cells, indicating a costimulatory signaling mechanism. The requirement for SLAMF7 expression on both effector and target cells suggests that Elo is capable of either (i) bridging SLAMF7 between the two cells or (ii) enhancing the SLAMF7–SLAMF7 interactions between the two cell types. This enhancement only occurred toward SLAMF7+ RPMI8226 and SLAMF7+ SKOV3, but not SLAMF7+ MM.1R myeloma target cells. We hypothesize that Elo engagement with SLAMF7 effectively costimulates additional NK cell–activating receptors engaging with ligands expressed on RPMI8226, but absent on MM.1R. This hypothesis is supported by our previous work showing that Elo engagement with SLAMF7 cannot induce calcium signaling in NK cells on its own, but Elo can potentiate calcium signaling triggered by engagement of the ITAM-linked NK cell receptor, NKp46, either with or without coengagement of NKG2D (16). Consistent with our previous work, we showed here that NKG2D plays at least a partial role in the enhanced cytotoxicity induced by Elo. Of note, the CD16-independent effect occurs at Elo concentrations in the range of 0.1 to 300 μg/mL, whereas ADCC responses are observed in the range of 1 ng/mL to 1 μg/mL. These concentrations are both well below the high serum concentrations achieved for Elo at therapeutically effective dosing regimens (46).

Surprisingly, of four SLAMF7 antibodies tested, only Elo enhanced SLAMF7–SLAMF7-dependent activation responses. This indicates that Elo binds to a unique epitope on SLAMF7, whereby it is either better oriented to effectively bridge the receptor between effector and target cell or the binding induces a conformational change that enhances homotypic interactions between SLAMF7 molecules on opposing cells. Furthermore, we showed that the costimulatory impact of Elo requires bivalent binding to SLAMF7, because F(ab')2 is effective, but Fab fragments are not. All four tested antibodies were found to bind distinct epitopes on SLAMF7. Elo reportedly binds to the membrane proximal C2 domain of SLAMF7, within an epitope encompassing amino acids 170-227 (U.S. patent 7842293B2). In contrast, PDL241 and ChLuc90 are reported to interact with the V domain (U.S. patent 7842293B2 and ref. 40), which mediates homotypic interactions between SLAM family members (48). These two antibodies may thereby block SLAMF7–SLAMF7 interactions, consistent with their capacities to slightly reduce cytotoxicity in our experiments. We could not detect enhanced conjugation between NK-92 cells and MM target cells by Elo or Elo Fc mut using standard flow cytometry–based assays (49). This result is consistent with a previous report that SLAMF7 signaling in YT-S NK cell line could potentiate killing of SLAMF7+ K562 target cells, but not enhance conjugate formation (50). Although the exact mechanism by which Elo promotes SLAMF7 interactions will require further experimentation, Elo has unique capacity to mediate this biological effect. It was shown that Elo can also facilitate macrophages to mediate ADCP toward MM cells in an Fc-dependent manner (14). As SLAMF7 is also highly expressed on subsets of macrophages (notably M1 macrophages; ref. 15), it is possible that Elo could also contribute to macrophage-mediated killing of SLAMF7+ tumors through a CD16-independent mechanism by promoting SLAMF7–SLAMF7 interactions.

We further demonstrated that CD16-independent costimulation by Elo requires the ITSM-containing cytoplasmic domain of SLAMF7 in NK cells, because Elo enhanced cytotoxicity by NK-92 cells reconstituted with SLAMF7-L, but not with the truncated receptor lacking ITSM (SLAMF7-S). Although we cannot rule out that another motif in the cytoplasmic domain is mediating the costimulation, our data suggest that the ITSM is responsible. Previous work showed that NK cell–mediated natural cytotoxicity toward SLAMF7+ target cells is potentiated via tyrosine phosphorylation of the ITSM motif on SLAMF7, which recruits EAT-2 (50) to recruit phospholipase Cγ-1, resulting in intracellular calcium signaling (50). This is consistent with the calcium signaling costimulation by Elo that we previously observed (16). Our results also suggest that extensive SLAMF7-S isoform expression on NK cells from MM patients may affect their clinical outcomes.

We used an Fc mutant form of Elo (Elo Fc mut) to demonstrate CD16-independent enhancement of natural cytotoxicity by NK cells from healthy donors toward SLAMF7+ target cells. Elo Fc mut enhanced cytotoxicity of fresh NK cells from three out of seven healthy donors. Interestingly, Elo Fc mut boosted NK cell cytotoxicity of five of these healthy donors if the cells were cultured for 48 hours in IL2, demonstrating that the CD16-independent effect of Elo on natural cytotoxicity is dependent upon the state of NK cell activation. This finding is of significant interest, because Elo is ineffective as a single agent in MM patients, but highly effective when coadministered with lenalidomide and dexamethasone (17). The immunomodulatory agent lenalidomide binds the E3 ubiquitin ligase cereblon and targets the degradation of the IKZF1 and IKZF3 transcription factors, resulting in increased production of IL2 by T cells (51). Therefore, it is interesting to speculate that lenalidomide coadministration with Elo may, at least in part, improve Elo activity toward MM cells by elevating IL2 levels to activate NK cells and thereby increase their susceptibility to Elo-mediated promotion of SLAMF7–SLAMF7 interactions. This is consistent with previous reports demonstrating lenalidomide can enhance Elo-mediated killing of MM cell lines in coculture with PBMCs and this activity can be partially blocked by antibodies neutralizing the effects of IL2 (anti-CD25/IL2R) on NK cells (21). Consistent with this, we also showed that Elo can induce IL2Rα (CD25) on NK cells, which may also contribute to enhanced function by lenalidomide in patients.

Our data further elucidate the mechanism of action of Elo beyond the previously established mechanism of ADCC. We showed that Elo coactivates NK cells and enhances cytotoxicity toward SLAMF7+ tumor cells by promoting SLAMF7–SLAMF7 interactions. Furthermore, these findings are specific to Elo compared with other SLAMF7 antibodies tested demonstrating the unique mechanistic attributes of NK coactivation and ADCC dually mediated by Elo.

T. Pazina reports receiving a commercial research grant from Bristol-Myers Squibb. A.Y. Kearney is Scientist II at Bristol-Myers Squibb. N.A. Bezman has ownership interest (including patents) in Bristol-Myers Squibb. M.D. Robbins has ownership interest (including patents) in Bristol-Myers Squibb. A.D. Cohen reports receiving a commercial research grant from Bristol-Myers Squibb, other commercial research support from Novartis, and is a consultant/advisory board member for Bristol-Myers Squibb, Celgene, Janssen, Takeda, Kite Pharma, Oncopeptides, GlaxoSmithKline, Seattle Genetics, and Array Biopharma. K.S. Campbell reports receiving a commercial research grant from Bristol-Myers Squibb and has ownership interest (including patents) in NantKwest. No potential conflicts of interest were disclosed by the other authors.

Conception and design: T. Pazina, A.M. James, A. Jhatakia, N.A. Bezman, M.D. Robbins, A.D. Cohen, K.S. Campbell

Development of methodology: T. Pazina, A.M. James, A. Gale, A. Jhatakia, N.A. Bezman, K.S. Campbell

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): T. Pazina, A.M. James, K.B. Colby, A.Y. Kearney, K.S. Campbell

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): T. Pazina, A.M. James, K.B. Colby, A. Jhatakia, A.Y. Kearney, R.F. Graziano, M.D. Robbins, A.D. Cohen, K.S. Campbell

Writing, review, and/or revision of the manuscript: T. Pazina, Y. Yang, A. Gale, R.F. Graziano, N.A. Bezman, M.D. Robbins, A.D. Cohen, K.S. Campbell

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): A.M. James, Y. Yang, A.Y. Kearney, K.S. Campbell

Study supervision: A.D. Cohen, K.S. Campbell

The authors thank Dr. Audie Rice for fostering initiation of this project, Drs. John Krais and Neil Johnson for help generating the CRISPR/Cas9 SLAMF7 KO NK-92 cell line, Lena Shally and Sanjna Shelukar for technical support, John Dowling for developing cytotoxicity assays using xCELLigence, and the following FCCC core facilities: Cell Culture, Flow Cytometry, Cell Sorting, and High-Throughput Screening. The authors also thank Bristol-Myers Squibb and AbbVie for valuable reagents, Bristol-Myers Squibb for financial support, and the Biological Resources Branch of NCI-Frederick for recombinant human IL2. This study was supported by Collaborative Science Center of Excellence grants CA204-013, CA-204-156, and CA204-214 to A.D. Cohen and K.S. Campbell from Bristol-Myers Squibb, FCCC Bucks County and Main Line Boards of Associates, NIH Training Grant T32 CA9035 (A.M. James), and NCI Comprehensive Cancer Center Support Grant CA06927 (FCCC).

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
Kumar
SK
,
Rajkumar
V
,
Kyle
RA
,
van Duin
M
,
Sonneveld
P
,
Mateos
MV
, et al
Multiple myeloma
.
Nat Rev Dis Primers
2017
;
3
:
17046
.
2.
Goldschmidt
H
,
Lokhorst
HM
,
Mai
EK
,
van der Holt
B
,
Blau
IW
,
Zweegman
S
, et al
Bortezomib before and after high-dose therapy in myeloma: long-term results from the phase III HOVON-65/GMMG-HD4 trial
.
Leukemia
2018
;
32
:
383
90
.
3.
Rajkumar
SV
,
Blood
E
,
Vesole
D
,
Fonseca
R
,
Greipp
PR
,
Eastern Cooperative Oncology G
. 
Phase III clinical trial of thalidomide plus dexamethasone compared with dexamethasone alone in newly diagnosed multiple myeloma: a clinical trial coordinated by the Eastern Cooperative Oncology Group
.
J Clin Oncol
2006
;
24
:
431
6
.
4.
Richardson
PG
,
Weller
E
,
Lonial
S
,
Jakubowiak
AJ
,
Jagannath
S
,
Raje
NS
, et al
Lenalidomide, bortezomib, and dexamethasone combination therapy in patients with newly diagnosed multiple myeloma
.
Blood
2010
;
116
:
679
86
.
5.
Baz
RC
,
Martin
TG
 3rd
,
Lin
HY
,
Zhao
X
,
Shain
KH
,
Cho
HJ
, et al
Randomized multicenter phase 2 study of pomalidomide, cyclophosphamide, and dexamethasone in relapsed refractory myeloma
.
Blood
2016
;
127
:
2561
8
.
6.
Berenson
JR
,
Cartmell
A
,
Bessudo
A
,
Lyons
RM
,
Harb
W
,
Tzachanis
D
, et al
CHAMPION-1: a phase 1/2 study of once-weekly carfilzomib and dexamethasone for relapsed or refractory multiple myeloma
.
Blood
2016
;
127
:
3360
8
.
7.
Kumar
S
,
Moreau
P
,
Hari
P
,
Mateos
MV
,
Ludwig
H
,
Shustik
C
, et al
Management of adverse events associated with ixazomib plus lenalidomide/dexamethasone in relapsed/refractory multiple myeloma
.
Br J Haematol
2017
;
178
:
571
82
.
8.
Ocio
EM
,
Richardson
PG
,
Rajkumar
SV
,
Palumbo
A
,
Mateos
MV
,
Orlowski
R
, et al
New drugs and novel mechanisms of action in multiple myeloma in 2013: a report from the international myeloma working group (IMWG)
.
Leukemia
2014
;
28
:
525
42
.
9.
Siegel
RL
,
Miller
KD
,
Jemal
A
. 
Cancer statistics, 2018
.
CA Cancer J Clin
2018
;
68
:
7
30
.
10.
Hsi
ED
,
Steinle
R
,
Balasa
B
,
Szmania
S
,
Draksharapu
A
,
Shum
BP
, et al
CS1, a potential new therapeutic antibody target for the treatment of multiple myeloma
.
Clin Cancer Res
2008
;
14
:
2775
84
.
11.
Tai
YT
,
Dillon
M
,
Song
W
,
Leiba
M
,
Li
XF
,
Burger
P
, et al
Anti-CS1 humanized monoclonal antibody HuLuc63 inhibits myeloma cell adhesion and induces antibody-dependent cellular cytotoxicity in the bone marrow milieu
.
Blood
2008
;
112
:
1329
37
.
12.
Campbell
KS
,
Cohen
AD
,
Pazina
T
. 
Mechanisms of NK cell activation and clinical activity of the therapeutic SLAMF7 antibody, elotuzumab in multiple myeloma
.
Front Immunol
2018
;
9
:
2551
.
13.
Chen
J
,
Zhong
MC
,
Guo
H
,
Davidson
D
,
Mishel
S
,
Lu
Y
, et al
SLAMF7 is critical for phagocytosis of haematopoietic tumour cells via Mac-1 integrin
.
Nature
2017
;
544
:
493
7
.
14.
Kurdi
AT
,
Glavey
SV
,
Bezman
NA
,
Jhatakia
A
,
Guerriero
JL
,
Manier
S
, et al
Antibody-dependent cellular phagocytosis by macrophages is a novel mechanism of action of elotuzumab
.
Mol Cancer Ther
2018
;
17
:
1454
63
.
15.
Beyer
M
,
Mallmann
MR
,
Xue
J
,
Staratschek-Jox
A
,
Vorholt
D
,
Krebs
W
, et al
High-resolution transcriptome of human macrophages
.
PLoS One
2012
;
7
:
e45466
.
16.
Pazina
T
,
James
AM
,
MacFarlane
AW
,
Bezman
NA
,
Henning
KA
,
Bee
C
, et al
The anti-SLAMF7 antibody elotuzumab mediates NK cell activation through both CD16-dependent and –independent mechanisms
.
Oncoimmunology
2017
;
6
:
e1339853
.
17.
Lonial
S
,
Dimopoulos
M
,
Palumbo
A
,
White
D
,
Grosicki
S
,
Spicka
I
, et al
Elotuzumab therapy for relapsed or refractory multiple myeloma
.
N Engl J Med
2015
;
373
:
621
31
.
18.
Zonder
JA
,
Mohrbacher
AF
,
Singhal
S
,
van Rhee
F
,
Bensinger
WI
,
Ding
H
, et al
A phase 1, multicenter, open-label, dose escalation study of elotuzumab in patients with advanced multiple myeloma
.
Blood
2012
;
120
:
552
9
.
19.
Dimopoulos
MA
,
Dytfeld
D
,
Grosicki
S
,
Moreau
P
,
Takezako
N
,
Hori
M
, et al
Elotuzumab plus pomalidomide and dexamethasone for multiple myeloma
.
New Engl J Med
2018
;
379
:
1811
22
.
20.
van Rhee
F
,
Szmania
SM
,
Dillon
M
,
van Abbema
AM
,
Li
X
,
Stone
MK
, et al
Combinatorial efficacy of anti-CS1 monoclonal antibody elotuzumab (HuLuc63) and bortezomib against multiple myeloma
.
Mol Cancer Ther
2009
;
8
:
2616
24
.
21.
Balasa
B
,
Yun
R
,
Belmar
NA
,
Fox
M
,
Chao
DT
,
Robbins
MD
, et al
Elotuzumab enhances natural killer cell activation and myeloma cell killing through interleukin-2 and TNF-alpha pathways
.
Cancer Immunol Immunother
2015
;
64
:
61
73
.
22.
Collins
SM
,
Bakan
CE
,
Swartzel
GD
,
Hofmeister
CC
,
Efebera
YA
,
Kwon
H
, et al
Elotuzumab directly enhances NK cell cytotoxicity against myeloma via CS1 ligation: evidence for augmented NK cell function complementing ADCC
.
Cancer Immunol Immunother
2013
;
62
:
1841
9
.
23.
Cartron
G
,
Dacheux
L
,
Salles
G
,
Solal-Celigny
P
,
Bardos
P
,
Colombat
P
, et al
Therapeutic activity of humanized anti-CD20 monoclonal antibody and polymorphism in IgG Fc receptor FcgammaRIIIa gene
.
Blood
2002
;
99
:
754
8
.
24.
Koene
HR
,
Kleijer
M
,
Algra
J
,
Roos
D
,
von dem Borne
AE
,
de Haas
M
. 
Fc gammaRIIIa-158V/F polymorphism influences the binding of IgG by natural killer cell Fc gammaRIIIa, independently of the Fc gammaRIIIa-48L/R/H phenotype
.
Blood
1997
;
90
:
1109
14
.
25.
Wu
J
,
Edberg
JC
,
Redecha
PB
,
Bansal
V
,
Guyre
PM
,
Coleman
K
, et al
A novel polymorphism of FcgammaRIIIa (CD16) alters receptor function and predisposes to autoimmune disease
.
J Clin Invest
1997
;
100
:
1059
70
.
26.
Jakubowiak
A
,
Offidani
M
,
Pegourie
B
,
De La Rubia
J
,
Garderet
L
,
Laribi
K
, et al
Randomized phase 2 study: elotuzumab plus bortezomib/dexamethasone vs bortezomib/dexamethasone for relapsed/refractory MM
.
Blood
2016
;
127
:
2833
40
.
27.
Cannons
JL
,
Tangye
SG
,
Schwartzberg
PL
. 
SLAM family receptors and SAP adaptors in immunity
.
Annu Rev Immunol
2011
;
29
:
665
705
.
28.
Kumaresan
PR
,
Lai
WC
,
Chuang
SS
,
Bennett
M
,
Mathew
PA
. 
CS1, a novel member of the CD2 family, is homophilic and regulates NK cell function
.
Mol Immunol
2002
;
39
:
1
8
.
29.
Cruz-Munoz
ME
,
Dong
Z
,
Shi
X
,
Zhang
S
,
Veillette
A
. 
Influence of CRACC, a SLAM family receptor coupled to the adaptor EAT-2, on natural killer cell function
.
Nat Immunol
2009
;
10
:
297
305
.
30.
Tassi
I
,
Colonna
M
. 
The cytotoxicity receptor CRACC (CS-1) recruits EAT-2 and activates the PI3K and phospholipase Cgamma signalling pathways in human NK cells
.
J Immunol
2005
Dec 15;
175
:
7996
8002
.
31.
Guo
H
,
Cruz-Munoz
ME
,
Wu
N
,
Robbins
M
,
Veillette
A
. 
Immune cell inhibition by SLAMF7 is mediated by a mechanism requiring src kinases, CD45, and SHIP-1 that is defective in multiple myeloma cells
.
Mol Cell Biol
2015
;
35
:
41
51
.
32.
Tassi
I
,
Colonna
M
. 
The cytotoxicity receptor CRACC (CS-1) recruits EAT-2 and activates the PI3K and phospholipase Cgamma signaling pathways in human NK cells
.
J Immunol
2005
;
175
:
7996
8002
.
33.
Lee
JK
,
Boles
KS
,
Mathew
PA
. 
Molecular and functional characterization of a CS1 (CRACC) splice variant expressed in human NK cells that does not contain immunoreceptor tyrosine-based switch motifs
.
Eur J Immunol
2004
;
34
:
2791
9
.
34.
Miah
SM
,
Campbell
KS
. 
Expression of cDNAs in human Natural Killer cell lines by retroviral transduction
.
Methods Mol Biol
2010
;
612
:
199
208
.
35.
Yusa
S
,
Catina
TL
,
Campbell
KS
. 
SHP-1- and phosphotyrosine-independent inhibitory signaling by a killer cell Ig-like receptor cytoplasmic domain in human NK cells
.
J Immunol
2002
;
168
:
5047
57
.
36.
Kinsella
TM
,
Nolan
GP
. 
Episomal vectors rapidly and stably produce high-titer recombinant retrovirus
.
Hum Gene Ther
1996
;
7
:
1405
13
.
37.
Anikeeva
N
,
Steblyanko
M
,
Fayngerts
S
,
Kopylova
N
,
Marshall
DJ
,
Powers
GD
, et al
Integrin receptors on tumor cells facilitate NK cell-mediated antibody-dependent cytotoxicity
.
Eur J Immunol
2014
;
44
:
2331
9
.
38.
Phelan
JD
,
Young
RM
,
Webster
DE
,
Roulland
S
,
Wright
GW
,
Kasbekar
M
, et al
A multiprotein supercomplex controlling oncogenic signalling in lymphoma
.
Nature
2018
;
560
:
387
91
.
39.
Wang
T
,
Wei
JJ
,
Sabatini
DM
,
Lander
ES
. 
Genetic screens in human cells using the CRISPR-Cas9 system
.
Science
2014
;
343
:
80
4
.
40.
Woo
J
,
Vierboom
MP
,
Kwon
H
,
Chao
D
,
Ye
S
,
Li
J
, et al
PDL241, a novel humanized monoclonal antibody, reveals CD319 as a therapeutic target for rheumatoid arthritis
.
Arthritis Res Ther
2013
;
15
:
R207
.
41.
Bouchon
A
,
Cella
M
,
Grierson
HL
,
Cohen
JI
,
Colonna
M
. 
Activation of NK cell-mediated cytotoxicity by a SAP-independent receptor of the CD2 family
.
J Immunol
2001
;
167
:
5517
21
.
42.
Kohrt
HE
,
Houot
R
,
Goldstein
MJ
,
Weiskopf
K
,
Alizadeh
AA
,
Brody
J
, et al
CD137 stimulation enhances the antilymphoma activity of anti-CD20 antibodies
.
Blood
2011
;
117
:
2423
32
.
43.
Borrego
F
,
Robertson
MJ
,
Ritz
J
,
Pena
J
,
Solana
R
. 
CD69 is a stimulatory receptor for natural killer cell and its cytotoxic effect is blocked by CD94 inhibitory receptor
.
Immunology
1999
;
97
:
159
65
.
44.
Kim
JR
,
Mathew
SO
,
Patel
RK
,
Pertusi
RM
,
Mathew
PA
. 
Altered expression of signalling lymphocyte activation molecule (SLAM) family receptors CS1 (CD319) and 2B4 (CD244) in patients with systemic lupus erythematosus
.
Clin Exp Immunol
2010
;
160
:
348
58
.
45.
Gandhi
AK
,
Kang
J
,
Havens
CG
,
Conklin
T
,
Ning
Y
,
Wu
L
, et al
Immunomodulatory agents lenalidomide and pomalidomide co-stimulate T cells by inducing degradation of T cell repressors Ikaros and Aiolos via modulation of the E3 ubiquitin ligase complex CRL4(CRBN.)
.
Br J Haematol
2014
;
164
:
811
21
.
46.
Lonial
S
,
Vij
R
,
Harousseau
JL
,
Facon
T
,
Moreau
P
,
Mazumder
A
, et al
Elotuzumab in combination with lenalidomide and low-dose dexamethasone in relapsed or refractory multiple myeloma
.
J Clin Oncol
2012
;
30
:
1953
9
.
47.
Weng
WK
,
Levy
R
. 
Two immunoglobulin G fragment C receptor polymorphisms independently predict response to rituximab in patients with follicular lymphoma
.
J Clin Oncol
2003
;
21
:
3940
7
.
48.
Calpe
S
,
Wang
N
,
Romero
X
,
Berger
SB
,
Lanyi
A
,
Engel
P
, et al
The SLAM and SAP gene families control innate and adaptive immune responses
.
Adv Immunol
2008
;
97
:
177
250
.
49.
Burshtyn
DN
,
Davidson
C
. 
Natural killer cell conjugate assay using two-color flow cytometry
.
Methods Mol Biol
2010
;
612
:
89
96
.
50.
Perez-Quintero
LA
,
Roncagalli
R
,
Guo
H
,
Latour
S
,
Davidson
D
,
Veillette
A
. 
EAT-2, a SAP-like adaptor, controls NK cell activation through phospholipase Cgamma, Ca++, and Erk, leading to granule polarization
.
J Exp Med
2014
;
211
:
727
42
.
51.
Kronke
J
,
Udeshi
ND
,
Narla
A
,
Grauman
P
,
Hurst
SN
,
McConkey
M
, et al
Lenalidomide causes selective degradation of IKZF1 and IKZF3 in multiple myeloma cells
.
Science
2014
;
343
:
301
5
.

Supplementary data