SLAMF6, a member of the SLAM (signaling lymphocyte activation molecules) family, is a homotypic-binding immune receptor expressed on NK, T, and B lymphocytes. Phosphorylation variance between T-cell subclones prompted us to explore its role in anti melanoma immunity. Using a 203-amino acid sequence of the human SLAMF6 (seSLAMF6) ectodomain, we found that seSLAMF6 reduced activation-induced cell death and had an antiapoptotic effect on tumor-infiltrating lymphocytes. CD8+ T cells costimulated with seSLAMF6 secreted more IFNγ and displayed augmented cytolytic activity. The systemic administration of seSLAMF6 to mice sustained adoptively transferred transgenic CD8+ T cells in comparable numbers to high doses of IL2. In a therapeutic model, lymphocytes activated by seSLAMF6 delayed tumor growth, and when further supported in vivo with seSLAMF6, induced complete tumor clearance. The ectodomain expedites the loss of phosphorylation on SLAMF6 that occurs in response to T-cell receptor triggering. Our findings suggest that seSLAMF6 is a costimulator that could be used in melanoma immunotherapy. Cancer Immunol Res; 6(2); 127–38. ©2018 AACR.

One of the approaches used in the search for new immunomodulatory mechanisms is to identify lymphocyte surface receptors that are likely to have a regulatory function. The clinical success of targeting the immunoglobulin superfamily receptors PD-1 and CTLA-4 fueled interest in the uncharacterized members of several structural Ig-like subfamilies (1).

One such family, the SLAM (signaling lymphocytic activation molecules) family of receptors, is typical of the homotypic-binding molecules involved in immunomodulation that are expressed on cells of hematopoietic origin.

Our previous results (2) showed differential expression of SLAMF6 on T-cell clones with identical epitope specificity but different functional capacity. These data sparked our interest in this receptor in the context of antimelanoma CD8+ T-cell responses and prompted us to explore whether SLAMF6 is a function-determining receptor or a secondary chaperone.

SLAMF6 is expressed on natural killer (NK), T, and B cells. It augments Th1 responses in CD4+ T cells and activates NK cells in a homotypic manner (3, 4). SLAMF6 is linked to immune pathology: the allele responsible for autoimmunity in lupus-prone B6.Sle1bz/z mice is a spliced isoform of Ly108, the mouse ortholog of SLAMF6 (5).

In this study, we focused on SLAMF6 as a potential target for improving the CD8+ T-cell response against melanoma. We show that a 203 amino acid–long polypeptide mimicking the extracellular domain of SLAMF6 exerted a costimulatory effect on activated antimelanoma CD8+ T cells. The soluble ectodomain (seSLAMF6) improved CD8+ T-cell expansion, cytokine secretion and cytotoxicity against melanoma. In vivo, the reagent supported adoptively transferred antimelanoma CD8+ T cells, mediated growth delay or complete tumor clearance, and was readily tolerated. We suggest that by interfering with the homotypic binding of the SLAMF6 receptor, seSLAMF6 has the features of a costimulator and could be relevant in melanoma immunotherapy.

Mice

Nude (athymic Foxn1−/−), C57BL/6, and BALB/c mice were purchased from Harlan Laboratories. Pmel-1 mice carry a rearranged T-cell receptor (TCR) Vbeta13, specific for the 9-mer epitope 25-32 from murine Pmel 17, the homolog of human gp100. Pmel-1 and Pmel-1-GFP were self-bred (kind gift from M. Baniyash, Hebrew University, Jerusalem). All experiments were performed with female 8- to 12-week-old mice.

Cells

Melanoma cells.

Cell line 888mel (HLA-A2−/MART-1+/gp100+; ref. 6) and 624mel (HLA-A2+/MART-1+gp100+) were a gift from M. Parkhurst (Surgery Branch, NCI, NIH, Bethesda, MD, 2004). Human melanoma 526mel is an HLA-A2+ cell line. Mouse melanoma B16-F10 and B16-F10/mhgp100 (B16-F10 melanoma cells transduced with pMSGV1 retrovirus that encodes a chimeric mouse gp100 with the human gp10025-33 sequence (gift from Ken-ichi Hanada, Surgery Branch, NCI, NIH, 2014). The cells were cultured in RPMI1640 supplemented with 10% heat-inactivated FCS, 2 mmol/L l-glutamine, and combined antibiotics (all from Invitrogen Life Technologies). Lines are kept in culture for no more than 14 to 20 days, are monthly tested for TCR-specific cognate T-cell reactivity (7), and are mycoplasma free.

TCR-transduced human lymphocytes.

DD154 is a lymphocyte line (HLA-A*0201) transduced to express a TCR cognate for gp100154-162 nonapeptide (gift from the Surgery Branch, NCI, NIH).

Aberrant slamf6 expression on melanoma cells.

Plasmid pDONR223-SLAMF6 was obtained from Addgene (deposited by William Hahn, plasmid # 23396). The full-length SLAMF6 sequence was inserted into PCDNA3.1-hygromycin plasmid (Invitrogen) using BamH1 and Xba1 restriction enzymes (NBT). 624mel human melanoma was transfected with the PCDNA3.1-hygromycin-SLAMF6 vector using lipofectamine. Hygromycin-resistant melanoma cells were subcloned, and the stably transfected cells were labeled with anti-SLAMF6 Ab and analyzed by flow cytometry.

Peripheral blood mononuclear cells.

Peripheral blood mononuclear cells (PBMC) were purified from healthy donors' buffy coats [Hadassah Blood Bank, obtained under Institutional Review Board (IRB) approval].

Tumor-infiltrating lymphocytes.

Microcultures were initiated and expanded from tumor specimens taken from resected metastases of melanoma patients, as described previously (8). Human lymphocytes were cultured in complete medium (CM) consisting of RPMI1640 supplemented with 10% heat-inactivated human AB serum, 2 mmol/l l-glutamine, 1 mmol/l sodium pyruvate, 1% nonessential amino acids, 25 mmol/L HEPES (pH 7.4), 50 μmol/L 2-ME, and combined antibiotics (all from Invitrogen Life Technologies). CM was supplemented with 6,000 IU/mL recombinant human IL2 (rhIL2, Chiron) every other day.

Cloning of peptide-specific TILs.

On day 14 after tumor-infiltrating lymphocyte (TIL) initiation, lymphocytes were washed with PBS, resuspended in PBS supplemented with 0.5% BSA, and stained with FITC-conjugated HLA-A*0201/MART-126–35 or HLA-A*0201/gp100209-217 dextramer (Immudex) for 30 minutes at 4°C. Lymphocytes were then incubated with allophycocyanin-conjugated mouse anti-human CD8 (eBioscience) for an additional 30 minutes at 4°C and washed. CD8+ lymphocytes, positively stained by dextramer (CD8+dextramer+ cells), were sorted by a BD FACSAria and directly cloned at one or two cells per well in 96-well plates in the presence of anti-CD3 (30 ng/mL, eBioscience), rhIL-2 (6,000 IU/mL), and 4 Gy-irradiated 5 × 104 allogeneic PBMCs as feeder cells. Five days later, rhIL2 (6,000 IU/mL) was added and renewed every 2 days thereafter. On day 14, the clones were assayed for IFNγ secretion in a peptide-specific manner following their coincubation with T2 cells pulsed with MART-126–35 or gp100209-217 [both commercially synthesized and purified (>95%) by reverse-phase HPLC by Biomer Technology] by ELISA (R&D Systems). The MART-126–35– or gp100209-217–reactive clones were further expanded in a second-round exposure to anti-CD3 (30 ng/mL) and rhIL2 (6,000 IU/mL) in the presence of 50-fold excess irradiated feeder cells. Supplementary Table S1 describes TILs used throughout the study.

Expansion of lymphocytes.

Human TILs were expanded by activation with 30 ng/mL of anti-CD3 and PBMCs as feeders for 12 days (9, 10). Where indicated, cells were expanded for 3 to 5 days with plate-bound anti-CD3 (1 μg/mL) in the presence of either IL2 (3,000 IU/ml for TILs and 300 IU/mL for PBMCs) or seSLAMF6 (50 μg/mL), as detailed for each experiment. Fresh medium containing IL2 or seSLAMF6 was added on day 5 and every other day thereafter.

Pmel-1 mouse splenocytes (2 × 106/mL) were activated with gp10025-33 peptide (1μg/mL) for 6 days. Cells were incubated with either IL2 (30 IU/mL) or seSLAMF6 (50 μg/mL). Fresh medium containing IL2 or seSLAMF6 was added every other day.

Cell viability assay.

Following expansion, human or mouse lymphocytes were washed, counted, and 1 × 105 cells were cultured in CM supplemented with IL2 or seSLAMF6, as specified in each experiment. At the indicated time points, cells were harvested, washed, and labeled with the Annexin V apoptosis Detection Kit (eBioscience), according to the manufacturer's instructions. Cells were analyzed by flow cytometry using a BD LSRII (BD Biosciences). The data were analyzed using FCS Express software (De Novo Software).

Proteins and antibodies

The following proteins and antibodies were used: seSLAMF6 (recombinant human protein, extracellular domain fused with polyhistidine tag at the C-terminus, Novoprotein); anti-SLAMF6 (NT-7, Biolegend); anti-CD3 (OKT3, eBioscience), anti-CD8, anti-CD107, anti-CD137, anti-OX40, anti-PD-1, anti-2B4, and anti-GITR (all from eBioscience).

In vitro assays

Binding competition assay.

TILs were incubated with increasing concentrations from 10 to 100 μg/mL seSLAMF6 or BSA (negative control) for 45 minutes on ice, washed twice, and labeled with anti-SLAMF6 for 45 minutes on ice. Antibody binding was measured by flow cytometry.

Quantitative RT-PCR.

PBMCs (2 × 106) were incubated in 48-well plates for 3 hours at 37°C in the presence of anti-CD3 with either IL2 (300 IU/mL) or seSLAMF6 (25 μg/mL). Cells were collected and RNA was isolated using GenElute Mammalian Total RNA Kit (Sigma, RTN70) according to the manufacturer's protocol. RNA was then transcribed to cDNA using qScript cDNA Synthesis Kit (Quanta, 95047-100). Real-time PCR was performed using PerfeCT SYBR Green FastMIX ROX (Quanta, 95073-012). Primers used were:

  • Actin: F 5′- CATTGGCAATGAGCGGTTGG R 5′-AGTGATCTCCTTCTGCATCC

  • BCL2: F 5′- CATGCTGGGGCCGTACAG R 5′-GAACCGGCACCTGCACAC

CD107a mobilization assay.

Expanded TILs were washed and cocultured with target melanoma cells for 1 hour at 37°C in the presence of anti-CD107a (eBioscience). Cells were washed and analyzed by flow cytometry.

BCL-2 intracellular staining.

Expanded PBMCs were harvested and washed twice with PBS. Following fixation and permeabilization (eBioscience protocol), intracellular BCL-2 was labeled with anti–BCL-2 (BioLegend) for 30 minutes at 4°C. Appropriate isotype controls were used. Cells were washed twice with permeabilization buffer, resuspended in FACS buffer, and subjected to flow cytometry.

Caspase-3 cleavage assay.

Intracellular staining of cleaved caspase-3 was performed as described previously (11). Briefly, melanoma cells were stained with Cell Trace Far Red DDAO-SE (Life Technologies), according to the manufacturer's protocol. CellTrace Far Red DDAO-SE–labeled melanoma cells were cocultured with effector lymphocytes for 90 minutes at a 1:1 (or 2:1 for DD154 cells) effector:target ratio. The cells were then washed, fixed, permeabilized (eBioscience protocol), and labeled with rabbit anti-cleaved caspase-3-PE (BD Pharmingen). Appropriate isotype controls were included. Washed cells were subjected to flow cytometry.

Phospho-protein measurement.

TILs were activated with plate-bound anti-CD3 with and without 50 μg/mL of seSLAMF6 for 5 and 15 minutes. Cells were harvested, lysed in the presence of phosphatase and protease inhibitors (Pierce IP lysis buffer with HALT phosphatse and protease inhibitor cocktail), and lysates were analyzed using Human Phospho-Immunoreceptor Proteome Profiler (R&D Systems) according to the manufacturer's instructions. Image was captured using GelDoc (Bio-Rad). Spot intensity was quantified (Image Lab software by Bio-Rad).

IFNγ secretion.

Human lymphocytes or mouse splenocytes (1 × 105) were cocultured overnight at a 1:1 ratio with the indicated target melanoma cells. Where indicated, seSLAMF6 50 μg/mL was added to the cultures. Conditioned medium was collected and IFNγ secretion was detected using ELISA (R&D Systems for human and BioLegend for mouse).

Confocal microscopy.

Half a million cells were loaded on slides using cytospin, fixed with 4% PFA in PBS, and blocked with PBS supplemented with 20% goat serum. Cells were stained with rabbit anti-SLAMF6 antibody (Proteintech) and Cy3 goat anti-rabbit (The Jackson Laboratory) was used as secondary antibody. Nuclear staining was performed with 1,5-bis{[2-(di methylamino) ethyl]amino}4,8-dihydroxyanthracene-9,10-dione (DRAQ5, Cell Signaling Technology). Images were captured with a Zeiss LSM 5 confocal microscope and analyzed with Zen software (Carl Zeiss).

Overlapping peptide spot array.

The Intavis spot synthesis peptide array (Koch Institute, MIT, Cambridge, MA) permits the synthesis of a 20 × 30 array (10 × 15 cm) of up to 600 distinct peptides immobilized on a cellulose solid support. Instrument software allows generation of spot sequences and residue offsets from an intact protein sequence. The synthesized peptides are 15 amino acids long, covering the whole protein sequence, with a 1 amino acid shift. Membrane blocking was performed with 5% BSA in TBST (tris buffered saline containing 50 mmol/L Tris, 150 mmol/L NaCl, and 0.05% Tween 20) overnight, followed by overnight incubation with seSLAMF6 (1 μg/mL) in TBST, overnight incubation with 1 μg/mL mouse anti-his antibody (BioLegend) in TBST and 1 hour incubation with goat anti-mouse HRP (The Jackson Laboratory) in TBST. Development was performed with Clarity Western ECL Substrate (Bio-Rad), and results were visualized using Image Lab software.

In vivo experiments

Winn assay.

Human TILs were expanded for 12 days as described above. At the end of activation, cells were washed and mixed at a 1:1 ratio (1 × 106 cells each) with 526mel and immediately injected subcutaneously into the back of 8- to 9-week-old female nude (athymic Foxn1−/−) mice. Tumor size was measured in two perpendicular diameters three times per week. Mice were sacrificed when tumors reached 15 mm diameter in one dimension or when the lesion necrotized. Tumor volume was calculated as L (length) × W (width)2 × 0.5.

Adoptive cell transfer experiments.

  • (i) B16-F10 and B16-F10/mhgp100 mouse melanoma cells (0.5 × 106) were injected subcutaneously into the left and right side of the back of C57BL/6 mice, respectively. Pmel-1 mouse splenocytes (2 × 106/mL) were expanded with 1 μg/mL of gp10025-33 peptide in the presence of IL2 30 IU/mL. Fresh medium containing IL2 was added every other day. On day 5, cells were labeled with CFSE (12). The following day, 107 cells were adoptively transferred into 500 CGy-irradiated tumor-bearing mice. A total of 0.25 × 106 IU/100 μL IL2 or 100 μg seSLAMF6 were administered intraperitoneally once a day for 2 days. The following day, mice were sacrificed and tumors and spleens were harvested for further analysis by flow cytometry.

  • (ii) Melanoma cells were injected as in (i). After 5 to 7 days, the mice were irradiated to 500 CGy. The following day, 5–10 × 106 gp100-specific Pmel-1-GFP lymphocytes (expanded for 6 days with IL2) were adoptively transferred intravenously (0.1 mL total volume) to the tail vein. A total of 0.25 × 106 IU/100 μL IL2 or 100 μg seSLAMF6 were administered intraperitoneally twice a day for 5 days. Mice were sacrificed on day 6, and tumors and draining lymph nodes were harvested for further analysis by flow cytometry.

  • (iii) B16-F10/mhgp100 mouse melanoma cells (0.5 × 106) were injected subcutaneously into the back of C57BL/6 mice. After 5 to 7 days, mice were irradiated to 500 CGy. The following day, 10 × 106 gp100-specific Pmel-1 lymphocytes (expanded for 6 days with IL2 or seSLAMF6) were adoptively transferred intravenously (0.1 mL total volume) to the tail vein. IL2 (0.25 × 106 IU/100 μL) or seSLAMF6 (100 μL) were administered intraperitoneally twice a day for 5 days. Tumor size was measured as above. Mouse weight was measured 3 times a week.

Generation of human lymphocyte cell lines was approved by the IRB (Hadassah Medical Organization IRB, approval number 395–16.09.05). Patients and donors gave their informed consent for the production of tumor cultures, release of TIL from resected metastases, and the use of PBMCs for immune evaluation.

All animal studies were approved by the IRB (approvals MD-14-14155-5 and MD-14-13991-5).

Statistical analysis

Statistical significance was determined by unpaired t test (two-tailed with equal SD) using Prism software (GraphPad). A P value <0.05 was considered statistically significant. Analysis of more than two groups was performed using one-way ANOVA test. Analysis of tumor size was performed by comparing tumor volumes measured on the last day on which all study animals were alive (*, P ≤ 0.05; **, P ≤ 0.01; ***, P ≤ 0.001). For each experiment, the number of replicates performed, the number of animals per group and the statistical test used are stated in the corresponding figure legend.

SLAMF6 ectodomain protects lymphocytes from activation-induced cell death

Because SLAMF6 is a homotypic binding receptor, expressed on resting and activated PBMCs (Supplementary Fig. S1A–S1C), a soluble polypeptide consisting of the ectodomain sequence (seSLAMF6) was used for ligation. To confirm that seSLAMF6 associates with the extracellular domain of the naturally expressed human (Supplementary Fig. S1D) and mouse (Ly108; Supplementary Fig. S1E) receptors, we used a binding competition assay. To identify the specific binding sites of the soluble ectodomain on full-length SLAMF6, an overlapping peptide spot array was utilized. seSLAMF6 bound to sequences extending from proline at position 41 to leucine at position 127 (Supplementary Fig. S2). This binding pattern is in agreement with the homophilic interface between two engaging IgV domains, as predicted by crystallography, encompassing 12 residues between Glu-37 and Ser-90 that form the linkage (13).

Initially, we evaluated the effect of seSLAMF6 on activation-induced cell death (AICD), which is the result of vigorous activation of T cells (14). In clinical protocols of adoptive transfer, large amounts of IL2 are used and administered at high doses to overcome AICD. The associated toxicity, however, is substantial and T-cell persistence posttransfer is still unsatisfactory (15, 16). SLAMF receptors were linked to AICD by the observation that in the absence of SLAM adaptor protein (SAP), the SLAM family activating adaptor, apoptosis is impaired and massive proliferation of CD8+ T cells ensues (17, 18).

To test the effect of seSLAMF6 on apoptosis and on the function of rapidly proliferating T cells, we used an expansion protocol that involves TCR activation and yields a large increase in cell number (9). Bulk (Fig. 1A, left) and dextramer-selected ("cloned", Fig. 1A, right) CD8+ TILs were used. At the end of the activation/expansion phases, lymphocytes were maintained for five additional days with either IL2 or seSLAMF6 added to the culture medium (postexpansion). We hypothesized that in the absence of IL2, T cells would die by neglect.

Figure 1.

SLAMF6 ectodomain protects lymphocytes from activation-induced cell death. A, Human bulk (left) or cloned (right) TIL412 activated for 12 days in IL2 or seSLAMF6 followed by 5 days maintenance in IL2, seSLAMF6 or complete medium alone (CM). Percentage of apoptotic and dead cells was measured by PI-Annexin V. B, Summary of three experiments showing percentage of viable (Annexin V/PI) human TILs 412 and 615 after 5 or 12 days of expansion in IL2 (left) or seSLAMF6 (right), followed by a 5-day maintenance phase (mean ± SEM). C, Human TILs (431, 209 bulk, 209 #4, 412 bulk, 412 sort, 209 #1) expanded for 12 days. Fold increase of cell number is shown. TIL 209 #4 was expanded twice, with two batches. D, Percentage viable mouse splenocytes (Annexin V/PI) expanded in vitro for 6 days in IL2 and maintained for 7 additional days supplemented with IL2, seSLAMF6, or CM alone (mean ± SEM). Left, Pmel-1 splenocytes (n = 3 mice); right, BALB/c splenocytes (n = 2 mice). E, Real-time PCR of Bcl-2 transcript in PBMCs stimulated with anti-CD3 supplemented with IL2, seSLAMF6, or CM (mean ± SEM). One representative experiment out of three performed is shown. F, Intracellular levels of BCL-2 protein were measured by flow cytometry in PBMCs activated with anti-CD3 and IL2, seSLAMF6, or CM. G, C57BL/6 splenocytes expanded with anti-CD3 and IL2 and maintained for 7 days in IL2, seSLAMF6, or CM (mean ± SEM). Summary of three experiments. Mann–Whitney t test. *, P < 0.05; **, P < 0.01.

Figure 1.

SLAMF6 ectodomain protects lymphocytes from activation-induced cell death. A, Human bulk (left) or cloned (right) TIL412 activated for 12 days in IL2 or seSLAMF6 followed by 5 days maintenance in IL2, seSLAMF6 or complete medium alone (CM). Percentage of apoptotic and dead cells was measured by PI-Annexin V. B, Summary of three experiments showing percentage of viable (Annexin V/PI) human TILs 412 and 615 after 5 or 12 days of expansion in IL2 (left) or seSLAMF6 (right), followed by a 5-day maintenance phase (mean ± SEM). C, Human TILs (431, 209 bulk, 209 #4, 412 bulk, 412 sort, 209 #1) expanded for 12 days. Fold increase of cell number is shown. TIL 209 #4 was expanded twice, with two batches. D, Percentage viable mouse splenocytes (Annexin V/PI) expanded in vitro for 6 days in IL2 and maintained for 7 additional days supplemented with IL2, seSLAMF6, or CM alone (mean ± SEM). Left, Pmel-1 splenocytes (n = 3 mice); right, BALB/c splenocytes (n = 2 mice). E, Real-time PCR of Bcl-2 transcript in PBMCs stimulated with anti-CD3 supplemented with IL2, seSLAMF6, or CM (mean ± SEM). One representative experiment out of three performed is shown. F, Intracellular levels of BCL-2 protein were measured by flow cytometry in PBMCs activated with anti-CD3 and IL2, seSLAMF6, or CM. G, C57BL/6 splenocytes expanded with anti-CD3 and IL2 and maintained for 7 days in IL2, seSLAMF6, or CM (mean ± SEM). Summary of three experiments. Mann–Whitney t test. *, P < 0.05; **, P < 0.01.

Close modal

Indeed, in the absence of IL2, the percentage of viable lymphocytes decreased to 31% and 37%. However, those maintained postactivation in seSLAMF6 had improved viability rates of 85% and 92% for the CD8+ clone and bulk TIL412. These values were higher than the viability rates of cells maintained in IL2 in these groups (63% and 65%, respectively). Lymphocytes that were maintained solely in seSLAMF6 both at the expansion and postexpansion phases, and had no IL2 added at any stage of their growth, still showed a high viability rate of 88% and 93% (Fig. 1A and B). The increase in cell number was similar in IL2 and seSLAMF6 expansion (Fig. 1C).

A similar phenomenon was observed using the soluble ectodomain of human SLAMF6 in mouse lymphocytes. Splenocytes from BALB/c and C57BL/6 TCR transgenic Pmel-1 mice displayed improved viability when supplied with seSLAMF6 after anti-CD3 activation or gp10025-32 peptide stimulation (Fig. 1D). In fact, the viability advantage of seSLAMF6 in mouse lymphocytes was greater than in human cells.

To determine whether the improved survival was related to a direct antiapoptotic effect, Bcl-2 transcript and protein were evaluated by quantitative RT-PCR and flow cytometry, respectively. The addition of seSLAMF6 restored Bcl-2 transcript in activated PBMCs to a significantly higher level than that induced by IL2 (P = 0.002; Fig. 1E). The trend was similar for the protein level, although the difference was smaller (Fig. 1F). In addition to improved viability, the antiapoptotic effect of seSLAMF6 resulted in a numerical expansion of lymphocytes at the end of the 7-day postexpansion phase. Splenocyte numbers increased about 4-fold when grown with seSLAMF6 compared with growth with IL2 (Fig. 1G). Anti-SLAMF6 had no effect on T-cell viability (Supplementary Fig. S3).

Taken together, these data demonstrate that seSLAMF6 reduces AICD and preserves the viability of rapidly proliferating T cells, exceeding the antiapoptotic effect of high IL2 levels, and, in mouse splenocytes, augmenting the T-cell proliferation rate.

seSLAMF6 improves effector function of antimelanoma CD8+ T cells

Because seSLAMF6 supports T-cell viability better than IL2, we examined whether the soluble ectodomain complies with other features of an agonistic immune modulator.

To this end, human PBMCs were expanded in CM supplemented with seSLAMF6 or IL2. IFNγ secretion was measured following overnight activation with anti-CD3. The results (Fig. 2A) show increased secretion of IFNγ by PBMCs expanded in seSLAMF6 compared with those expanded in IL2 (P = 0.008). Enhanced IFNγ secretion was also observed in mice using Pmel-1 splenocytes expanded for 6 days with murine gp10025-33 peptide, and rechallenged with B16-F10/mhgp100 mouse melanoma when supplemented with seSLAMF6 compared with IL2 (Fig. 2B).

Figure 2.

Improved antimelanoma function of seSLAMF6-treated lymphocytes. A, PBMCs from five healthy donors were expanded for 3 days with seSLAMF6, IL2, or CM and activated overnight with anti-CD3. IFNγ secretion was measured by ELISA. The figure summarizes two repeats from each donor (mean ± SEM). B, IFNγ secretion by Pmel-1 splenocytes expanded for 6 days with gp10025-33 peptide and seSLAMF6, IL2 or CM (mean ± SEM). C, Human CD8+ TILs (209 clone #4) were expanded for 12 days in IL2 or seSLAMF6 and then incubated for 1 hour with cognate (526mel) or HLA-non cognate melanoma control (888mel) or CM. CD107 surface mobilization was measured by flow cytometry (mean ± SEM). Data summarize four experiments. D, Human CD8+ TILs (209 clone #4) were expanded as in C and then incubated with cognate (624mel and 526mel) or noncognate control (888mel) DDAO-SE-melanoma for 90 minutes. Cytotoxicity was determined by cleaved caspase-3 expression in target melanoma cells. One experiment of two performed is shown. E, DD154 cells were expanded for 5 or 12 days with anti-CD3 in IL2 or seSLAMF6 and then incubated with cognate (624mel and 526mel) or control (888mel) DDAO-SE-melanoma for 90 minutes. Cytotoxicity was determined by cleaved caspase-3 expression in target melanoma cells. F, Summary of four repeats from two experiments, described in E. G, Human T-cell clone 209 was expanded in vitro for 12 days with anti-CD3 in the presence of IL2 or seSLAMF6. Expanded cells were mixed 1:1 with 526mel and injected subcutaneously into athymic Foxn1−/− (nude) mice (Winn assay). Tumor volume was measured on the last day on which all mice were alive (mean ± SEM). Summary of two experiments is shown (n = 10–11 mice/group; 5 mice in the control group). No Tx: 526mel injected without lymphocytes. Unpaired Student t test. *, P < 0.05; **, P < 0.01, ***, P < 0.001.

Figure 2.

Improved antimelanoma function of seSLAMF6-treated lymphocytes. A, PBMCs from five healthy donors were expanded for 3 days with seSLAMF6, IL2, or CM and activated overnight with anti-CD3. IFNγ secretion was measured by ELISA. The figure summarizes two repeats from each donor (mean ± SEM). B, IFNγ secretion by Pmel-1 splenocytes expanded for 6 days with gp10025-33 peptide and seSLAMF6, IL2 or CM (mean ± SEM). C, Human CD8+ TILs (209 clone #4) were expanded for 12 days in IL2 or seSLAMF6 and then incubated for 1 hour with cognate (526mel) or HLA-non cognate melanoma control (888mel) or CM. CD107 surface mobilization was measured by flow cytometry (mean ± SEM). Data summarize four experiments. D, Human CD8+ TILs (209 clone #4) were expanded as in C and then incubated with cognate (624mel and 526mel) or noncognate control (888mel) DDAO-SE-melanoma for 90 minutes. Cytotoxicity was determined by cleaved caspase-3 expression in target melanoma cells. One experiment of two performed is shown. E, DD154 cells were expanded for 5 or 12 days with anti-CD3 in IL2 or seSLAMF6 and then incubated with cognate (624mel and 526mel) or control (888mel) DDAO-SE-melanoma for 90 minutes. Cytotoxicity was determined by cleaved caspase-3 expression in target melanoma cells. F, Summary of four repeats from two experiments, described in E. G, Human T-cell clone 209 was expanded in vitro for 12 days with anti-CD3 in the presence of IL2 or seSLAMF6. Expanded cells were mixed 1:1 with 526mel and injected subcutaneously into athymic Foxn1−/− (nude) mice (Winn assay). Tumor volume was measured on the last day on which all mice were alive (mean ± SEM). Summary of two experiments is shown (n = 10–11 mice/group; 5 mice in the control group). No Tx: 526mel injected without lymphocytes. Unpaired Student t test. *, P < 0.05; **, P < 0.01, ***, P < 0.001.

Close modal

To evaluate cytotoxicity, CD107a mobilization (Fig. 2C) and caspase-3 cleavage (Fig. 2D–F) were used as indicators of lymphocyte lysosomal degranulation and tumor cell damage, respectively. Cognate (526mel and 624mel, A2+) or noncognate (888mel, A2) melanoma lines were incubated with a CD8+ clone derived from TIL209 (clone #4) or TCR-transduced DD154 lymphocytes, expanded in seSLAMF6 or IL2. Figures 2C–F show that T cells expanded in seSLAMF6 induced augmented CD107a mobilization in T cells and caspase-3 cleavage in melanoma targets compared with T cells expanded in IL2. Of note, seSLAMF6 did not induce a cytotoxic response against the noncognate 888mel target, demonstrating that specific recognition is required for the costimulatory effect to take place. The addition of seSLAMF6 to IL2 had no synergistic effect on cytotoxic activity (Supplementary Fig. S4).

Using a Winn assay, we assessed the effect of human antimelanoma CD8+ T cells expanded in seSLAMF6 and mixed with cognate tumor on the growth of human melanoma in athymic Foxn1−/− (nude) mice. The A2-restricted TIL 209 was mixed with 1 × 106 526mel at a ratio of 1:1 and injected subcutaneously into the back of nude mice. TILs expanded in seSLAMF6 were as effective as TILs expanded in high IL2 concentration for preventing melanoma growth. In a control group of TILs that were not supplemented ex vivo by seSLAMF6 or IL2, 3 of 5 mice developed tumors (Fig. 2G).

Overall, these data indicate that activation and expansion of antimelanoma CD8+ T cells with seSLAMF6 was more effective than the standard IL2 protocol at augmenting cytokine secretion and cytotoxic capacity.

seSLAMF6 affects expression of immune receptors and differentiation markers

In the next series of experiments, we aimed to characterize the effect of seSLAMF6 on the expression of other immune checkpoints and differentiation markers in activated CD8+ T cells.

Overnight coculturing of TIL209 with 526mel showed that cell subsets coexpressing GITR, PD-1, 2B4 (SLAMF4), and 4-1BB (CD137) increased in lymphocytes expanded in seSLAMF6 (Fig. 3A). Prolonged and exclusive exposure to seSLAMF6 increased the expression of 2B4 in all CD8+ T cells (Fig. 3A and B). The improved functional capacity and reduced AICD (Fig. 1A) of seSLAMF6-expanded cells implies that, in this context, overexpression of 2B4 is not an indicator of exhaustion (Fig. 3C). In addition, a larger population of 4-1BB–expressing (Fig. 3D) and PD-1–expressing (Fig. 3E) CD8+ T cells was found in seSLAMF6-expanded T cells, whereas OX40 (Fig. 3F) showed lower expression in seSLAMF6-expanded compared with IL2-expanded cells. Figure 3G shows a distinct population of CD4+5RO+, CD62Lhigh/CCR7high double positive lymphocytes that diminished in the presence of IL2 but were retained with seSLAMF6. This phenotype characterizes T cells with a central memory differentiation, which are more likely to persist (19).

Figure 3.

seSLAMF6 affects the expression of other immunomodulatory receptors and differentiation markers. A, TIL209 was expanded for 12 days in CM alone, or supplemented with IL2 or seSLAMF6, and cocultured for 24 hours with cognate (526mel) melanoma. The expression of immune receptors GITR, PD-1, and 2B4 on CD8+ T cells was evaluated by flow cytometry. B, Summary of three TILs (412 bulk, 209 bulk, 412 sorted), cocultured with cognate melanoma 526mel and 624mel, showing 2B4 expression (mean ± SEM). Paired Student t test. C, TIL412 was activated for 12 days in IL2 followed by 5 days of maintenance in IL2, seSLAMF6, or CM. 2B4 expression on CD8+ T cells was measured by flow cytometry. D–F, TIL412 was expanded as in A and cocultured with melanomas 526mel, 624mel, and 888mel. The expression of 4-1BB, PD-1, and OX40 was measured. G, Human PBMCs expanded for 7 days with anti-CD3 and IL2, seSLAMF6, or CM. CD62L and CCR7 expression on CD4+5RO+ cells was determined by flow cytometry. Unpaired Student t test. *, P < 0.05; ***, P < 0.0001.

Figure 3.

seSLAMF6 affects the expression of other immunomodulatory receptors and differentiation markers. A, TIL209 was expanded for 12 days in CM alone, or supplemented with IL2 or seSLAMF6, and cocultured for 24 hours with cognate (526mel) melanoma. The expression of immune receptors GITR, PD-1, and 2B4 on CD8+ T cells was evaluated by flow cytometry. B, Summary of three TILs (412 bulk, 209 bulk, 412 sorted), cocultured with cognate melanoma 526mel and 624mel, showing 2B4 expression (mean ± SEM). Paired Student t test. C, TIL412 was activated for 12 days in IL2 followed by 5 days of maintenance in IL2, seSLAMF6, or CM. 2B4 expression on CD8+ T cells was measured by flow cytometry. D–F, TIL412 was expanded as in A and cocultured with melanomas 526mel, 624mel, and 888mel. The expression of 4-1BB, PD-1, and OX40 was measured. G, Human PBMCs expanded for 7 days with anti-CD3 and IL2, seSLAMF6, or CM. CD62L and CCR7 expression on CD4+5RO+ cells was determined by flow cytometry. Unpaired Student t test. *, P < 0.05; ***, P < 0.0001.

Close modal

In summary, the enhanced response of CD8+ T cells that were preexposed to seSLAMF6 was associated with a change in immune receptor expression. The altered pattern may play a role in the improved response or may merely represent the outcome of vigorous activation.

Systemic administration of seSLAMF6 sustains adoptively transferred T cells in vivo

We evaluated whether the improved in vitro viability of activated CD8+ T cells that were supplemented with seSLAMF6 would also manifest itself in vivo. To this end, persistence and proliferation of transferred CFSE-labeled Pmel-1 T cells was measured. Labeled splenocytes were administered to melanoma-bearing irradiated mice. Posttransfer, the animals were injected with two doses of intraperitoneal IL2, seSLAMF6, or PBS, 24 hours apart. Mice were sacrificed on day 3, and the spleens were harvested.

Mice treated with IL2 or seSLAMF6 had a higher percent of CFSE-positive cells in their spleens than PBS-treated controls (P < 0.01). The percent was similar in IL2-treated mice and in seSLAMF6-treated mice (Fig. 4A and B). CFSE dilution showed that a majority of Pmel-1 T cells from mice treated with IL2 underwent 2 to 4 divisions, while T cells from seSLAMF6-treated mice were still in their first or second division cycle (Fig. 4C).

Figure 4.

Systemic seSLAMF6 administration enhances persistence of transferred lymphocytes in vivo. A–C, Pmel-1 splenocytes were expanded with gp10025-33 cognate peptide (1 μg/mL) in IL2 (30 IU/mL) for 6 days. On day 5, cells were labeled with CFSE and injected one day later intravenously into irradiated B16-F10- and B16-F10/mhgp100–bearing C57BL/6 mice. IL2 (0.25 × 106 IU), seSLAMF6 (100 μg), or PBS were administered intraperitoneally once a day for 2 days. On the third day, mice were sacrificed and spleens were harvested. A, Flow cytometry of splenocytes from treated mice (one representative mouse from each group is shown). B, Summary of all mice (n = 4/group; mean ± SEM). C, Progressive dilution of CFSE dye in dividing splenocytes (“0” denotes undivided cells, “1” denotes cells after one division, etc.) was analyzed by flow cytometry. The graph shows percentage (mean ± SEM) of splenocytes in successive divisions. D and E, Pmel-1-GFP splenocytes were expanded with gp10025-33 peptide (1 μg/mL) in IL2 (30 IU/mL) for 6 days and injected intravenously into irradiated B16-F10- and B16-F10/mhgp100–bearing C57BL/6 mice. IL2 (0.25 × 106 IU) or seSLAMF6 (100μg) were administered intraperitoneally twice daily for 5 days. On the sixth day, mice were sacrificed, and draining lymph nodes were harvested and analyzed by flow cytometry (mean ± SEM). D and E, One representative mouse from each group (D) and comparison of pooled data from both tumor-draining lymph nodes from all mice receiving IL2 (n = 10) or seSLAMF6 (E; n = 8). Unpaired Student t test. *, P < 0.05; **, P < 0.01; ***, P < 0.001.

Figure 4.

Systemic seSLAMF6 administration enhances persistence of transferred lymphocytes in vivo. A–C, Pmel-1 splenocytes were expanded with gp10025-33 cognate peptide (1 μg/mL) in IL2 (30 IU/mL) for 6 days. On day 5, cells were labeled with CFSE and injected one day later intravenously into irradiated B16-F10- and B16-F10/mhgp100–bearing C57BL/6 mice. IL2 (0.25 × 106 IU), seSLAMF6 (100 μg), or PBS were administered intraperitoneally once a day for 2 days. On the third day, mice were sacrificed and spleens were harvested. A, Flow cytometry of splenocytes from treated mice (one representative mouse from each group is shown). B, Summary of all mice (n = 4/group; mean ± SEM). C, Progressive dilution of CFSE dye in dividing splenocytes (“0” denotes undivided cells, “1” denotes cells after one division, etc.) was analyzed by flow cytometry. The graph shows percentage (mean ± SEM) of splenocytes in successive divisions. D and E, Pmel-1-GFP splenocytes were expanded with gp10025-33 peptide (1 μg/mL) in IL2 (30 IU/mL) for 6 days and injected intravenously into irradiated B16-F10- and B16-F10/mhgp100–bearing C57BL/6 mice. IL2 (0.25 × 106 IU) or seSLAMF6 (100μg) were administered intraperitoneally twice daily for 5 days. On the sixth day, mice were sacrificed, and draining lymph nodes were harvested and analyzed by flow cytometry (mean ± SEM). D and E, One representative mouse from each group (D) and comparison of pooled data from both tumor-draining lymph nodes from all mice receiving IL2 (n = 10) or seSLAMF6 (E; n = 8). Unpaired Student t test. *, P < 0.05; **, P < 0.01; ***, P < 0.001.

Close modal

The presence of transferred T cells in the tumor and its draining lymph nodes correlates with treatment outcome (20). To detect the effect of seSLAMF6 on lymphocyte localization in draining lymph nodes, Pmel-1-GFP T cells were transferred to C57BL/6 mice bearing established B16-F10 and B16-F10/mhgp100 melanomas on each side of the back. The double tumor model was selected to identify the role of melanoma immunogenicity on T-cell accumulation in the draining lymph nodes. On day 6 after tumor implantation, mice were irradiated to 500 CGy and injected with Pmel-1-GFP T cells. Following transfer, the animals received two daily doses of IL2 or seSLAMF6, for 5 successive days. One day later, mice were sacrificed and tumors and draining lymph nodes were harvested. The tumors were largely necrotic and did not yield lymphocytes, but a distinct population of Pmel-1-GFP lymphocytes was detected in the lymph nodes draining both melanomas (Fig. 4D). A higher ratio of Pmel-1 cells was found in mice treated with seSLAMF6, in lymph nodes of both B16-F10 and the immunogenic B16-F10/mhgp100 tumors (Fig. 4E).

These experiments demonstrate that systemically administered seSLAMF6 was able to sustain adoptively transferred lymphocytes in recipient mice, with enhanced accumulation in tumor draining lymph nodes.

Melanoma growth delay in mice receiving T cells expanded ex vivo with seSLAMF6

Our next goal was to evaluate the effect of seSLAMF6 on the antitumor activity of adoptively transferred, melanoma-cognate cytotoxic T lymphocytes (CTL) against established melanoma tumors. To assess this, peptide-activated splenocytes from Pmel-1 transgenic mice were administered to irradiated C57BL/6 mice carrying 6-day-old B16-F10/mhgp100 melanoma (Fig. 5A). During the pretransfer (expansion) phase, we compared the standard IL2 protocol with a seSLAMF6-supplemented regimen for the ability to expand and preserve viability and function of proliferating CD8+ T cells (Fig. 1C and F). Following transfer, we compared the effect of systemic administration of high IL2 doses and seSLAMF6 on the antimelanoma response of seSLAMF6-expanded Pmel-1 T cells. The standard IL2 expansion/IL2 injection protocol was used as a reference.

Figure 5.

Melanoma growth is delayed or prevented in mice receiving antigen-specific CD8+ T cells expanded in vitro and supported in vivo by seSLAMF6. Adoptive cell therapy in C57BL/6 tumor-bearing mice. A, Experimental design. B, Spider plot showing tumor volume calculated as L (length) × W (width)2 × 0.5. TF, tumor free. No Tx: No lymphocytes injected. C, Kaplan–Meier survival curve. No Tx: No lymphocytes injected. D, Tumor size measured on day 31, the last day on which all mice were alive (in the negative control group, all mice died by day 25). E, Average weight of mice receiving systemic IL2 versus seSLAMF6. One experiment, including 7 mice per group, and 5 mice in the systemic SLAMF6 group, was performed. Unpaired Student t test. *, P < 0.05; ***, P < 0.001.

Figure 5.

Melanoma growth is delayed or prevented in mice receiving antigen-specific CD8+ T cells expanded in vitro and supported in vivo by seSLAMF6. Adoptive cell therapy in C57BL/6 tumor-bearing mice. A, Experimental design. B, Spider plot showing tumor volume calculated as L (length) × W (width)2 × 0.5. TF, tumor free. No Tx: No lymphocytes injected. C, Kaplan–Meier survival curve. No Tx: No lymphocytes injected. D, Tumor size measured on day 31, the last day on which all mice were alive (in the negative control group, all mice died by day 25). E, Average weight of mice receiving systemic IL2 versus seSLAMF6. One experiment, including 7 mice per group, and 5 mice in the systemic SLAMF6 group, was performed. Unpaired Student t test. *, P < 0.05; ***, P < 0.001.

Close modal

All groups of mice receiving Pmel-1 T cells showed a delay in tumor outburst (Fig. 5B and C). However, although tumors eventually grew in all mice receiving lymphocytes expanded in seSLAMF6 and supported in vivo by IL2, the systemic administration of seSLAMF6 to support seSLAMF6-expanded Pmel-1 cells resulted in complete clearance of melanoma in 2 of 5 mice; melanoma did not recur during 80 days of follow-up. The optimized reference protocol of IL2-expanded/IL2-supported adoptive transfer resulted in the longest growth delay and best clearance rate of melanoma (4/7). On day 31, the last day on which all mice were alive (except the group with no injected lymphocytes), tumor size was similar in mice treated exclusively with seSLAMF6 and those treated with the IL2 reference protocol (Fig. 5D). During the 5 days in which the mice received high-dose IL2 injections, they lost 16% of their body weight on average. In contrast, mice receiving seSLAMF6 lost less than 5% of their body weight (Fig. 5E), attesting to the reduced systemic toxicity of seSLAMF6.

In summary, although IL2 is the accepted protocol for preparation and maintenance of adoptively transferred CD8+ T cells, seSLAMF6 is a potential alternative that may be better tolerated.

Aberrant expression of SLAMF6 inhibits antimelanoma T-cell reactivity

To elucidate the mechanism of action of seSLAMF6, we evaluated the net effect of transactivation of the SLAMF6 receptor in the context of effector–target interaction. To this end, we engineered 624mel melanoma cells, which lack SLAMF6, to aberrantly express the full-length receptor (624mel-slamf6 cells; Fig. 6A). Melanoma cognate TILs (209, 412, 431) were cocultured overnight with aberrantly expressing 624mel-slamf6. IFNγ secretion decreased after stimulation with the 624mel- slamf6 compared with the natural target. The addition of seSLAMF6 to the coculture resulted in increased IFNγ secretion, but did not restore the original reactivity (Fig. 6B). These results imply that SLAMF6 engagement in trans reduces lymphocyte response. In this context, the soluble ectodomain lowers SLAMF6's natural inhibitory role.

Figure 6.

Aberrant expression of SLAMF6 inhibits antimelanoma T-cell reactivity. A, 624mel was stably transfected with the pCDNA3.1-hygromycin-SLAMF6 vector (624mel-slamf6). SLAMF6-expressing cells were labeled with anti-SLAMF6 mAb and analyzed by flow cytometry. B, hHuman TILs 209, 412, and 431 were cocultured overnight with parental and 624mel-slamf6 target melanoma cells. Where indicated, seSLAMF6 (50 μg/mL) was added to the culture. Supernatant was collected, and IFNγ secretion was measured by ELISA (mean ± SEM). Unpaired Student t test. ***, P < 0.001.

Figure 6.

Aberrant expression of SLAMF6 inhibits antimelanoma T-cell reactivity. A, 624mel was stably transfected with the pCDNA3.1-hygromycin-SLAMF6 vector (624mel-slamf6). SLAMF6-expressing cells were labeled with anti-SLAMF6 mAb and analyzed by flow cytometry. B, hHuman TILs 209, 412, and 431 were cocultured overnight with parental and 624mel-slamf6 target melanoma cells. Where indicated, seSLAMF6 (50 μg/mL) was added to the culture. Supernatant was collected, and IFNγ secretion was measured by ELISA (mean ± SEM). Unpaired Student t test. ***, P < 0.001.

Close modal

seSLAMF6 induces receptor dephosphorylation in activated TILs

To identify the initial signal induced by seSLAMF6, we used the Proteome Profiler array to follow SLAMF6 phosphorylation upon seSLAMF6 engagement. Phosphorylation was measured in anti-CD3–activated melanoma cognate TILs 209 clone #1 and clone #4 and TIL 412 at 0, 5, and 15 minutes after stimulation. The addition of seSLAMF6 was compared with basal CD3 activation.

As shown in Fig. 7, SLAMF6 is phosphorylated in nonactivated TILs. Following CD3 activation, SLAMF6 is dephosphorylated. However, although dephosphorylation induced by CD3 activation is apparent after 15 minutes, the addition of seSLAMF6 induced dephosphorylation after only 5 minutes. Thus, T-cell activation, and, to a greater extent, seSLAMF6 costimulation, induced dephosphorylation of the SLAMF6 receptor.

Figure 7.

seSLAMF6 induces receptor dephosphorylation in activated TILs. TIL 209 clones #1 and #4 and TIL412 were activated with plate-bound anti-CD3 with or without the addition of seSLAMF6 for 5 and 15 minutes. Cells were harvested, lysed in the presence of phosphatase and protease inhibitors, and lysates were analyzed using Human Phospho-Immunoreceptor Proteome Profiler. Image was captured (bottom) and dot intensity was calculated using Image Lab software (mean ± SEM; top). P-SLAMF6, phosphorylated SLAMF6. Unpaired Student t test. **, P < 0.01.

Figure 7.

seSLAMF6 induces receptor dephosphorylation in activated TILs. TIL 209 clones #1 and #4 and TIL412 were activated with plate-bound anti-CD3 with or without the addition of seSLAMF6 for 5 and 15 minutes. Cells were harvested, lysed in the presence of phosphatase and protease inhibitors, and lysates were analyzed using Human Phospho-Immunoreceptor Proteome Profiler. Image was captured (bottom) and dot intensity was calculated using Image Lab software (mean ± SEM; top). P-SLAMF6, phosphorylated SLAMF6. Unpaired Student t test. **, P < 0.01.

Close modal

We conclude that SLAMF6 acts as a CD8+ T-cell inhibitory receptor, which is dephosphorylated upon TCR stimulation. The soluble ectodomain of SLAMF6 enhances receptor dephosphorylation acting as an agonistic immune modulator.

In this work, we showed that the soluble ectodomain of SLAMF6, which binds to sequences of the extracellular part of the receptor by ligand mimicry, in vitro and in vivo, presents features of a costimulatory immune modulator. The magnitude of the activating binding effect was substantial and unexpected, in light of the uncertain role attributed to this receptor per se, outside the context of the SLAM family. In fact, it is difficult to assess the net contribution of SLAMF6 to normal immune response because much of the data on this receptor were acquired in disease-prone animal models.

The human disease that drew attention to SLAMF receptors, X-linked lymphoproliferative disorder, indicated a dual function for SLAMF6, as a negative or a positive coreceptor, depending on competition between SAP and phosphatase SHP-1 for binding to the immunoreceptor tyrosine-based switch motif (ITSM) on its cytoplasmic tail (21). Schwartzberg and colleagues showed that although SLAMF6−/− mice have intact antiviral CD4-mediated responses, the deletion of SLAMF6 repairs the defect that occurs in SAP deficiency (22). The observation that SLAMF6 exerts a negative effect on synapse organization in the absence of SAP was also shown in CD8+ T cells. Enhanced diffuse SHP-1 localization was detected in the defective immune synapse, further supporting the concept that in the absence of SAP, SLAMF6 recruits SHP-1, which consecutively dephosphorylates kinases such as Lck, which is required for TCR signaling (23).

Further support for the inhibitory role of SLAMF6 was provided by adoptive transfer of SLAMF6−/− CD4+ T cells, which caused abnormalities related to systemic lupus erythematosus in a susceptible mouse strain (24). The complexity of deciphering the role of SLAMF6 was further exemplified by Veillette and his group, who showed that SLAMF6 is required for NK-cell priming in a phase prior to target encounter, and that the effect of the receptor is exerted in cis, that is, in the same cell or cell populations of the same hematopoietic ancestry (25).

The main enhancing effects of seSLAMF6 in our systems, which focused on CD8+ T cells, were improved viability and enhanced cytotoxicity against melanoma targets. We chose to compare SLAMF6 with IL2 because this cytokine, itself a product of strongly costimulated T cells (26, 27), circumvents the need for costimulation and drives CD8+ T-cell function to a theoretical maximum. IL2 is also a cytokine approved for use in the clinic, for the preparatory phase and posttransfer support of T cell–based therapies. In the early phases of development, strategies of ACT were mainly based on TILs in melanoma (28). Since then, the field of ACT has expanded to many other cancers, with the introduction of genetically engineered TCRs, leveraging the recognition of unique tumor antigens or surface markers by antibodies (29–31). In the future, it is not unlikely that “off-the-shelf” TCRs and hybrids may become part of cancer care, especially because regressions of epithelial cancers have occurred and mutated oncogenes have been successfully targeted (32). However, the problem remains that even with inserts of optimized costimulatory signals, the resistance of engineered cells to apoptosis still poses a challenge, as rebound overexpression of inhibitory receptors results in decreased function (33), and IL2 is still required (31). Our data suggest that seSLAMF6 can be given systemically and could replace IL2, simultaneously providing costimulation and opposing suppressive signals. The use of seSLAMF6 highlights the difference between a costimulator and a cytokine, as with costimulation only antigen-specific CTLs are subjected to function enhancement, whereas nonactivated T cells remain unaffected.

The mechanism of action of seSLAMF6 is addressed indirectly in our data. The reduced antimelanoma activity of CTLs against SLAMF6-expressing melanoma implies that ligation of this receptor in trans exerts an inhibitory effect. It is possible that the same effect occurs in cis, between lymphocytes. We show that SLAMF6 in nonactivated T cells is in the phosphorylated form. Rapid dephosphorylation, which occurs upon activation, is accelerated by seSLAMF6. We suggest that SLAMF6 acts as a rheostat: close contact between lymphocytes provides binding opportunities for the receptor with extracellular domains of other cells, leading to SLAMF6 phosphorylation. In this state, lymphocytes are less susceptible to activating signals. seSLAMF6 interferes with this autoregulatory status, increasing the magnitude of T-cell functional capacity and making the lymphocytes less susceptible to apoptosis.

Although inhibitory receptors can be targeted by mAbs, our literature review showed little study of SLAMF6-targeting antibodies that augment immune response. Only Valdez and colleagues, who used a mixture of mAbs against human SLAMF6 in CD4+ T cells, showed improved IFNγ secretion in naïve T cells, but not in memory cells (3). From the peptide binding map, we conclude that seSLAMF6, which is a monomer at relevant concentrations, adheres to the native receptor in a spatial conformation that may not be reproducible by an antibody. In this way, seSLAMF6 suppresses an autoregulatory effect, unleashing an armed phenotype in addition to external TCR triggering.

In summary, the soluble ectodomain of SLAMF6 activates CD8+ T cells in a manner that complies with the characteristics of costimulation: when signal 1 is generated, lymphocytes exposed to seSLAMF6 display improved capacity to secrete IFNγ and lyse tumor targets. Increased expression of BCL-2, induced by seSLAMF6, combats activation-induced cell death, and as a result sustains lymphocyte proliferation and numerical expansion. When given to melanoma-bearing mice, antigen-specific CTLs supported in vivo by seSLAMF6 inhibit melanoma growth and circumvent the need for cytokine support. This highlights seSLAMF6 as an attractive component in strategies of adoptive T-cell transfer.

A. Machlenkin is the head of Immuno Oncology at Compugen Ltd. M. Lotem has received speakers bureau honoraria from BMS and is a consultant/advisory board member for MSD. No potential conflicts of interest were disclosed by the other authors.

Conception and design: G. Eisenberg, R. Uzana, A. Machlenkin, M. Lotem

Development of methodology: G. Eisenberg, R. Uzana, A. Rutenberg

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): G. Eisenberg, R. Engelstein, A. Geiger-Maor, E. Hajaj, A. Rutenberg

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): G. Eisenberg, R. Engelstein, A. Geiger-Maor, E. Hajaj, S. Frankenburg, G. Frei, T. Peretz, M. Lotem

Writing, review, and/or revision of the manuscript: G. Eisenberg, R. Engelstein, A. Geiger-Maor, S. Merims, S. Frankenburg, M. Lotem

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): G. Eisenberg, S. Frankenburg

Study supervision: G. Eisenberg, M. Lotem

This work was supported by research grants from Dr. Miriam and Sheldon G. Adelson Medical Research Foundation (AMRF); the Canadian Institutes of Health Research (CIHR), the International Development Research Centre (IDRC), the Israel Science Foundation (ISF), the Azrieli Foundation; Melanoma Research Alliance; Deutsche Forschungsgemeinschaft (DFG); and The Israel Cancer Research Fund, Rosetrees Trust, and the Perlstein Family Fund.

The authors wish to acknowledge the devoted technical work of Inna Ben David, Anna Kuznetz and Yael Gelfand. We thank Andre Veillette, Ofer Mandelboim, Eli Pikarsky, Philipp Beckhove, and Tillmann Michels for helpful discussions. Special gratitude to Stewart Greenberg for his endless support of this work.

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.
Yap
EH
,
Rosche
T
,
Almo
S
,
Fiser
A
. 
Functional clustering of immunoglobulin superfamily proteins with protein-protein interaction information calibrated hidden Markov model sequence profiles
.
J Mol Biol
2014
;
426
:
945
61
.
2.
Uzana
R
,
Eisenberg
G
,
Sagi
Y
,
Frankenburg
S
,
Merims
S
,
Amariglio
N
, et al
Trogocytosis is a gateway to characterize functional diversity in melanoma-specific CD8+ T cell clones
.
J Immunol
2012
;
188
:
632
40
.
3.
Valdez
PA
,
Wang
H
,
Seshasayee
D
,
van Lookeren Campagne
M
,
Gurney
A
,
Lee
WP
, et al
NTB-A, a new activating receptor in T cells that regulates autoimmune disease
.
J Biol Chem
2004
;
279
:
18662
9
.
4.
Flaig
RM
,
Stark
S
,
Watzl
C
. 
Cutting edge: NTB-A activates NK cells via homophilic interaction
.
J Immunol
2004
;
172
:
6524
7
.
5.
Kumar
KR
,
Li
L
,
Yan
M
,
Bhaskarabhatla
M
,
Mobley
AB
,
Nguyen
C
, et al
Regulation of B cell tolerance by the lupus susceptibility gene Ly108
.
Science
2006
;
312
:
1665
9
.
6.
Parkhurst
MR
,
DePan
C
,
Riley
JP
,
Rosenberg
SA
,
Shu
S
. 
Hybrids of dendritic cells and tumor cells generated by electrofusion simultaneously present immunodominant epitopes from multiple human tumor-associated antigens in the context of MHC class I and class II molecules
.
J Immunol
2003
;
170
:
5317
25
.
7.
Uzana
R
,
Eisenberg
G
,
Merims
S
,
Frankenburg
S
,
Pato
A
,
Yefenof
E
, et al
Human T cell crosstalk is induced by tumor membrane transfer
.
PLoS One
2015
;
10
:
e0118244
.
8.
Lotem
M
,
Peretz
T
,
Drize
O
,
Gimmon
Z
,
Ad El
D
,
Weitzen
R
, et al
Autologous cell vaccine as a post operative adjuvant treatment for high-risk melanoma patients (AJCC stages III and IV). The new American Joint Committee on Cancer
.
Br J Cancer
2002
;
86
:
1534
9
.
9.
Dudley
ME
,
Wunderlich
JR
,
Shelton
TE
,
Even
J
,
Rosenberg
SA
. 
Generation of tumor-infiltrating lymphocyte cultures for use in adoptive transfer therapy for melanoma patients
.
J Immunother
2003
;
26
:
332
42
.
10.
Eisenberg
G
,
Uzana
R
,
Pato
A
,
Frankenburg
S
,
Merims
S
,
Yefenof
E
, et al
Imprinting of lymphocytes with melanoma antigens acquired by trogocytosis facilitates identification of tumor-reactive T cells
.
J Immunol
2013
;
190
:
5856
65
.
11.
He
L
,
Hakimi
J
,
Salha
D
,
Miron
I
,
Dunn
P
,
Radvanyi
L
. 
A sensitive flow cytometry-based cytotoxic T-lymphocyte assay through detection of cleaved caspase 3 in target cells
.
J Immunol Methods
2005
;
304
:
43
59
.
12.
Quah
BJ
,
Warren
HS
. 
Parish CR, Monitoring lymphocyte proliferation in vitro and in vivo with the intracellular fluorescent dye carboxyfluorescein diacetate succinimidyl ester
.
Nat Protoc
2007
;
2
:
2049
56
.
13.
Cao
E
,
Ramagopal
UA
,
Fedorov
A
,
Fedorov
E
,
Yan
Q
,
Lary
JW
, et al
NTB-A receptor crystal structure: insights into homophilic interactions in the signaling lymphocytic activation molecule receptor family
.
Immunity
2006
;
25
:
559
70
.
14.
Krammer
PH
,
Arnold
R
,
Lavrik
IN
. 
Life and death in peripheral T cells
.
Nat Rev Immunol
2007
;
7
:
532
42
.
15.
Chhabra
A
,
Mukherji
B
,
Batra
D
. 
Activation induced cell death (AICD)of human melanoma antigen-specific TCR engineered CD8 T cells involves JNK, Bim and p53
.
Expert Opin Ther Targets
2017
;
21
:
117
29
.
16.
Robbins
PF
,
Dudley
ME
,
Wunderlich
J
,
El-Gamil
M
,
Li
YF
,
Zhou
J
, et al
Cutting edge: persistence of transferred lymphocyte clonotypes correlates with cancer regression in patients receiving cell transfer therapy
.
J Immunol
2004
;
173
:
7125
30
.
17.
Chen
G
,
Tai
AK
,
Lin
M
,
Chang
F
,
Terhorst
C
,
Huber
BT
. 
Increased proliferation of CD8+ T cells in SAP-deficient mice is associated with impaired activation-induced cell death
.
Eur J Immunol
2007
;
37
:
663
74
.
18.
Snow
AL
,
Marsh
RA
,
Krummey
SM
,
Roehrs
P
,
Young
LR
,
Zhang
K
, et al
Restimulation-induced apoptosis of T cells is impaired in patients with X-linked lymphoproliferative disease caused by SAP deficiency
.
J Clin Invest
2009
;
119
:
2976
89
.
19.
Gattinoni
L
,
Klebanoff
CA
.
Restifo
NP
. 
Paths to stemness: building the ultimate antitumour T cell
.
Nat Rev Cancer
2012
;
12
:
671
84
.
20.
Pockaj
BA
,
Sherry
RM
,
Wei
JP
,
Yannelli
JR
,
Carter
CS
,
Leitman
SF
, et al
Localization of 111indium-labeled tumor infiltrating lymphocytes to tumor in patients receiving adoptive immunotherapy. Augmentation with cyclophosphamide and correlation with response
.
Cancer
1994
;
73
:
1731
7
.
21.
Cannons
JL
,
Tangye
SG
,
Schwartzberg
PL
. 
SLAM family receptors and SAP adaptors in immunity
.
Annu Rev Immunol
2011
;
29
:
665
705
.
22.
Kageyama
R
,
Cannons
JL
,
Zhao
F
,
Yusuf
I
,
Lao
C
,
Locci
M
, et al
The receptor Ly108 functions as a SAP adaptor-dependent on-off switch for T cell help to B cells and NKT cell development
.
Immunity
2012
;
36
:
986
1002
.
23.
Zhao
F
,
Cannons
JL
,
Dutta
M
,
Griffiths
GM
,
Schwartzberg
PL
. 
Positive and negative signaling through SLAM receptors regulate synapse organization and thresholds of cytolysis
.
Immunity
2012
;
36
:
1003
16
.
24.
Wang
N
,
Keszei
M
,
Halibozek
P
,
Yigit
B
,
Engel
P
,
Terhorst
C
. 
Slamf6 negatively regulates autoimmunity
.
Clin Immunol
2016
;
173
:
19
26
.
25.
Wu
N
,
Zhong
MC
,
Roncagalli
R
,
Pérez-Quintero
LA
,
Guo
H
,
Zhang
Z
, et al
A hematopoietic cell-driven mechanism involving SLAMF6 receptor, SAP adaptors and SHP-1 phosphatase regulates NK cell education
.
Nat Immunol
2016
;
17
:
387
96
.
26.
Parry
RV
,
Rumbley
CA
,
Vandenberghe
LH
,
June
CH
,
Riley
JL
. 
CD28 and inducible costimulatory protein Src homology 2 binding domains show distinct regulation of phosphatidylinositol 3-kinase, Bcl-xL, and IL-2 expression in primary human CD4 T lymphocytes
.
J Immunol
2003
;
171
:
166
74
.
27.
Barsoumian
HB
,
Yolcu
ES
,
Shirwan
H
. 
4-1BB signaling in conventional T cells drives IL-2 production that overcomes CD4+CD25+FoxP3+ T regulatory cell suppression
.
PLoS One
2016
;
11
:
e0153088
.
28.
Rosenberg
SA
,
Packard
BS
,
Aebersold
PM
,
Solomon
D
,
Topalian
SL
,
Toy
ST
, et al
Use of tumor-infiltrating lymphocytes and interleukin-2 in the immunotherapy of patients with metastatic melanoma. A preliminary report
.
N Engl J Med
1988
;
319
:
1676
80
.
29.
Morgan
RA
,
Dudley
ME
,
Wunderlich
JR
,
Hughes
MS
,
Yang
JC
,
Sherry
RM
, et al
Cancer regression in patients after transfer of genetically engineered lymphocytes
.
Science
2006
;
314
:
126
9
.
30.
Gross
G
,
Waks
T
,
Eshhar
Z
. 
Expression of immunoglobulin-T-cell receptor chimeric molecules as functional receptors with antibody-type specificity
.
Proc Natl Acad Sci U S A
1989
;
86
:
10024
8
.
31.
Brown
CE
,
Alizadeh
D
,
Starr
R
,
Weng
L
,
Wagner
JR
,
Naranjo
A
, et al
Regression of glioblastoma after chimeric antigen receptor T-Cell Therapy
.
N Engl J Med
2016
;
375
:
2561
9
.
32.
Tran
E
,
Robbins
PF
,
Lu
YC
,
Prickett
TD
,
Gartner
JJ
,
Jia
L
, et al
T-Cell transfer therapy targeting mutant KRAS in cancer
.
N Engl J Med
2016
;
375
:
2255
62
.
33.
Moon
EK
,
Wang
LC
,
Dolfi
DV
,
Wilson
CB
,
Ranganathan
R
,
Sun
J
, et al
Multifactorial T-cell hypofunction that is reversible can limit the efficacy of chimeric antigen receptor-transduced human T cells in solid tumors
.
Clin Cancer Res
2014
;
20
:
4262
73
.