Chimeric Ag receptor T cells derived from universal donors are susceptible to recipient immunologic rejection, which may limit their in vivo persistence and compromise treatment efficacy. In this study, we generated HLA class I–deficient T cells by disrupting β2-microglobulin to evade recognition by HLA-mismatched CD8+ T cells, and then restored NK cell tolerance by forced expression of an HLA-E single-chain receptor. We specifically report on an optimized hypoimmunogenic disulfide trap HLA-E4 (dtHLA-E4) molecule that exhibited increased surface expression, enhanced NK cell inhibitory potential, and abrogated CD8-dependent T cell recognition. Our dtHLA-E4 molecule comprised the CD4 (4) transmembrane domain and truncated cytoplasmic region, as well as disulfide trap mutations to anchor an HLA class I signal sequence-derived peptide. Functional comparison of dtHLA-E4 molecules fused to different VL9 epitopes showed that peptides derived from HLA-A and HLA-C allotypes maximized NK cell inhibition and minimized NKG2C+ NK cell activation. Furthermore, incorporation of mutations into the α3 domain of HLA-E diminished the immunogenicity of dtHLA-E4 by reducing CD8+ T cell recognition, but crucially, these mutations left NK cell inhibitory function intact. These findings demonstrate the systematic construction of a hypoimmunogenic dtHLA-E4 molecule, which promises to facilitate persistence of allogeneic HLA class I–deficient chimeric Ag receptor T cells by overcoming NK cell missing-self recognition.

Autologous chimeric Ag receptor (CAR) T cells have transformed the treatment paradigm for patients with hematological malignancies. Yet, the production of these CAR-T cells is lengthy, expensive, and there is an inherent risk of manufacturing failure due to poor T cell fitness that may result from advanced disease and prior lines of therapy (1–3). Thus, CAR-T cells derived from healthy allogeneic donors is an attractive solution that could reduce patient wait time to treatment and cost by standardizing a scalable manufacturing process (4). However, HLA mismatch between an allogeneic donor and recipient patient challenges the clinical feasibility of allogeneic CAR-T cells. For example, disruption of the endogenous TCR in allogeneic T cells is necessary to prevent life-threatening graft-versus-host disease (5–7), but retention of non-self HLA molecules renders these cells vulnerable to immunologic rejection by the recipient (8), which may serve to limit their in vivo persistence. Consequently, additional genome engineering is required to facilitate clinical investigation of allogeneic CAR-T cells.

The elimination of surface HLA class I and HLA class II expression by inactivating β2-microglobulin (B2M) and HLA class II transcriptional activator (CIITA), respectively, renders allogeneic CAR-T cells invisible to HLA-mismatched T cells (8, 9). However, the absence of HLA class I triggers missing-self recognition and lysis by NK cells (10). Previous attempts to restore NK cell tolerance to HLA class I–deficient T cells include genetic disruption of NK cell activating ligands (11) and adhesion molecules (12) or forced expression of an inhibitory ligand (11, 13–17). The latter approach necessitates that the inhibitory ligand is nonimmunogenic insofar as not to induce alloreactive T cell or Ab responses, and that the corresponding inhibitory receptor is widely expressed on NK cell subsets.

HLA-E is an attractive NK cell inhibitory ligand because this molecule is minimally polymorphic (18) and binds the heterodimeric inhibitory receptor NKG2A/CD94 (19, 20), which is expressed by ∼50% of circulating NK cells (21, 22). Similar to other HLA class I molecules, HLA-E requires noncovalent interactions with B2M and a cognate peptide ligand to correctly assemble and traffic to the cell surface, so it, as well, would not be expressed in B2M-deficient cells. Hence, previous attempts to restore HLA-E expression have required constructing a single-chain HLA-E (scHLA-E) molecule that fused together the HLA-E H chain, B2M, and loading peptide using intervening flexible linkers (23). After forced expression in xenografts (23, 24) and allogeneic human T cells or pluripotent stem cells (13–15, 25), scHLA-E has been shown to mitigate NK cell–dependent lysis.

In this study, we systematically optimized the scHLA-E molecule to improve cell surface expression and NK cell inhibitory capacity while minimizing immunogenic potential. To do so, we structurally modified the scHLA-E chassis by replacing the HLA-E transmembrane and cytoplasmic regions with that of the CD4 (4) coreceptor. We then functionally compared scHLA-E4 molecules fused to eight independent VL9 epitopes derived from the signal sequences of common HLA class Ia alleles and HLA-G. scHLA-E molecules that comprised VL9 epitopes from HLA-A and HLA-C allotypes, as well as disulfide trap mutations to anchor the loading peptide, maximized NK cell inhibition and minimally stimulated NKG2C+ NK cells. Lastly, our disulfide trap HLA-E4 (dtHLA-E4) variants induced HLA-E–specific CD8+ T cell responses, which necessitated introducing mutations into the α3 domain of HLA-E H chain to abrogate CD8-dependent T cell recognition. Taken together, these findings describe a hypoimmunogenic dtHLA-E4 molecule that effectively averts NK cell–driven rejection of HLA class I-deficient T cells.

HLA-E single-chain molecules were constructed in the following N- to C-terminal orientation: B2M signal peptide, VL9 loading peptide, (G4S)3 linker, B2M chain, (G4S)4 linker, and HLA-E*01:03 H chain. scHLA-E8 and scHLA-E4 molecules retained the HLA-E*01:03 extracellular domain but used the transmembrane domain and truncated cytoplasmic regions from CD8α or CD4, respectively. The VL9 epitopes VMAPRTLFL (v1), VMAPRTLLL (v2), VMAPRTLVL (v3), VMAPRTLIL (v4), VMAPRALLL (v5), VTAPRTVLL (v6), VMAPRTVLL (v7), and VTAPRTLLL (v8) were derived from signal sequences of HLA-G*01:01, HLA-A*01:01, HLA-A*02:01/HLA-C*02:01, HLA-C*01:02, HLA-C*07:01, HLA-B*35:01, HLA-B*07:02, and HLA-B*13:01, respectively. A comprehensive list of HLA class I alleles comprising these VL9 epitopes can be found elsewhere (26). scHLA-A*02 molecules were constructed by fusing the B2M signal peptide, B2M chain, (G4S)4 linker, and HLA-A*02:01 H chain. Disulfide trap scHLA-E4 molecules contained mutations within the (G4S)3 linker (GCGGS(G4S)2) and HLA-E*01:03 H chain (Y84C). Hypoimmunogenic scHLA-A*02 and dtHLA-E4v4 molecules contained D227K, T228A, and A245V mutations within the α3 domain of the HLA H chain. All amino acid sequences are reported in Supplemental Table I. Each scHLA-E molecule was separated by a T2A linker to methotrexate-resistant dihydrofolate reductase (27). All gene fragments were codon optimized (IDT) and then custom synthesized and cloned into a self-inactivating lentiviral vector (GenScript).

Packaging plasmids encoding HIV Gag/Pol (pALD-GagPol), HIV Rev (pALD-Rev), and VSV glycoprotein (pALD-VSV-G) were purchased from Aldevron and used to generate lentiviral particles. The self-inactivating transfer plasmid and packaging plasmids were transfected into HEK293T cells using Lipofectamine 2000 (Life Technologies). Lentivirus supernatant was collected at 24 h posttransfection and then clarified through a 0.45-µm filter. Filtered lentivirus supernatant was mixed with PEG-it virus precipitation solution (System Biosciences) according to the manufacturer’s instructions and stored at 4°C overnight, after which the virus solution was concentrated for 30 min at 1500 × g, 4°C. The supernatant was aspirated, and the virus pellet was resuspended in 600 μl of ImmunoCult-XF T cell expansion medium (STEMCELL Technologies) and stored at −80°C. Lenti-X GoStix Plus (Takara) was used to quantify lentivirus titer.

Adult human leukopaks were purchased from HemaCare (Charles River) and then T cells were isolated using the StraightFrom Leukopak CD4/CD8 MicroBead kit (Miltenyi Biotec) following the manufacturer’s instructions and cryopreserved in CS10 (STEMCELL Technologies). T cells were thawed and resuspended at 1 × 106 cells/ml in expansion medium consisting of ImmunoCult XF T cell expansion medium, 2 mM GlutaMAX, 25 mM HEPES buffer, and 1% penicillin-streptomycin, and 5% CTS immune cell SR (serum replacement) (Thermo Fisher Scientific). Expansion medium was supplemented with 10 ng/ml IL-7 and IL-15 (BioLegend). T cells were stimulated with ImmunoCult human CD3/CD28/CD2 T cell activator (STEMCELL Technologies) following the manufacturer’s protocol and incubated at 37°C, 5% CO2, and 95% humidity. Forty-eight hours following stimulation, T cells were washed with PBS and resuspended in P3 buffer (Lonza) at 5 × 107 cells/ml. T cells were then electroporated using the Lonza 4DNucleofector system, program DH-102. The electroporation reaction consisted of 2 µg of mRNA encoding ABE8.20m and 1 µg of B2M-specific single-guide RNA (sgRNA) from IDT (protospacer sequence, 5′-CTTACCCCACTTAACTATCT-3′) per 1 × 106 cells. T cells were recovered in expansion medium at 1 × 106 cells/ml and were transduced with lentiviral vector supernatant (∼100 ng of p24). T cell donors were transduced with independent lots of lentiviruses. Four days later, T cell cultures were divided and left untreated or treated with 220 nM methotrexate (MilliporeSigma). Medium was exchanged every second day to adjust cell concentration to 5 × 105 cells/ml until day 13 when cells were cryopreserved in CS10.

Cells were stained in 50 µl of PBS containing 2% FBS and 2 mM EDTA with anti-human Abs: CD3 (OKT3), CD2 (RPA-2.10), CD4 (OKT4), CD8 (SK1), CD56 (5.1H11), CD107a (H4A3), HLA-ABC (W6/32), HLA-E (3D12), HLA-A2 (BB7.2), B2M (A17082A), CD86 (IT2.2), NKG2A (S19004C), and NKG2C (S19005E). Intracellular cytokines were detected using cell fixation and cell permeability kits (Invitrogen) per the manufacturer’s instructions using anti-human TNF-α (MAb11) (BioLegend). Surface-bound NKG2A and NKG2C recombinant proteins were detected by incubating T cells with biotinylated human NKG2A&CD94 protein, Fc Avitag and biotinylated human NKG2C&CD94 Protein, His Avitag (Acro Biosystems) followed by staining with fluorophore-labeled streptavidin (BioLegend). Flow cytometry data were acquired on the MACSQuant analyzer 16 (Miltenyi Biotec) and analyzed using FlowJo software v10 (Tree Star). All analyses were performed on live cells within the lymphocyte gate (forward scatter area versus side scatter area) following doublet exclusion (side scatter height versus side scatter width) and staining negative for fixable viability dye eFluor 780 (eBioscience).

PBMCs were obtained from human leukopaks by Ficoll separation. To generate effector cells, CD8+ T cells were isolated from PBMCs (donor 1) using a CD8 Microbead kit (Miltenyi Biotec). To generate stimulator cells, CD3+ cells were depleted from PBMCs of an HLA-mismatched donor (donor 2) using a CD3 Microbead kit (Miltenyi Biotec). The CD3-positive fraction from donor 2 was then used to generate B2M knockout (B2MKO) T cells and unmodified T cells. All cell isolations were performed following the manufacturer’s protocols. Effector CD8+ T cells (donor 1) were cultured at a 1:1 ratio with stimulator CD3-depleted PBMCs (donor 2) for 1 wk in expansion medium supplemented with 300 U/ml IL-2 (Sartorius). Afterward, these activated effector CD8+ T cells were labeled with CellTrace Violet (Thermo Fisher Scientific) according to the manufacturer’s protocol, and then restimulated with a 1:1 ratio of B2MKO or unmodified T cells (donor 2) at 20:1, 10:1, 5:1, 2.5:1, 1.25:1 or 0:1 E:T ratios. After 48 h, the assay was analyzed by flow cytometry to detect the change in the frequency of B2MKO and unmodified T cells. Flow cytometry gating strategy follows viable lymphocytes/CD3+/CellTrace Violet/with/without B2M.

NK cells were isolated from human leukopaks (Charles River) using a StraightFrom Leukopak REAlease CD56 MicroBead kit (Miltenyi Biotec) or CD56 MicroBeads (Miltenyi Biotec). NK cells were cultured for 3 d in expansion medium supplemented with 300 U/ml IL-2 and 5 ng/ml IL-15. Afterward, NK cells were cultured at different ratios with unmodified or B2MKO allogeneic T cells. B2MKO T cells engineered to express scHLA-E molecules were methotrexate (MTX) selected prior to assay setup. Unmodified and B2MKO allogeneic T cells were combined at an equal ratio before adding NK cells to measure the relative change in frequency of B2MKO T cells compared with unmodified T cells in the absence of NK cells. NK cell–dependent lysis of B2MKO T cells was measured 48 h later by flow cytometry using the following formula: NK cell–dependent lysis (%) = 100 – [100 × (% survival in the presence of NK cells/% survival in the absence of NK cells)]. Flow cytometry gating strategy follows viable lymphocytes/CD56/CD3+/CD4+CD8+/with/without HLA class I identified by HLA-ABC, B2M, or HLA-A2 staining. NK cell degranulation and TNF-α production were measured by culturing 1 × 105 NK cells with 1 × 105 B2MKO or unmodified T cells, or alone (media only control). Anti-CD107a Ab was added at the start of stimulation followed by the addition of 1× monensin solution and 1× brefeldin A (BioLegend) 1 h later. Cells were incubated for 6 h total before Ab staining and analysis by flow cytometry. Flow cytometry gating strategy follows viable lymphocytes/CD3/CD56+/CD107a+ or TNF-α+, or viable lymphocytes/CD3/CD56+/NKG2A+ or NKG2C+/CD107a+ or TNF-α+.

Artificial APCs (aAPCs) were generated by transducing 5 × 105 K-562 cells (American Type Culture Collection) with lentivirus encoding CD86. The resulting K.86 cell line recovered in R10 medium comprising RPMI 1640 supplemented with 2 mM GlutaMAX, 25 mM HEPES buffer, 1% penicillin-streptomycin, and 10% FBS (Thermo Fisher Scientific). After recovery, K.86 cells were transduced with lentiviral vectors encoding individual dtHLA-E4 or scHLA-A*02 molecules, and then expanded in R10. Cell surface expression of CD86 and B2M were detected on these aAPC lines by flow cytometry. To perform the HLA immunogenicity assays, aAPCs were treated with 10 µg/ml mitomycin C (MMC) (MilliporeSigma) for 3 h at 37°C, and then washed twice to remove residual MMC. T cells were thawed and mixed with MMC-treated aAPCs at a 2:1 ratio in expansion medium supplemented with 300 U/ml IL-2 and incubated at 37°C, 5% CO2, and 95% humidity. At 7 d poststimulation, samples were analyzed for T cell proliferation and were restimulated with the appropriate aAPC and K.86 cell lines at a 1:1 ratio in the presence of an anti-CD107a Ab. One hour later, 1× monensin solution and 1× brefeldin A were added to the culture and then incubated at 37°C for 5 h before Ab staining and analysis by flow cytometry. The percentage of responding T cells was calculated by subtracting background production after stimulation with K.86 cells. Flow cytometry gating strategy follows viable lymphocytes/CD3+/CD8+ or CD4+/CD107a+ or TNF-α+.

Genomic DNA samples from B2MKO T cells were prepared to determine on-target genomic editing efficiency as previously described (28). Briefly, ∼1 × 106 cells were lysed using QuickExtract DNA extraction solution (Lucigen) following the manufacturer’s instructions. Two microliters of genomic DNA was added to a 25-µl PCR containing Q5 high-fidelity DNA polymerase (New England Biolabs) and 0.5 µM forward and 0.5 µM reverse primers. Forward and revers primer sequences are 5′-TGTCTTTCAGCAAGGACTGGTCTTTCTA-3′ and 5′-GACTCATTCAGGGTAGTATGGCCATAGA-3′, respectively. Products were then amplified using Illumina barcoding primer pairs, and then the amplicon was purified using solid phase reversible immobilization beads (Beckman Coulter) and quantified using a NanoDrop spectrophotometer (Thermo Fischer Scientific). Barcoded products were sequenced using an Illumina MiSeq instrument according to the manufacturer’s protocol.

Statistical comparisons of paired samples were performed using a two-sided nonparametric Wilcoxon matched-pairs signed rank test or Friedman test followed by Dunn’s test for multiple comparisons. Comparisons of unmatched samples were performed using a nonparametric Wilcoxon rank sum test. All statistical analyses were performed using GraphPad Prism version 9.3.0 (GraphPad).

Recognition of mismatched HLA class I alleles expressed on allogeneic CAR-T cells by the patient immune system may result in rapid clearance by cytotoxic CD8+ T cells. However, genome editing approaches to disrupt HLA class I expression in allogeneic CAR-T cells could alleviate recipient CD8+ T cell recognition, thereby improving their in vivo lifespan and therapeutic window (Fig. 1A). Thus, we leveraged an adenosine base editor (ABE8.20m) that when complexed with a sgRNA could disrupt B2M expression by inactivating a conserved splice donor site (exon|GT-intron) via a transition mutation (A·G) (Fig. 1B). To do so, primary human T cells were activated and then electroporated with mRNA encoding ABE8.20m and a B2M-specific sgRNA. This pairing achieved a mean on-target genomic editing efficiency of 95.5% (Fig. 1C), corresponding to a profound loss of HLA class I cell surface expression (Fig. 1D). The resulting B2MKO T cells were invisible to alloreactive CD8+ T cells from an unrelated donor, whereas unmodified T cells were eliminated (Fig. 1E, 1F). Although disrupting B2M in allogeneic T cells prevented rejection by HLA-mismatched CD8+ T cells, in turn the absence of HLA class I expression could activate NK cells by missing self-recognition (Fig. 1A). As a result, B2MKO T cells induced degranulation by NK cells (Fig. 1G) and were rapidly eliminated after in vitro culture with NK cells (Fig. 1H, 1I). These data indicate that the status of HLA class I expression on allogeneic T cells dictates their susceptibility to either T cell– or NK cell–mediated responses, and, importantly, additional genome engineering approaches are required to avert NK cell–driven allorejection.

FIGURE 1.

B2M-deficient T cells evade alloreactive T cells but are susceptible to NK cell–mediated lysis. (A) Schematic demonstrating that the status of HLA class I expression dictates susceptibility of allogeneic T cells to rejection by T cells or NK cells from an unrelated donor. (B) Schematic showing B2M-specific single-guide RNA (sgRNA) disrupting the splice donor site at the exon/intron junction (B2MKO). (C) Frequency of on-target A > G nucleotide conversion by next-generation sequencing in primary human T cells base-edited with ABE8.20m mRNA and B2M-specific sgRNA. Symbols indicate four independent T cell donors. (D) Histogram shows cell surface expression of HLA class I in B2MKO and unmodified T cells. (E and F) T cell mixed leukocyte assay as described in Materials and Methods. FACS plots (E) and summarized data (F) are shown for the frequency of B2MKO and unmodified T cells at 48 h after culture in triplicate at the indicated E:T ratios with HLA-mismatched CD8+ T cells that were labeled with CellTrace Violet. (G) Frequency of CD107a+ NK cells after stimulation with B2MKO and unmodified T cells. Symbols represent three independent NK cell donors in duplicate. (H and I) NK cell cytotoxicity assay as described in Materials and Methods. FACS plots indicate frequency (H) and summarized data indicate NK cell–dependent lysis (I) of B2MKO and unmodified T cells at 48 h after culture with NK cells at the indicated E:T ratios. Symbols represent four independent NK cell donors in duplicate. For all data, bars represent mean and error bars indicate ±SEM.

FIGURE 1.

B2M-deficient T cells evade alloreactive T cells but are susceptible to NK cell–mediated lysis. (A) Schematic demonstrating that the status of HLA class I expression dictates susceptibility of allogeneic T cells to rejection by T cells or NK cells from an unrelated donor. (B) Schematic showing B2M-specific single-guide RNA (sgRNA) disrupting the splice donor site at the exon/intron junction (B2MKO). (C) Frequency of on-target A > G nucleotide conversion by next-generation sequencing in primary human T cells base-edited with ABE8.20m mRNA and B2M-specific sgRNA. Symbols indicate four independent T cell donors. (D) Histogram shows cell surface expression of HLA class I in B2MKO and unmodified T cells. (E and F) T cell mixed leukocyte assay as described in Materials and Methods. FACS plots (E) and summarized data (F) are shown for the frequency of B2MKO and unmodified T cells at 48 h after culture in triplicate at the indicated E:T ratios with HLA-mismatched CD8+ T cells that were labeled with CellTrace Violet. (G) Frequency of CD107a+ NK cells after stimulation with B2MKO and unmodified T cells. Symbols represent three independent NK cell donors in duplicate. (H and I) NK cell cytotoxicity assay as described in Materials and Methods. FACS plots indicate frequency (H) and summarized data indicate NK cell–dependent lysis (I) of B2MKO and unmodified T cells at 48 h after culture with NK cells at the indicated E:T ratios. Symbols represent four independent NK cell donors in duplicate. For all data, bars represent mean and error bars indicate ±SEM.

Close modal

To protect B2MKO T cells from allorecognition, we constructed an scHLA-E molecule that could inhibit NK cell–mediated lysis through binding to NKG2A/CD94 (Fig. 2A). Four types of scHLA-E molecules were designed for our experiments (Fig. 2B). The scHLA-E dimer (D) comprised an HLA-E*01:03 H chain covalently fused to the signal peptide and mature chain of B2M via an intervening (G4S)4 linker, enabling the scHLA-ED to bind endogenous peptide ligands for proper assembly and presentation. The scHLA-E trimer (T) contained an additional N-terminal (G4S)3 linker fused to a peptide ligand from the signal sequence of HLA-G*01 (VMAPRTLFL). In addition, we constructed scHLA-ET molecules where the HLA-E transmembrane and cytoplasmic domains were replaced with those of CD8α (8) or CD4 (4), except to prevent undesired signaling, the cytoplasmic regions were truncated before the CXCP motif required for interactions with Lck (29–31). In this way, we hypothesized that surface expression of scHLA-E8 and scHLA-E4 may be greater than either construct containing the HLA-E cytoplasmic tail, which facilitates rapid recycling of endogenous HLA-E molecules (32).

FIGURE 2.

scHLA-E4 exhibits improved cell surface expression and inhibits NK cell–driven lysis. (A) Schematic demonstrating that expression of an HLA-E single-chain (sc) molecule in B2MKO T cells inhibits NK cells by engaging the NKG2A/CD94 heterodimer. (B) Schematic detailing components of scHLA-E fusion constructs, including scHLA-E dimer (D), scHLA-E trimer (T), and scHLA-E molecules containing the CD8α (8) and CD4 (4) transmembrane domain and truncated cytoplasmic regions. (C and D) B2MKO T cells were transduced with an equivalent titer of lentivirus that encoded a unique scHLA-E variant and mDHFR. (C) FACS plots demonstrate the cell surface expression of the scHLA-E variants. (D) Summarized data show the geometric median fluorescence intensity (GMFI) of HLA-E on total nontransduced B2MKO T cells, and HLA-E–positive unmodified T cells and B2MKO T cells that were transduced with scHLA-E4 variants. Nontransduced B2MKO T cells served as a gating control to discriminate positive and negative HLA-E expression. Symbols represent four independent T cell donors in duplicate at 11 d posttransduction. (E and F) Histograms (E) and summarized data (F) indicate GMFI of scHLA-E variants bound to recombinant human NKG2A/CD94 heterodimer. Symbols represent three independent T cell donors. (G and H) Frequency of NKG2A+ NK cells and total CD56+ NK cells that upregulated expression of CD107a (G) and TNF-α (H) after stimulation with B2MKO T cells from three independent donors that were nontransduced or expressed scHLA-E4. Symbols represent eight independent NK cell donors in duplicate. (I) Correlation between frequency of NKG2A+ NK cells and percentage change in frequency of total responding NK cells after stimulation with scHLA-E4B2MKO T cells from stimulation with nontransduced B2MKO T cells. Symbols represent eight independent NK cell donors. (J) NK cell cytotoxicity assay as described in Materials and Methods. NK cell–dependent lysis of B2MKO T cells from three independent donors that were nontransduced or expressed scHLA-E4 at 48 h after culture with NK cells at the 4:1 E:T ratio. Symbols represent five independent NK cell donors in duplicate. For all data, bars represent mean and error bars indicate ±SEM. Statistical significance was calculated by a Wilcoxon matched-pairs signed rank test (D, F, G, H, and J) and Spearman correlation (I).

FIGURE 2.

scHLA-E4 exhibits improved cell surface expression and inhibits NK cell–driven lysis. (A) Schematic demonstrating that expression of an HLA-E single-chain (sc) molecule in B2MKO T cells inhibits NK cells by engaging the NKG2A/CD94 heterodimer. (B) Schematic detailing components of scHLA-E fusion constructs, including scHLA-E dimer (D), scHLA-E trimer (T), and scHLA-E molecules containing the CD8α (8) and CD4 (4) transmembrane domain and truncated cytoplasmic regions. (C and D) B2MKO T cells were transduced with an equivalent titer of lentivirus that encoded a unique scHLA-E variant and mDHFR. (C) FACS plots demonstrate the cell surface expression of the scHLA-E variants. (D) Summarized data show the geometric median fluorescence intensity (GMFI) of HLA-E on total nontransduced B2MKO T cells, and HLA-E–positive unmodified T cells and B2MKO T cells that were transduced with scHLA-E4 variants. Nontransduced B2MKO T cells served as a gating control to discriminate positive and negative HLA-E expression. Symbols represent four independent T cell donors in duplicate at 11 d posttransduction. (E and F) Histograms (E) and summarized data (F) indicate GMFI of scHLA-E variants bound to recombinant human NKG2A/CD94 heterodimer. Symbols represent three independent T cell donors. (G and H) Frequency of NKG2A+ NK cells and total CD56+ NK cells that upregulated expression of CD107a (G) and TNF-α (H) after stimulation with B2MKO T cells from three independent donors that were nontransduced or expressed scHLA-E4. Symbols represent eight independent NK cell donors in duplicate. (I) Correlation between frequency of NKG2A+ NK cells and percentage change in frequency of total responding NK cells after stimulation with scHLA-E4B2MKO T cells from stimulation with nontransduced B2MKO T cells. Symbols represent eight independent NK cell donors. (J) NK cell cytotoxicity assay as described in Materials and Methods. NK cell–dependent lysis of B2MKO T cells from three independent donors that were nontransduced or expressed scHLA-E4 at 48 h after culture with NK cells at the 4:1 E:T ratio. Symbols represent five independent NK cell donors in duplicate. For all data, bars represent mean and error bars indicate ±SEM. Statistical significance was calculated by a Wilcoxon matched-pairs signed rank test (D, F, G, H, and J) and Spearman correlation (I).

Close modal

scHLA-E molecules were cloned into a bicistronic lentiviral vector containing methotrexate-resistant dihydrofolate reductase (mDHFR) (27) separated by an intervening T2A peptide. After lentiviral transduction, each scHLA-E molecule was successfully detected on the cell surface of B2MKO T cells by flow cytometry (Fig. 2C). Forced expression of each scHLA-E molecule substantially improved the expression level of HLA-E on transduced B2MKO T cells over the endogenous level of HLA-E in unmodified T cells (Fig. 2D). Moreover, the inclusion of the VMAPRTLFL epitope stabilized longitudinal expression of the scHLA-ET over the scHLA-ED (Supplemental Fig. 1A), but this effect was nullified when the CD8α transmembrane domain and cytoplasmic region replaced their HLA-E counterparts (Supplemental Fig. 1A). The scHLA-E4 molecule exhibited similar surface expression levels as the scHLA-ET at 4 d posttransduction, but thereafter the scHLA-E4 sustained a higher level of steady-state expression at latter timepoints of manufacturing (Fig. 2D, Supplemental Fig. 1A), including in both CD4+ and CD8+ T cells (Supplemental Fig. 1B). Furthermore, B2MKO T cells expressing the scHLA-E4 construct exhibited a 2-fold increase in fluorescence intensity of bound recombinant NKG2A/CD94 protein relative to scHLA-ET-positive T cells (Fig. 2E, 2F). These data suggest that the scHLA-E4 molecule is most likely to sustain adequate cell surface expression, which may confer B2MKO T cells greater long-term protection against NK cell–mediated lysis.

Given that the scHLA-E4 molecule exhibited high-level cell surface expression, we carried this construct forward into functional assays. To do so, we leveraged expression of the mDHFR cassette to render scHLA-E4-positive B2MKO T cells resistant to lymphotoxic concentrations of MTX, whereas nontransduced B2MKO T cells were eliminated (Supplemental Fig. 1C). In this way, MTX selection facilitated evaluation of the scHLA-E4 in the absence of nontransduced B2MKO T cells within the same culture that in turn could stimulate NK cells. Importantly, MTX treatment of B2MKO T cells did not impact their susceptibility to alloreactive NK cell responses, as the frequency of degranulating and TNF-α+ NK cells (Supplemental Fig. 1D), as well as the magnitude of NK cell–dependent lysis (Supplemental Fig. 1E), was unchanged from stimulation with untreated B2MKO T cells. We then MTX treated B2MKO T cells transduced with scHLA-E4 and cultured these cells with NK cells from unrelated donors. Notably, the scHLA-E4 potently decreased the frequency of NKG2A+ and total NK cells that degranulated (Fig. 2G) and produced TNF-α (Fig. 2H). The ability of scHLA-E4 to decrease the frequency of total responding NK cells relative to nontransduced B2MKO T cells correlated with the prevalence of NKG2A expression (Fig. 2I), which could explain the divergent extent to which scHLA-E4 decreases the susceptibility of B2MKO T cells to NK cells from unique donors. Furthermore, the scHLA-E4 molecule mitigated in vitro NK cell–driven lysis (Fig. 2J) relative to stimulation with nontransduced B2MKO T cells. Taken together, inclusion of the CD4 transmembrane and cytoplasmic components into scHLA-ET improved surface expression, resulting in a molecule that inhibits NK cell reactivity against B2MKO T cells.

HLA-E binds nonamer peptides derived from the signal sequences of HLA-A, HLA-B, HLA-C, and HLA-G (33, 34). However, only a subset of HLA class I signal sequence variants are efficiently processed to generate epitopes that bind HLA-E and engage NKG2A/CD94 on NK cells (26). In this study, we used the scHLA-E4 chassis to evaluate whether eight different VL9 epitopes from common HLA-A, HLA-B, and HLA-C allotypes and HLA-G stabilize scHLA-E4 expression and impact its ability to inhibit NK cells (Fig. 3A). The scHLA-E4 variants exhibited differential cell surface expression levels, where molecules comprising the VMAPRTLFL (v1), VMAPRTLLL (v2), VMAPRTLIL (v4), and VMAPRTVLL (v7) epitopes were highly expressed at near-equivalent levels (Fig. 3B, 3C). In contrast, scHLA-E4 molecules fused to HLA-B allotype-derived VL9 epitopes with threonine at position 2 (2T), VTAPRTVLL (v6) and VTAPRTLLL (v8), expressed poorly on the cell surface (Fig. 3B, 3C), which is congruent with reports indicating that these peptides bind HLA-E with lower affinity (20).

FIGURE 3.

VL9 epitope variants modulate surface expression and NK cell inhibitory potential of scHLA-E4 molecules. (A) Schematic of a scHLA-E4 molecule and VL9 epitope variants derived from signal sequences of HLA-G*01 and representative HLA class Ia alleles. (B and C) B2MKO T cells were transduced with an equivalent titer of lentivirus that encoded a unique scHLA-E4 VL9 variant, and HLA-E cell surface expression was quantified at 11 d posttransduction. (B) FACS plots indicate cell surface expression of each scHLA-E4 VL9 variant and corresponding GMFI of HLA-E on transduced B2MKO T cells. (C) Heatmap shows GMFI of HLA-E on total nontransduced B2MKO T cells and HLA-E–positive unmodified T cells and B2MKO T cells expressing each scHLA-E4 VL9 variant at 11 d posttransduction. Nontransduced B2MKO T cells served as a gating control to discriminate positive and negative HLA-E expression. Data represent the mean of four independent T cell donors in duplicate. (DH) NK cells from unrelated donors were stimulated with unmodified T cells or B2MKO T cells from three independent donors that were nontransduced or expressed unique scHLA-E4 VL9 variants. (D) FACS plots indicate frequency of NKG2A+ NK cells that express CD107a and TNF-α poststimulation with B2MKO T cells that were nontransduced or expressed scHLA-E4v4. (E–H) Summarized data indicate the frequency of CD107a+ (E) and TNF-α+ (F) NKG2A+ NK cells, and CD107a+ (G) and TNF-α+ (H) total CD56+ NK cells. Symbols represent data from eight (E and F) and seven (G and H) independent NK cell donors in duplicate. (I) NK cell cytotoxicity assay as described in Materials and Methods. NK cell–dependent lysis of B2MKO T cells from four independent donors that were nontransduced or expressed unique scHLA-E4 VL9 variants at 48 h after culture with NK cells at the 4:1 E:T ratio. Symbols represent seven independent NK cell donors in duplicate. For all data, bars represent mean and error bars indicate ±SEM. For (C), a Wilcoxon matched-pairs signed rank test was used to calculate significance. For (E)–(I), statistical significance between B2MKO T cells that were nontransduced and each scHLA-E4 VL9 variant was calculated using a Wilcoxon matched-pairs signed rank test, whereas the Friedman test with a Dunn test for multiple comparisons calculated statistical significance between the scHLA-E4 VL9 variants.

FIGURE 3.

VL9 epitope variants modulate surface expression and NK cell inhibitory potential of scHLA-E4 molecules. (A) Schematic of a scHLA-E4 molecule and VL9 epitope variants derived from signal sequences of HLA-G*01 and representative HLA class Ia alleles. (B and C) B2MKO T cells were transduced with an equivalent titer of lentivirus that encoded a unique scHLA-E4 VL9 variant, and HLA-E cell surface expression was quantified at 11 d posttransduction. (B) FACS plots indicate cell surface expression of each scHLA-E4 VL9 variant and corresponding GMFI of HLA-E on transduced B2MKO T cells. (C) Heatmap shows GMFI of HLA-E on total nontransduced B2MKO T cells and HLA-E–positive unmodified T cells and B2MKO T cells expressing each scHLA-E4 VL9 variant at 11 d posttransduction. Nontransduced B2MKO T cells served as a gating control to discriminate positive and negative HLA-E expression. Data represent the mean of four independent T cell donors in duplicate. (DH) NK cells from unrelated donors were stimulated with unmodified T cells or B2MKO T cells from three independent donors that were nontransduced or expressed unique scHLA-E4 VL9 variants. (D) FACS plots indicate frequency of NKG2A+ NK cells that express CD107a and TNF-α poststimulation with B2MKO T cells that were nontransduced or expressed scHLA-E4v4. (E–H) Summarized data indicate the frequency of CD107a+ (E) and TNF-α+ (F) NKG2A+ NK cells, and CD107a+ (G) and TNF-α+ (H) total CD56+ NK cells. Symbols represent data from eight (E and F) and seven (G and H) independent NK cell donors in duplicate. (I) NK cell cytotoxicity assay as described in Materials and Methods. NK cell–dependent lysis of B2MKO T cells from four independent donors that were nontransduced or expressed unique scHLA-E4 VL9 variants at 48 h after culture with NK cells at the 4:1 E:T ratio. Symbols represent seven independent NK cell donors in duplicate. For all data, bars represent mean and error bars indicate ±SEM. For (C), a Wilcoxon matched-pairs signed rank test was used to calculate significance. For (E)–(I), statistical significance between B2MKO T cells that were nontransduced and each scHLA-E4 VL9 variant was calculated using a Wilcoxon matched-pairs signed rank test, whereas the Friedman test with a Dunn test for multiple comparisons calculated statistical significance between the scHLA-E4 VL9 variants.

Close modal

We next determined the inhibitory potential of each scHLA-E4 variant to attenuate NK cell reactivity to B2MKO T cells. Nontransduced B2MKO T cells stimulated NKG2A+ NK cells responses, but importantly, each scHLA-E4 variant considerably decreased the frequency of CD107a+ (Fig. 3D, 3E) and TNF-α+ (Fig. 3D, 3F) NKG2A+ NK cells to levels comparable to stimulation with unmodified T cells. Moreover, each scHLA-E4 molecule exerted a substantial inhibitory effect such that the frequencies of responding NK cells were also reduced at the population level (Fig. 3G, 3H). Notably, the scHLA-E4 variants comprising the VL9 epitopes v1, v4, and v7 demonstrated enhanced inhibitory potential relative to other peptide variants (Fig. 3E, 3G). Interestingly, the scHLA-E4 variants comprising v1, v2, v4, and v7 peptides sustained the highest surface expression (Fig. 3B, 3C), but unlike the rest, the scHLA-E4v2 molecule did not exhibit enhanced inhibitory potential compared with the other variants (Fig. 3E, 3G). Lastly, all scHLA-E4 molecules, except scHLA-E4v8, attenuated NK cell–driven lysis of B2MKO T cells, even reducing lysis by as much as 50% on average relative to nontransduced B2MKO T cells (Fig. 3I). The scHLA-E4 variants presenting peptides from HLA-A (v3) and HLA-C (v4) allotypes prevented NK cell–dependent lysis of B2MKO T cells to a greater extent than scHLA-E4v2 and scHLA-E4v8. Taken together, these data indicate that selection of a VL9 epitope can substantially impact both the surface expression and inhibitory capacity of scHLA-E4 molecules.

We further modified the scHLA-E4 variants by incorporating mutations that stabilize the VL9 peptides into the HLA-E binding groove to improve expression and inhibitory potency. To do so, we introduced cysteine residues into the flexible (G4S)3 linker (GCGG(G4S)2) and tyrosine-84 (Y84C) of the HLA-E H chain (Fig. 4A). The resulting disulfide trap has been demonstrated to anchor relatively weak binding peptides and prolong cell surface half-life of scHLA class Ia trimers (35). The incorporation of the disulfide trap into each scHLA-E4 variant improved cell surface expression over their respective scHLA-E4 counterparts (Fig. 4B). Of note, dtHLA-E4v6 and dtHLA-E4v8 molecules, which comprised the VL9 epitope with the 2T substitutions, exhibited a 3- and 4-fold increase in surface expression, respectively (Fig. 4B). We then evaluated whether dtHLA-E4 variants could mitigate NK cell responses against B2MKO T cells. Indeed, each dtHLA-E4 variant reduced the frequency of CD107a+ and TNF-α+ NKG2A+ NK cells (Fig. 4C, 4D) and total NK cells (Supplemental Fig. 2A, 2B), as well as mitigated NK cell–driven lysis of B2MKO T cells (Fig. 4E). We previously showed that VL9 epitopes modulate the inhibitory potential of scHLA-E4 molecules (Fig. 3E, 3G, 3I), but now, we failed to detect any substantial difference in potency among the dtHLA-E4 variants (Fig. 4C, 4D), although B2MKO T cells expressing dtHLA-E4 molecules bound to v3 and v4 peptides exhibited the lowest degree of NK cell–dependent lysis (Fig. 4E).

FIGURE 4.

Disulfide trap mutations improve surface expression and NK cell inhibitory potential of scHLA-E4 variants. (A) Schematic of scHLA-E4 molecule that is either nonmutated or comprises the disulfide trap (dt) mutations in the (G4S)3 flexible linker and HLA-E H chain. (B) B2MKO T cells were transduced with an equivalent titer of lentivirus that encoded a unique scHLA-E4 and dtHLA-E4 VL9 variant. HLA-E cell surface expression was quantified as GMFI of HLA-E on HLA-E–positive B2MKO T cells that were transduced. Nontransduced B2MKO T cells served as a gating control to discriminate positive and negative HLA-E expression. Symbols represent four independent T cell donors in duplicate at 11 d posttransduction. (C and D) Frequency of CD107a+ (C) and TNF-α+ (D) NKG2A+ NK cells poststimulation with unmodified T cells or B2MKO T cells that were nontransduced or expressed a unique dtHLA-E4 VL9 variant from five (C) and three (D) independent donors. Symbols represent 12 (C) and 8 (D) independent NK cell donors in duplicate. (E) NK cell cytotoxicity assay as described in Materials and Methods. NK cell–dependent lysis of B2MKO T cells that were nontransduced or expressed a unique dtHLA-E4 VL9 variant from four independent donors at 48 h after culture with NK cells at the 4:1 E:T ratio. Symbols represent seven independent NK cell donors in duplicate. (F and G) Frequency of CD107a+ (F) and TNF-α+ (G) NKG2A+ NK cells poststimulation with B2MKO T cells that expressed a unique scHLA-E4 VL9 variant or the corresponding dtHLA-E4 VL9 variant from three independent donors. Symbols represent eight independent NK cell donors in duplicate. (H) NK cell cytotoxicity assay as described in Materials and Methods. NK cell–dependent lysis of B2MKO T cells that expressed a unique scHLA-E4 VL9 variant or the corresponding dtHLA-E4 VL9 variant from four independent donors at 48 h after culture with NK cells at the 4:1 E:T ratio. Symbols represent six independent NK cell donors in duplicate. For all data, bars represent mean and error bars indicate ±SEM. For (B)–(H), a Wilcoxon matched-pairs signed rank test was used to calculate significance. For (E), a Friedman test with a Dunn test for multiple comparisons calculated statistical significance between dtHLA-E4 VL9 variants.

FIGURE 4.

Disulfide trap mutations improve surface expression and NK cell inhibitory potential of scHLA-E4 variants. (A) Schematic of scHLA-E4 molecule that is either nonmutated or comprises the disulfide trap (dt) mutations in the (G4S)3 flexible linker and HLA-E H chain. (B) B2MKO T cells were transduced with an equivalent titer of lentivirus that encoded a unique scHLA-E4 and dtHLA-E4 VL9 variant. HLA-E cell surface expression was quantified as GMFI of HLA-E on HLA-E–positive B2MKO T cells that were transduced. Nontransduced B2MKO T cells served as a gating control to discriminate positive and negative HLA-E expression. Symbols represent four independent T cell donors in duplicate at 11 d posttransduction. (C and D) Frequency of CD107a+ (C) and TNF-α+ (D) NKG2A+ NK cells poststimulation with unmodified T cells or B2MKO T cells that were nontransduced or expressed a unique dtHLA-E4 VL9 variant from five (C) and three (D) independent donors. Symbols represent 12 (C) and 8 (D) independent NK cell donors in duplicate. (E) NK cell cytotoxicity assay as described in Materials and Methods. NK cell–dependent lysis of B2MKO T cells that were nontransduced or expressed a unique dtHLA-E4 VL9 variant from four independent donors at 48 h after culture with NK cells at the 4:1 E:T ratio. Symbols represent seven independent NK cell donors in duplicate. (F and G) Frequency of CD107a+ (F) and TNF-α+ (G) NKG2A+ NK cells poststimulation with B2MKO T cells that expressed a unique scHLA-E4 VL9 variant or the corresponding dtHLA-E4 VL9 variant from three independent donors. Symbols represent eight independent NK cell donors in duplicate. (H) NK cell cytotoxicity assay as described in Materials and Methods. NK cell–dependent lysis of B2MKO T cells that expressed a unique scHLA-E4 VL9 variant or the corresponding dtHLA-E4 VL9 variant from four independent donors at 48 h after culture with NK cells at the 4:1 E:T ratio. Symbols represent six independent NK cell donors in duplicate. For all data, bars represent mean and error bars indicate ±SEM. For (B)–(H), a Wilcoxon matched-pairs signed rank test was used to calculate significance. For (E), a Friedman test with a Dunn test for multiple comparisons calculated statistical significance between dtHLA-E4 VL9 variants.

Close modal

Given that the dtHLA-E4 variants exhibited comparable abilities to inhibit NK cells, we assessed whether the inclusion of the disulfide trap improved inhibitory activity over their scHLA-E4 counterparts. Inclusion of the disulfide trap mutations largely improved the ability of scHLA-E4 molecules to reduce the frequency of CD107a+ and/or TNF-α+ NKG2A+ NK cells (Fig. 4F, 4G) and total NK cells (Supplemental Fig. 2C, 2D) and reduce NK cell–dependent lysis of B2MKO T cells to a greater extent than their scHLA-E4 counterparts (Fig. 4H). Strikingly, the addition of the disulfide trap substantially enhanced the function of scHLA-E4 molecules comprising v2, v6, and v8 epitopes, which previously underperformed relative to the other scHLA-E4 molecules (Fig. 3E, 3G, 3I). However, the inclusion of the disulfide trap did not similarly augment NK cell inhibition of high-activity molecules, such as scHLA-E4v7 (Fig. 4F–H). Collectively, these data indicate that introducing the disulfide trap can enhance both the surface expression and inhibitory potency of scHLA-E4 variants comprising different VL9 epitopes. This effect was emphasized in VL9 epitopes from HLA-B allotypes with the 2T substitution, which is consistent with the ability of the disulfide trap to anchor low-affinity peptides.

HLA-E binds to the activating receptor NKG2C/CD94, albeit with lower affinity than NKG2A (36), so we investigated whether dtHLA-E4-driven activation of NKG2C+ NK cells could nullify the protective benefit of these molecules. We found that B2MKO T cells expressing dtHLA-E4v1 bound recombinant human NKG2C/CD94 protein greater than endogenous HLA-E by unmodified T cells (Fig. 5A), and each dtHLA-E4 variant markedly increased the frequency of degranulating NKG2C+ NK cells over stimulation with nontransduced B2MKO T cells (Fig. 5B, 5C). The dtHLA-E4v1 molecule induced the greatest NKG2C+ NK cell degranulation, whereas in comparison the dtHLA-E4 molecules comprising v3, v4, and v5 epitopes from HLA-A and HLA-C allotypes exhibited lower activation of NKG2C+ NK cells (Fig. 5C). Importantly, inclusion of the disulfide trap did not induce greater NKG2C+ NK cell degranulation compared with their matched scHLA-E4 variants (Supplemental Fig. 2E). Despite each dtHLA-E4 driving activation of NKG2C+ NK cells, all variants previously mitigated NK cell–driven lysis of B2MKO T cells (Fig. 4E), indicating that NKG2C+ NK cells were too infrequent to counter the inhibitory effect afforded by these molecules. Indeed, NKG2C expression among those donors (termed NKG2Clow donors) was <10% (0.25–6.02%), whereas NKG2A expression exceeded 50% (58.2–73.2%) of total NK cells (Fig. 5D, 5E). We next identified NKG2Chigh donors in which the frequency of NKG2C expression surpassed 10% (15–77.9%) of total NK cells (Fig. 5D, 5E) and then determined which dtHLA-E4 variants could avert rejection of B2MKO T cells. In contrast to our previous findings, now only dtHLA-E4 molecules comprising v3, v4, and v5 epitopes reduced NK cell–dependent lysis of B2MKO T cells by NKG2Chigh donors (Fig. 5F). Collectively, these findings support utilizing a dtHLA-E4 variant fused to either VMAPRTLVL (v3), VMAPRTLIL (v4), or VMAPRALLL (v5) to maximize NK cell inhibition through NKG2A engagement and decrease activation of NKG2C+ NK cells.

FIGURE 5.

dtHLA-E4 molecules bound to HLA-A and HLA-C–derived VL9 epitopes mitigate activation of NKG2C+ NK cells. (A) FACS plot indicates binding of recombinant human NKG2C/CD94 heterodimer to unmodified T cells and B2MKO T cells that expressed dtHLA-E4v1. (B and C) FACS plots (B) and summarized data (C) show the frequency of CD107a+NKG2C+ NK cells after stimulation with B2MKO T cells that were nontransduced or expressed a unique dtHLA-E4 VL9 variant from three independent donors. Symbols represent six independent NK cell donors in duplicate. (D and E) FACS plots (D) and summarized data (E) indicate the frequency of NKG2A and NKG2C expression of total NK cells from donors classified as NKG2Clow and NKG2Chigh based on frequency of NKG2C+ NK cells being less than or greater than 10%, respectively. Each symbol represents an independent NK cell donor. (F) NK cell cytotoxicity assay as described in Materials and Methods. NK cell–dependent lysis of B2MKO T cells that were nontransduced or expressed a unique dtHLA-E4 VL9 variant from three independent donors at 48 h after culture with NK cells from NKG2Chigh donors at the 4:1 E:T ratio. Symbols represent four independent NK cell donors in duplicate. For all data, bars represent mean and error bars indicate ±SEM. For (C) and (F), a Wilcoxon matched-pairs signed rank test was used to calculate significance between nontransduced and dtHLA-E4 variant–expressing B2MKO T cells. For (C), a Friedman test with a Dunn test for multiple comparisons calculated statistical significance between dtHLA-E4 VL9 variants.

FIGURE 5.

dtHLA-E4 molecules bound to HLA-A and HLA-C–derived VL9 epitopes mitigate activation of NKG2C+ NK cells. (A) FACS plot indicates binding of recombinant human NKG2C/CD94 heterodimer to unmodified T cells and B2MKO T cells that expressed dtHLA-E4v1. (B and C) FACS plots (B) and summarized data (C) show the frequency of CD107a+NKG2C+ NK cells after stimulation with B2MKO T cells that were nontransduced or expressed a unique dtHLA-E4 VL9 variant from three independent donors. Symbols represent six independent NK cell donors in duplicate. (D and E) FACS plots (D) and summarized data (E) indicate the frequency of NKG2A and NKG2C expression of total NK cells from donors classified as NKG2Clow and NKG2Chigh based on frequency of NKG2C+ NK cells being less than or greater than 10%, respectively. Each symbol represents an independent NK cell donor. (F) NK cell cytotoxicity assay as described in Materials and Methods. NK cell–dependent lysis of B2MKO T cells that were nontransduced or expressed a unique dtHLA-E4 VL9 variant from three independent donors at 48 h after culture with NK cells from NKG2Chigh donors at the 4:1 E:T ratio. Symbols represent four independent NK cell donors in duplicate. For all data, bars represent mean and error bars indicate ±SEM. For (C) and (F), a Wilcoxon matched-pairs signed rank test was used to calculate significance between nontransduced and dtHLA-E4 variant–expressing B2MKO T cells. For (C), a Friedman test with a Dunn test for multiple comparisons calculated statistical significance between dtHLA-E4 VL9 variants.

Close modal

Given that HLA-E–restricted CD8+ T cell responses against self and nonself VL9 epitopes have been identified from human donors (37–39), we hypothesized that supraphysiologic cell surface expression of dtHLA-E4 variants could elicit recipient CD8+ T cell responses to HLA-E–bound peptide ligands. To examine the immunogenic potential of dtHLA-E4 variants, we first generated aAPCs by transducing K-562 cells that are devoid of endogenous HLA expression (40) with lentiviruses encoding the CD86 costimulatory ligand and each dtHLA-E4 variant (Supplemental Fig. 3A, 3B). Next, T cells from a panel of human donors were stimulated with aAPCs, but we did not detect an increase in the frequency of responding CD8+ T cells after subtracting the signal from stimulation with K-562 cells expressing CD86 alone (K.86) (Fig. 6A). This finding suggests that the population of pre-existing HLA-E–specific T cells may be too infrequent for quantification, so we then leveraged the aAPC platform to expand these low-frequency precursors in vitro. One week after primary stimulation, these T cells were restimulated with their respective aAPCs and K.86 control cells, and notably we detected a substantial increase in the frequency of responding CD107a+ and TNF-α+ CD8+ T cells (Fig. 6B, 6C), but not CD4+ T cells (Supplemental Fig. 3C, 3D). These findings are indicative of functional dtHLA-E4–specific CD8+ T cell responses, which could limit the in vivo persistence of allogeneic B2MKO T cells expressing dtHLA-E4.

FIGURE 6.

Hypoimmunogenic dtHLA-E4v4 evades HLA-E–reactive CD8+ T cell responses. (A) Frequency of responding CD107a+ and TNF-α+ CD8+ T cells after in vitro stimulation with the indicated aAPC.dtHLA-E4 variant. (B and C) T cells were stimulated with aAPCs expressing a unique dtHLA-E4 variant and cultured for 1 wk. FACS plots (B) and summarized data (C) indicate the frequency of CD107a+ and TNFα+ CD8+ T cells after restimulation with the appropriate aAPC and K.86 cell lines. (D–F) T cells were stimulated with K.86 cells, aAPC.hypoA2, and aAPC.scA2 cells and then cultured for 1 wk. (D) Total CD8+ T cells were enumerated at 1 wk poststimulation. Dotted line indicates average CD8+ T cell count when cultured in the absence of K-562 cells. (E and F) Frequency of CD107a+ (E) and TNF-α+ (F) CD8+ T cells after restimulation with aAPC.scA2 and aAPC.hypoA2 cells. Symbols represents nine independent T cell donors. (G) Amino acid alignment of the α3 domain region for the indicated HLA class I alleles. (H and I) T cells were stimulated with aAPC.dtHLA-E4v4 cells for 1 wk in culture. FACS plots (H) and summarized data (I) indicate frequency of CD107a+ and TNF-α+ CD8+ T cells after restimulation with aAPC.dtHLA-E4v4 and aAPC.hypoHLA-E4v4 cells. (J) NK cell cytotoxicity assay as described in Materials and Methods. NK cell–dependent lysis of B2MKO T cells that were nontransduced or expressed dtHLA-E4v4 or hypoHLA-E4v4. Symbols represent mean of three independent NK cell donors in duplicate. For (A), (C), and (I), symbols represent nine independent T cell donors in duplicate. For (A), (C), (E), (F), and (I), values are background subtracted from restimulation with K.86 cells. For (E), (F), and (I), a Wilcoxon matched-pairs signed rank test was used to calculate significance. For all data, bars represent mean and error bars indicate ±SEM.

FIGURE 6.

Hypoimmunogenic dtHLA-E4v4 evades HLA-E–reactive CD8+ T cell responses. (A) Frequency of responding CD107a+ and TNF-α+ CD8+ T cells after in vitro stimulation with the indicated aAPC.dtHLA-E4 variant. (B and C) T cells were stimulated with aAPCs expressing a unique dtHLA-E4 variant and cultured for 1 wk. FACS plots (B) and summarized data (C) indicate the frequency of CD107a+ and TNFα+ CD8+ T cells after restimulation with the appropriate aAPC and K.86 cell lines. (D–F) T cells were stimulated with K.86 cells, aAPC.hypoA2, and aAPC.scA2 cells and then cultured for 1 wk. (D) Total CD8+ T cells were enumerated at 1 wk poststimulation. Dotted line indicates average CD8+ T cell count when cultured in the absence of K-562 cells. (E and F) Frequency of CD107a+ (E) and TNF-α+ (F) CD8+ T cells after restimulation with aAPC.scA2 and aAPC.hypoA2 cells. Symbols represents nine independent T cell donors. (G) Amino acid alignment of the α3 domain region for the indicated HLA class I alleles. (H and I) T cells were stimulated with aAPC.dtHLA-E4v4 cells for 1 wk in culture. FACS plots (H) and summarized data (I) indicate frequency of CD107a+ and TNF-α+ CD8+ T cells after restimulation with aAPC.dtHLA-E4v4 and aAPC.hypoHLA-E4v4 cells. (J) NK cell cytotoxicity assay as described in Materials and Methods. NK cell–dependent lysis of B2MKO T cells that were nontransduced or expressed dtHLA-E4v4 or hypoHLA-E4v4. Symbols represent mean of three independent NK cell donors in duplicate. For (A), (C), and (I), symbols represent nine independent T cell donors in duplicate. For (A), (C), (E), (F), and (I), values are background subtracted from restimulation with K.86 cells. For (E), (F), and (I), a Wilcoxon matched-pairs signed rank test was used to calculate significance. For all data, bars represent mean and error bars indicate ±SEM.

Close modal

One approach to disrupt CD8+ T cell recognition is to abrogate the CD8/HLA interaction by introducing mutations into critical CD8 contact sites within the α3 domain of the HLA class I H chain (4142). To demonstrate this, we generated aAPCs expressing scHLA-A*02 molecules comprising either the wild-type HLA-A*02 H chain (scA2) or the hypoimmunogenic α3 domain mutations D227K, T228A, and A245V (hypoA2) (Supplemental Fig. 3A, 3B). T cells from HLA-A*02+ and HLA-A*02 donors were then stimulated with each aAPC and K.86 cell line. As expected, aAPC.scA2 cells induced CD8+ T cell expansion mainly driven by T cells from mismatched HLA-A*02 donors (Fig. 6D). In contrast, T cells stimulated by aAPC.hypoA2 cells failed to expand to the same extent, and instead exhibited near equivalent persistence to T cells activated by K.86 control cells (Fig. 6D). CD4+ T cells did not expand in response to either aAPC, confirming that this response is mediated by HLA class I–restricted CD8+ T cells (Supplemental Fig. 3E). Importantly, the hypoA2 molecule did not restimulate responses by HLA-A*02-specific CD8+ T cells that were previously primed by aAPC.A2 cells (Fig. 6E, 6F). These data indicate that α3 domain mutations are sufficient to abrogate CD8-dependent T cell responses in the context of HLA-A*02.

Amino acid residues critical for CD8 binding to HLA class I are well conserved, and indeed, the α3 domain of HLA-E*01:03 contains the amino acids D227, T228, and A245 (Fig. 6G). Thus, we introduced these mutations into dtHLA-E4v4, which previously exhibited potent NK cell inhibitory potential, and then engineered aAPCs to express this molecule (Supplemental Fig. 3A, 3B). Importantly, the resulting hypoHLA-E4v4 substantially reduced the frequency of CD107a+ and TNF-α+ CD8+ T cells that were previously primed with aAPCs expressing the nonmutated dtHLA-E4v4 variant (Fig. 6H, 6I). Moreover, the incorporation of α3 mutations did not interfere with the ability of the dtHLA-E4v4 molecule to inhibit NK cell–dependent lysis of B2MKO T cells (Fig. 6J). Taken together, these data describe the development of hypoHLA-E4 that is capable of evading CD8-dependent T cell responses while retaining NK cell inhibitory capacity.

In this study, we systematically constructed a scHLA-E molecule that exhibited improved surface expression and enhanced NK cell inhibitory function, as well as decreased the risk of immunogenicity. Our scHLA-E comprised the CD4 transmembrane domain and truncated cytoplasmic region, as well as disulfide trap mutations to anchor an HLA class I signal peptide-derived VL9 epitope, and, lastly, mutations in the α3 domain of HLA-E to abrogate CD8 coreceptor binding. Forced expression of a hypoimmunogenic dtHLA-E4 molecule in B2MKO T cells substantially mitigated NK cell and CD8+ T cell reactivity. These findings support the utility of this molecule to increase the therapeutic potential of allogeneic CAR-T cells by extending their in vivo persistence.

NK cell activity is governed by the balance of activating and inhibitory signals, and attempts to restore tolerance to HLA class I–deficient cells have overexpressed inhibitory molecules, including HLA class Ia killer Ig-like receptor (KIR) ligands (43), HLA-G (11, 13), CD47 (1617), PD-L1 (16), and HLA-E (11, 13–15). However, the corresponding immune checkpoint receptors for HLA-G (LILRB1), CD47 (SIRPα), and PD-1 (PD-L1) are restricted to minor subpopulations of NK cells and/or require cytokine stimulation to induce meaningful expression at steady state (44–46), which challenges their ability to broadly inhibit NK cells. Likewise, forced expression of an HLA class Ia single-chain molecule may be insufficient to inhibit the total NK cell population given that NK cells display a variegated expression pattern of inhibitory KIRs (47, 48). The extensive genetic diversity of KIRs influences both KIR expression level and frequency of KIR+ NK cells (49). As a result, individual NK cells may only express a subset of available KIRs that can sense allogeneic cells with altered expression of HLA class I or that do not express the appropriate KIR ligand. Given that NKG2A inversely correlates with KIR expression (50, 51), NK cell repertoires characterized by low KIR expression or KIR-negative NK cells are reported to rely on NKG2A expression for functional competence (52). In these individuals, forced expression of a scHLA-E molecule may be more effective at inhibiting NK cells than in individuals with KIR-dominated repertoires. Notably, NK cell repertoires are likely skewed following lymphodepleting chemotherapy, which is required therapy prior to the infusion of CAR-T cells. Indeed, NK cells are first to recover after lymphodepleting chemotherapy (53, 54), and data following hematopoietic stem cell transplant indicate that early repopulating NK cells are predominately immature and ∼90% are NKG2A+ (55). These findings imply that targeting the NKG2A inhibitory pathway with an HLA-E–based strategy could inhibit most rebounding NK cells following conditioning, thereby enabling maximal CAR-T cell expansion, which is a correlate of long-term remission in patients with hematological malignancies (56).

To restore HLA-E expression in B2M-disrupted T cells, it was necessary to construct a scHLA-ET molecule that couples the HLA-E H chain, B2M L chain, and peptide ligand. Previous reports indicated that the scHLA-ET in this format exhibited short cell surface half-life and stability (23). This phenomenon may be due to the cytoplasmic tail of HLA-E, which promotes intracellular accumulation and rapid internalization relative to HLA class Ia molecules (32). We found that replacing the HLA-E transmembrane and cytoplasmic regions with the corresponding domains from CD8α decreased surface expression relative to scHLA-ET, which may reflect improper assembly due to the propensity of the CD8α transmembrane domain to homodimerize (57). In contrast, the scHLA-E4 molecule sustained the highest level of surface expression, which could be attributed to the truncated cytoplasmic region because critical serine residues were omitted that when phosphorylated mediate CD4 coreceptor internalization (58, 59). We suspect that sustaining high-level surface expression of scHLA-E4 will be necessary to maximize long-term protection of HLA class I–deficient T cells from NK cell–mediated lysis.

We then functionally characterized the scHLA-E4 molecule bound to the HLA-G*01 signal peptide-derived epitope VMAPRTLFL. However, polymorphisms within HLA class Ia signal peptides yield alternative VL9 epitopes that bind HLA-E and influence its cell surface stability and affinity for NKG2X/CD94 receptors (26). Indeed, scHLA-E4 VL9 variants were differentially expressed on the surface, and molecules bound to HLA-B allotype-derived epitopes with a 2T substitution, VTAPRTVLL (v6) and VTAPRTLLL (v8), exhibited the lowest surface expression. Notably, disulfide trap mutations improved the expression of all dtHLA-E4 VL9 variants, particularly for weak binding HLA-B allotype-derived epitopes. These findings are congruent with previous observations that disulfide bond engineering generates robust scHLA complexes by anchoring low-affinity epitopes into the HLA binding groove, thereby resisting displacement from high-affinity competitor peptides (35). Moreover, all dtHLA-E4 variants, except dtHLA-E4v5 and dtHLA-E4v7, exhibited greater NK cell inhibitory capacity than their scHLA-E4 counterparts, suggesting that surface stabilization of these molecules augmented their inhibitory potential.

The NK cell donors from our initial assays were biased toward NKG2A expression, but HLA-E also binds the NKG2C/CD94 heterodimer (20, 36) and may stimulate adaptive NKG2C+ NK cells. Adaptive NK cells expand in response to human CMV infection (60), meaning some individuals may harbor a sizable frequency of NKG2C+ NK cells that could nullifying the protective benefit of dtHLA-E4 molecules. Indeed, in NKG2Chigh NK cell donors the dtHLA-E4 variant comprising the VMAPRTLFL (v1) epitope drove substantial activation of NKG2C+ NK cells, whereas epitopes derived from HLA-A and HLA-C allotypes, VMAPRTLVL (v3), VMAPRTLIL (v4), and VMAPRALLL (v5), minimized the magnitude of their responses. These data substantiated previous findings (26) and indicate that HLA class Ia signal peptide-derived VL9 epitopes influence NKG2X/CD94 engagement.

Although HLA-E is minimally polymorphic, we suspected that forced expression of dtHLA-E4 VL9 variants may induce detrimental CD8+ T cell–driven responses. For example, HLA-E–specific T cells may recognize, on allogeneic cells, HLA-E bound to nonself peptides (39), including the human CMV UL40 VL9 mimic epitopes VMAPRTLLL (v2), VMAPRTLVL (v3), and VMAPRTLIL (v4) (3738). Moreover, it is possible to activate self-reactive T cells by providing abundant stimulation (61), which suggests that supraphysiologic cell surface expression of dtHLA-E4 molecules presenting VL9 epitopes of host origin could also stimulate autoreactive CD8+ T cells. To this end, we detected in vitro HLA-E–specific T cell responses to aAPCs expressing dtHLA-E4 VL9 variants, which necessitated additional protein engineering to mitigate their immunogenicity. CD8 serves a critical role in the generation of HLA class I–restricted T cell responses, particularly when T cells display low-affinity autoreactive or alloreactive TCRs (62–65). Indeed, disruption of the CD8–HLA interaction using anti-CD8 nondepleting Abs has ameliorated pathogenesis of autoimmune type 1 diabetes (66–68), suggesting that a similar approach could prevent priming of dtHLA-E4–specific CD8+ T cells. Thus, we introduced mutations (D227K/T228/A245V) into the α3 domain of HLA-E. These mutations have been previously described to abolish CD8 binding to HLA class Ia molecules (41, 42), and when incorporated into dtHLA-E4v4 these mutations substantially reduced CD8+ T cell reactivity without compromising its ability to inhibit NK cell–mediated lysis.

Notably, if the TCR–dtHLA-E4 interaction is of sufficient affinity, it may obviate CD8 coreceptor dependence (64, 69), thereby rendering these allospecific T cells insensitive to the α3 domain mutations. TCRs have been reported to interact with HLA-E bound to self-peptides with micromolar affinity; however, these T cells are hyporesponsive, as their activity is regulated by the expression of HLA-C–specific inhibitory KIRs (70). In our setting, it is conceivable that the activation of CD8-independent HLA-E–reactive T cells could eliminate allogeneic cells expressing dtHLA-E4, especially upon B2M disruption, which abrogates expression of inhibitory KIR ligands. Thus, it may be necessary to include a compensatory approach to inhibit recipient T cell–mediated allorejection. We recently demonstrated that short-course immunosuppressant therapy comprising rapamycin or tacrolimus permitted the engraftment and expansion of HLA-mismatched CAR-T cells in lymphoreplete humanized mice (71). Genetic inactivation of FKBP1A was necessary to render CAR-T cells drug-resistant, and this strategy could be combined with multiplex base editing of TCR and B2M to enable treatment with allogeneic CAR-T cells expressing dtHLA-E4.

dtHLA-E4 variants bound to VMAPRTLVL (v3), VMAPRTLIL (v4), or VMAPRALLL (v5) substantially protected B2MKO T cells from lysis by NK cells that were biased for either NKG2A or NKG2C expression. As well, the insertion of mutations into the HLA-E α3 domain was necessary to mitigate the likelihood of stimulating host CD8+ T cell responses. Although we evaluated these dtHLA-E4 variants in allogeneic T cells, forced expression of these molecules may also facilitate the stable engraftment of pluripotent stem cells (25), as well as allogeneic and xenogeneic tissues (23, 24, 72). However, it may be necessary to enhance resistance to NK cell–driven rejection, particularly against cytolytic NKG2AKIR+CD56dim NK cell subsets. To accomplish this, we could envision combining expression of a hypoHLA-E4 molecule with multiplex base editing of NK cell adhesion (12) and activating ligands (1173), which have demonstrated the ability on their own to inhibit NK cell alloreactivity. Collectively, these findings demonstrate the systematic development and comprehensive in vitro characterization of hypoimmunogenic dtHLA-E4 molecules to increase the therapeutic potential of allogeneic cell therapies by overcoming allorejection.

All authors were employees of Beam Therapeutics when the work was conducted and are shareholders in the company. Beam Therapeutics has filed patent applications based on this work. Funding was provided by Beam Therapeutics.

We thank J. Decker and the Lab Automation team at Beam Therapeutics for next-generation sequencing support. We acknowledge W. Schmidt, N. Mastrangelo and E. Chiang for facilitating patent filings based on this work. Some figures were created using BioRender.com.

The online version of this article contains supplemental material.

aAPC

artificial APC

B2M

β2-microglobulin

B2MKO

B2M knockout

CAR

chimeric Ag receptor

dtHLA-E4

disulfide trap HLA-E4

KIR

killer Ig-like receptor

mDHFR

methotrexate-resistant dihydrofolate reductase

MMC

mitomycin C

MTX

methotrexate

scHLA

single-chain HLA

sgRNA

single-guide RNA

1
Das
,
R. K.
,
L.
Vernau
,
S. A.
Grupp
,
D. M.
Barrett
.
2019
.
Naïve T-cell deficits at diagnosis and after chemotherapy impair cell therapy potential in pediatric cancers
.
Cancer Discov.
9
:
492
499
.
2
Singh
,
N.
,
J.
Perazzelli
,
S. A.
Grupp
,
D. M.
Barrett
.
2016
.
Early memory phenotypes drive T cell proliferation in patients with pediatric malignancies
.
Sci. Transl. Med.
8
:
320ra3
.
3
Baguet
,
C.
,
J.
Larghero
,
M.
Mebarki
.
2024
.
Early predictive factors of failure in autologous CAR T-cell manufacturing and/or efficacy in hematologic malignancies
.
Blood Adv.
8
:
337
342
.
4
Depil
,
S.
,
P.
Duchateau
,
S. A.
Grupp
,
G.
Mufti
,
L.
Poirot
.
2020
.
“Off-the-shelf” allogeneic CAR T cells: development and challenges
.
Nat. Rev. Drug Discov.
19
:
185
199
.
5
Kernan
,
N. A.
,
N. H.
Collins
,
L.
Juliano
,
T.
Cartagena
,
B.
Dupont
,
R. J.
O’Reilly
.
1986
.
Clonable T lymphocytes in T cell-depleted bone marrow transplants correlate with development of graft-v-host disease
.
Blood
68
:
770
773
.
6
Qasim
,
W.
,
H.
Zhan
,
S.
Samarasinghe
,
S.
Adams
,
P.
Amrolia
,
S.
Stafford
,
K.
Butler
,
C.
Rivat
,
G.
Wright
,
K.
Somana
, et al
2017
.
Molecular remission of infant B-ALL after infusion of universal TALEN gene-edited CAR T cells
.
Sci. Transl. Med.
9
:
eaaj2013
.
7
Philip
,
L. P. B.
,
C.
Schiffer-Mannioui
,
D.
Le Clerre
,
I.
Chion-Sotinel
,
S.
Derniame
,
P.
Potrel
,
C.
Bas
,
L.
Lemaire
,
R.
Galetto
,
C.
Lebuhotel
, et al
2015
.
Multiplex genome-edited T-cell manufacturing platform for “off-the-shelf” adoptive T-cell immunotherapies
.
Cancer Res.
75
:
3853
3864
.
8
Kagoya
,
Y.
,
T.
Guo
,
B.
Yeung
,
K.
Saso
,
M.
Anczurowski
,
C. H.
Wang
,
K.
Murata
,
K.
Sugata
,
H.
Saijo
,
Y.
Matsunaga
, et al
2020
.
Genetic ablation of HLA class I, class II, and the T-cell receptor enables allogeneic T cells to be used for adoptive T-cell therapy
.
Cancer Immunol. Res.
8
:
926
936
.
9
Lee
,
J.
,
J. H.
Sheen
,
O.
Lim
,
Y.
Lee
,
J.
Ryu
,
D.
Shin
,
Y. Y.
Kim
,
M.
Kim
.
2020
.
Abrogation of HLA surface expression using CRISPR/Cas9 genome editing: a step toward universal T cell therapy
.
Sci. Rep.
10
:
17753
.
10
Kärre
,
K.
,
H. G.
Ljunggren
,
G.
Piontek
,
R.
Kiessling
.
1986
.
Selective rejection of H-2-deficient lymphoma variants suggests alternative immune defence strategy
.
Nature
319
:
675
678
.
11
Wang
,
B.
,
S.
Iriguchi
,
M.
Waseda
,
N.
Ueda
,
T.
Ueda
,
H.
Xu
,
A.
Minagawa
,
A.
 
Ishikawa
,
H.
Yano
,
T.
Ishi
, et al
2021
.
Generation of hypoimmunogenic T cells from genetically engineered allogeneic human induced pluripotent stem cells
.
Nat. Biomed. Eng.
5
:
429
440
.
12
Hammer
,
Q.
,
K.
Perica
,
R. M.
Mbofung
,
H.
van Ooijen
,
K. E.
Martin
,
P.
 
Momayyezi
,
E.
Varady
,
Y.
Pan
,
M.
Jelcic
,
B.
Groff
, et al
2024
.
Genetic ablation of adhesion ligands mitigates rejection of allogeneic cellular immunotherapies
.
Cell Stem Cell
31
:
1376
1386.e8
.
13
Guo
,
Y.
,
B.
Xu
,
Z.
Wu
,
J.
Bo
,
C.
Tong
,
D.
Chen
,
J.
Wang
,
H.
Wang
,
Y.
Wang
,
W.
Han
.
2021
.
Mutant B2M-HLA-E and B2M-HLA-G fusion proteins protects universal chimeric antigen receptor-modified T cells from allogeneic NK cell-mediated lysis
.
Eur. J. Immunol.
51
:
2513
2521
.
14
Li
,
W.
,
X.
Zhu
,
Y.
Xu
,
J.
Chen
,
H.
Zhang
,
Z.
Yang
,
Y.
Qi
,
J.
Hong
,
Y.
Li
,
G.
 
Wang
, et al
2022
.
Simultaneous editing of TCR, HLA-I/II and HLA-E resulted in enhanced universal CAR-T resistance to allo-rejection
.
Front. Immunol.
13
:
1052717
.
15
Jo
,
S.
,
S.
Das
,
A.
Williams
,
A. S.
Chretien
,
T.
Pagliardini
,
A.
Le Roy
,
J. P.
Fernandez
,
D.
Le Clerre
,
B.
Jahangiri
,
I.
Chion-Sotinel
, et al
2022
.
Endowing universal CAR T-cell with immune-evasive properties using TALEN-gene editing
.
Nat. Commun.
13
:
3453
.
16
Han
,
X.
,
M.
Wang
,
S.
Duan
,
P. J.
Franco
,
J. H.-R.
Kenty
,
P.
Hedrick
,
Y.
Xia
,
A.
Allen
,
L. M. R.
Ferreira
,
J. L.
Strominger
, et al
2019
.
Generation of hypoimmunogenic human pluripotent stem cells
.
Proc. Natl. Acad. Sci. USA
116
:
10441
10446
.
17
Hu
,
X.
,
K.
Manner
,
R.
DeJesus
,
K.
White
,
C.
Gattis
,
P.
Ngo
,
C.
Bandoro
,
E.
 
Tham
,
E. Y.
Chu
,
C.
Young
, et al
2023
.
Hypoimmune anti-CD19 chimeric antigen receptor T cells provide lasting tumor control in fully immunocompetent allogeneic humanized mice
.
Nat. Commun.
14
:
2020
.
18
Felício
,
L. P.
,
I. O. P.
Porto
,
C. T.
Mendes-Junior
,
L. C.
Veiga-Castelli
,
K. E.
Santos
,
R. P.
Vianello-Brondani
,
A.
Sabbagh
,
P.
Moreau
,
E. A.
Donadi
,
E. C.
Castelli
.
2014
.
Worldwide HLA-E nucleotide and haplotype variability reveals a conserved gene for coding and 3′ untranslated regions
.
Tissue Antigens
83
:
82
93
.
19
Lee
,
N.
,
M.
Llano
,
M.
Carretero
,
A.
Ishitani
,
F.
Navarro
,
M.
López-Botet
,
D. E.
Geraghty
.
1998
.
HLA-E is a major ligand for the natural killer inhibitory receptor CD94/NKG2A
.
Proc. Natl. Acad. Sci. USA
95
:
5199
5204
.
20
Braud
,
V. M.
,
D. S. J.
Allan
,
C. A.
O’Callaghan
,
K.
Söderström
,
A.
D’Andrea
,
G. S.
Ogg
,
S.
Lazetic
,
N. T.
Young
,
J. I.
Bell
,
J. H.
Phillips
, et al
1998
.
HLA-E binds to natural killer cell receptors CD94/NKG2A, B and C
.
Nature
391
:
795
799
.
21
Manser
,
A. R.
,
M.
Uhrberg
.
2016
.
Age-related changes in natural killer cell repertoires: impact on NK cell function and immune surveillance
.
Cancer Immunol. Immunother.
65
:
417
426
.
22
Mahapatra
,
S.
,
E. M.
Mace
,
C. G.
Minard
,
L. R.
Forbes
,
A.
Vargas-Hernandez
,
T. K.
Duryea
,
G.
Makedonas
,
P. P.
Banerjee
,
W. T.
Shearer
,
J. S.
Orange
.
2017
.
High-resolution phenotyping identifies NK cell subsets that distinguish healthy children from adults
.
PLoS One
12
:
e0181134
.
23
Crew
,
M. D.
,
M. J.
Cannon
,
B.
Phanavanh
,
C. N.
Garcia-Borges
.
2005
.
An HLA-E single chain trimer inhibits human NK cell reactivity towards porcine cells
.
Mol. Immunol.
42
:
1205
1214
.
24
Lilienfeld
,
B. G.
,
M. D.
Crew
,
P.
Forte
,
B. C.
Baumann
,
J. D.
Seebach
.
2007
.
Transgenic expression of HLA-E single chain trimer protects porcine endothelial cells against human natural killer cell-mediated cytotoxicity
.
Xenotransplantation
14
:
126
134
.
25
Gornalusse
,
G. G.
,
R. K.
Hirata
,
S. E.
Funk
,
L.
Riolobos
,
V. S.
Lopes
,
G.
Manske
,
D.
Prunkard
,
A. G.
Colunga
,
L.-A.
Hanafi
,
D. O.
Clegg
, et al
2017
.
HLA-E-expressing pluripotent stem cells escape allogeneic responses and lysis by NK cells
.
Nat. Biotechnol.
35
:
765
772
.
26
Lin
,
Z.
,
A. A.
Bashirova
,
M.
Viard
,
L.
Garner
,
M.
Quastel
,
M.
Beiersdorfer
,
W. K.
Kasprzak
,
M.
Akdag
,
Y.
Yuki
,
P.
Ojeda
, et al
2023
.
HLA class I signal peptide polymorphism determines the level of CD94/NKG2-HLA-E-mediated regulation of effector cell responses
.
Nat. Immunol.
24
:
1087
1097
.
27
Jonnalagadda
,
M.
,
C. E.
Brown
,
W. C.
Chang
,
J. R.
Ostberg
,
S. J.
Forman
,
M. C.
Jensen
.
2013
.
Efficient selection of genetically modified human T cells using methotrexate-resistant human dihydrofolate reductase
.
Gene Ther.
20
:
853
860
.
28
Diorio
,
C.
,
R.
Murray
,
M.
Naniong
,
L.
Barrera
,
A.
Camblin
,
J.
Chukinas
,
L.
Coholan
,
A.
Edwards
,
T.
Fuller
,
C.
Gonzales
, et al
2022
.
Cytosine base editing enables quadruple-edited allogeneic CART cells for T-ALL
.
Blood
140
:
619
629
.
29
Shaw
,
A. S.
,
J.
Chalupny
,
J. A.
Whitney
,
C.
Hammond
,
K. E.
Amrein
,
P.
Kavathas
,
B. M.
Sefton
,
J. K.
Rose
.
1990
.
Short related sequences in the cytoplasmic domains of CD4 and CD8 mediate binding to the amino-terminal domain of the p56lck tyrosine protein kinase
.
Mol. Cell. Biol.
10
:
1853
1862
.
30
Turner
,
J. M.
,
M. H.
Brodsky
,
B. A.
Irving
,
S. D.
Levin
,
R. M.
Perlmutter
,
D. R.
Littman
.
1990
.
Interaction of the unique N-terminal region of tyrosine kinase p56lck with cytoplasmic domains of CD4 and CD8 is mediated by cysteine motifs
.
Cell
60
:
755
765
.
31
Shaw
,
A. S.
,
K. E.
Amrein
,
C.
Hammond
,
D. F.
Stern
,
B. M.
Sefton
,
J. K.
Rose
.
1989
.
The lck tyrosine protein kinase interacts with the cytoplasmic tail of the CD4 glycoprotein through its unique amino-terminal domain
.
Cell
59
:
627
636
.
32
He
,
W.
,
E.
Gea-Mallorquí
,
H.
Colin-York
,
M.
Fritzsche
,
G. M.
Gillespie
,
S.
 
Brackenridge
,
P.
Borrow
,
A. J.
McMichael
.
2023
.
Intracellular trafficking of HLA-E and its regulation
.
J. Exp. Med.
220
:
e20221941
.
33
Braud
,
V. M.
,
D. S.
Allan
,
D.
Wilson
,
A. J.
McMichael
.
1998
.
TAP- and tapasin-dependent HLA-E surface expression correlates with the binding of an MHC class I leader peptide
.
Curr. Biol.
8
:
1
10
.
34
Braud
,
V.
,
E. Y.
Jones
,
A.
McMichael
.
1997
.
The human major histocompatibility complex class Ib molecule HLA-E binds signal sequence-derived peptides with primary anchor residues at positions 2 and 9
.
Eur. J. Immunol.
27
:
1164
1169
.
35
Truscott
,
S. M.
,
L.
Lybarger
,
J. M.
Martinko
,
V. E.
Mitaksov
,
D. M.
Kranz
,
J. M.
Connolly
,
D. H.
Fremont
,
T. H.
Hansen
.
2007
.
Disulfide bond engineering to trap peptides in the MHC class I binding groove
.
J. Immunol.
178
:
6280
6289
.
36
Valés-Gómez
,
M.
,
H. T.
Reyburn
,
R. A.
Erskine
,
M.
López-Botet
,
J. L.
Strominger
.
1999
.
Kinetics and peptide dependency of the binding of the inhibitory NK receptor CD94/NKG2-A and the activating receptor CD94/NKG2-C to HLA-E
.
EMBO J.
18
:
4250
4260
.
37
Pietra
,
G.
,
C.
Romagnani
,
P.
Mazzarino
,
M.
Falco
,
E.
Millo
,
A.
Moretta
,
L.
Moretta
,
M. C.
Mingari
.
2003
.
HLA-E-restricted recognition of cytomegalovirus-derived peptides by human CD8+ cytolytic T lymphocytes
.
Proc. Natl. Acad. Sci. USA
100
:
10896
10901
.
38
Mazzarino
,
P.
,
G.
Pietra
,
P.
Vacca
,
M.
Falco
,
D.
Colau
,
P.
Coulie
,
L.
Moretta
,
M. C.
Mingari
.
2005
.
Identification of effector-memory CMV-specific T lymphocytes that kill CMV-infected target cells in an HLA-E-restricted fashion
.
Eur. J. Immunol.
35
:
3240
3247
.
39
Romagnani
,
C.
,
G.
Pietra
,
M.
Falco
,
E.
Millo
,
P.
Mazzarino
,
R.
Biassoni
,
A.
Moretta
,
L.
Moretta
,
M. C.
Mingari
.
2002
.
Identification of HLA-E-specific alloreactive T lymphocytes: a cell subset that undergoes preferential expansion in mixed lymphocyte culture and displays a broad cytolytic activity against allogeneic cells
.
Proc. Natl. Acad. Sci. USA
99
:
11328
11333
.
40
Turtle
,
C. J.
,
S. R.
Riddell
.
2010
.
Artificial antigen-presenting cells for use in adoptive immunotherapy
.
Cancer J.
16
:
374
381
.
41
Gao
,
G. F.
,
B. E.
Willcox
,
J. R.
Wyer
,
J. M.
Boulter
,
C. A.
O’Callaghan
,
K.
Maenaka
,
D. I.
Stuart
,
E.
Jones
,
P.
Van Der Merwe
,
J. I.
Bell
,
B. K.
Jakobsen
.
2000
.
Classical and nonclassical class I major histocompatibility complex molecules exhibit subtle conformational differences that affect binding to CD8αα
.
J. Biol. Chem.
275
:
15232
15238
.
42
Purbhoo
,
M. A.
,
J. M.
Boulter
,
D. A.
Price
,
A. L.
Vuidepot
,
C. S.
Hourigan
,
P. R.
Dunbar
,
K.
Olson
,
S. J.
Dawson
,
R. E.
Phillips
,
B. K.
Jakobsen
, et al
2001
.
The human CD8 coreceptor effects cytotoxic T cell activation and antigen sensitivity primarily by mediating complete phosphorylation of the T cell receptor ζ chain
.
J. Biol. Chem.
276
:
32786
32792
.
43
Seebach
,
J. D.
,
C.
Comrack
,
S.
Germana
,
C.
LeGuern
,
D. H.
Sachs
,
H.
DerSimonian
.
1997
.
HLA-Cw3 expression on porcine endothelial cells protects against xenogeneic cytotoxicity mediated by a subset of human NK cells
.
J. Immunol.
159
:
3655
3661
.
44
Deuse
,
T.
,
X.
Hu
,
S.
Agbor-Enoh
,
M. K.
Jang
,
M.
Alawi
,
C.
Saygi
,
A.
Gravina
,
G.
Tediashvili
,
V. Q.
Nguyen
,
Y.
Liu
, et al
2021
.
The SIRPα-CD47 immune checkpoint in NK cells
.
J. Exp. Med.
218
:
e20200839
.
45
Pesce
,
S.
,
M.
Greppi
,
G.
Tabellini
,
F.
Rampinelli
,
S.
Parolini
,
D.
Olive
,
L.
Moretta
,
A.
Moretta
,
E.
Marcenaro
.
2017
.
Identification of a subset of human natural killer cells expressing high levels of programmed death 1: a phenotypic and functional characterization
.
J. Allergy Clin. Immunol.
139
:
335
346.e3
.
46
Davidson
,
C. L.
,
N. L.
Li
,
D. N.
Burshtyn
.
2010
.
LILRB1 polymorphism and surface phenotypes of natural killer cells
.
Hum. Immunol.
71
:
942
949
.
47
Valiante
,
N. M.
,
M.
Uhrberg
,
H. G.
Shilling
,
K.
Lienert-Weidenbach
,
K. L.
Arnett
,
A.
D’Andrea
,
J. H.
Phillips
,
L. L.
Lanier
,
P.
Parham
.
1997
.
Functionally and structurally distinct NK cell receptor repertoires in the peripheral blood of two human donors
.
Immunity
7
:
739
751
.
48
Brennan
,
J.
,
D.
Mager
,
W.
Jefferies
,
F.
Takei
.
1994
.
Expression of different members of the Ly-49 gene family defines distinct natural killer cell subsets and cell adhesion properties
.
J. Exp. Med.
180
:
2287
2295
.
49
Yawata
,
M.
,
N.
Yawata
,
M.
Draghi
,
A.-M.
Little
,
F.
Partheniou
,
P.
Parham
.
2006
.
Roles for HLA and KIR polymorphisms in natural killer cell repertoire selection and modulation of effector function
.
J. Exp. Med.
203
:
633
645
.
50
Fauriat
,
C.
,
S.
Andersson
,
A. T.
Björklund
,
M.
Carlsten
,
M.
Schaffer
,
N. K.
Björkström
,
B. C.
Baumann
,
J.
Michaëlsson
,
H.-G.
Ljunggren
,
K.-J.
Malmberg
.
2008
.
Estimation of the size of the alloreactive NK cell repertoire: studies in individuals homozygous for the group A KIR haplotype
.
J. Immunol.
181
:
6010
6019
.
51
Shilling
,
H. G.
,
N.
Young
,
L. A.
Guethlein
,
N. W.
Cheng
,
C. M.
Gardiner
,
D.
Tyan
,
P.
Parham
.
2002
.
Genetic control of human NK cell repertoire
.
J. Immunol.
169
:
239
247
.
52
Andersson
,
S.
,
C.
Fauriat
,
J.-A.
Malmberg
,
H.-G.
Ljunggren
,
K.-J.
Malmberg
.
2009
.
KIR acquisition probabilities are independent of self-HLA class I ligands and increase with cellular KIR expression
.
Blood
114
:
95
104
.
53
Galli
,
E.
,
A.
Fresa
,
S.
Bellesi
,
E.
Metafuni
,
E.
Maiolo
,
I.
Pansini
,
F.
Frioni
,
F.
 
Autore
,
M. A.
Limongiello
,
I.
Innocenti
, et al
2024
.
Hematopoiesis and immune reconstitution after CD19 directed chimeric antigen receptor T-cells (CAR-T): a comprehensive review on incidence, risk factors and current management
.
Eur. J. Haematol.
112
:
184
196
.
54
Wang
,
Y.
,
H.
Li
,
X.
Song
,
K.
Qi
,
H.
Cheng
,
J.
Cao
,
M.
Shi
,
Z.
Yan
,
G.
Jing
,
B.
 
Pan
, et al
2021
.
Kinetics of immune reconstitution after anti-CD19 chimeric antigen receptor T cell therapy in relapsed or refractory acute lymphoblastic leukemia patients
.
Int. J. Lab. Hematol.
43
:
250
258
.
55
Orrantia
,
A.
,
I.
Terrén
,
G.
Astarloa-Pando
,
C.
González
,
A.
Uranga
,
J. J.
Mateos-Mazón
,
J. C.
García-Ruiz
,
M.
Riñón
,
M.
Rey
,
S.
Pérez-Fernandez
, et al
2021
.
NK cell reconstitution after autologous hematopoietic stem cell transplantation: association between NK cell maturation stage and outcome in multiple myeloma
.
Front. Immunol.
12
:
748207
.
56
Cappell
,
K. M.
,
J. N.
Kochenderfer
.
2023
.
Long-term outcomes following CAR T cell therapy: what we know so far
.
Nat. Rev. Clin. Oncol.
20
:
359
371
.
57
Hennecke
,
S.
,
P.
Cosson
.
1993
.
Role of transmembrane domains in assembly and intracellular transport of the CD8 molecule
.
J. Biol. Chem.
268
:
26607
26612
.
58
Pitcher
,
C.
,
S.
Höning
,
A.
Fingerhut
,
K.
Bowers
,
M.
Marsh
.
1999
.
Cluster of differentiation antigen 4 (CD4) endocytosis and adaptor complex binding require activation of the CD4 endocytosis signal by serine phosphorylation
.
Mol. Biol. Cell
10
:
677
691
.
59
Shin
,
J.
,
C.
Doyle
,
Z.
Yang
,
D.
Kappes
,
J. L.
Strominger
.
1990
.
Structural features of the cytoplasmic region of CD4 required for internalization
.
EMBO J.
9
:
425
434
.
60
Gumá
,
M.
,
A.
Angulo
,
C.
Vilches
,
N.
Gómez-Lozano
,
N.
Malats
,
M.
López-Botet
.
2004
.
Imprint of human cytomegalovirus infection on the NK cell receptor repertoire
.
Blood
104
:
3664
3671
.
61
Tallquist
,
M. D.
,
A. J.
Weaver
,
L. R.
Pease
.
1998
.
Degenerate recognition of alloantigenic peptides on a positive-selecting class I molecule
.
J. Immunol.
160
:
802
809
.
62
Holler
,
P. D.
,
D. M.
Kranz
.
2003
.
Quantitative analysis of the contribution of TCR/pepMHC affinity and CD8 to T cell activation
.
Immunity
18
:
255
264
.
63
Garcia
,
K. C.
,
M. D.
Tallquist
,
L. R.
Pease
,
A.
Brunmark
,
C. A.
Scott
,
M.
Degano
,
E. A.
Stura
,
P. A.
Peterson
,
I. A.
Wilson
,
L.
Teyton
.
1997
.
αβ T cell receptor interactions with syngeneic and allogeneic ligands: affinity measurements and crystallization
.
Proc. Natl. Acad. Sci. USA
94
:
13838
13843
.
64
Laugel
,
B.
,
H. A.
van den Berg
,
E.
Gostick
,
D. K.
Cole
,
L.
Wooldridge
,
J.
Boulter
,
A.
Milicic
,
D. A.
Price
,
A. K.
Sewell
.
2007
.
Different T cell receptor affinity thresholds and CD8 coreceptor dependence govern cytotoxic T lymphocyte activation and tetramer binding properties
.
J. Biol. Chem.
282
:
23799
23810
.
65
Bridgeman
,
J. S.
,
A. K.
Sewell
,
J. J.
Miles
,
D. A.
Price
,
D. K.
Cole
.
2012
.
Structural and biophysical determinants of αβ T-cell antigen recognition
.
Immunology
135
:
9
18
.
66
Phillips
,
J. M.
,
N. M.
Parish
,
T.
Raine
,
C.
Bland
,
Y.
Sawyer
,
H.
De La Peña
,
A.
Cooke
.
2009
.
Type 1 diabetes development requires both CD4+ and CD8+ T cells and can be reversed by non-depleting antibodies targeting both T cell populations
.
Rev. Diabet. Stud.
6
:
97
103
.
67
Yi
,
Z.
,
R.
Diz
,
A. J.
Martin
,
Y. M.
Morillon
,
D. E.
Kline
,
L.
Li
,
B.
Wang
,
R.
 
Tisch
.
2012
.
Long-term remission of diabetes in NOD mice is induced by nondepleting anti-CD4 and anti-CD8 antibodies
.
Diabetes
61
:
2871
2880
.
68
Clement
,
M.
,
J. A.
Pearson
,
S.
Gras
,
H. A.
van den Berg
,
A.
Lissina
,
S.
Llewellyn-Lacey
,
M. D.
Willis
,
T.
Dockree
,
J. E.
McLaren
,
J.
Ekeruche-Makinde
, et al
2016
.
Targeted suppression of autoreactive CD8+ T-cell activation using blocking anti-CD8 antibodies
.
Sci. Rep.
6
:
35332
.
69
Cole
,
D. K.
,
B.
Laugel
,
M.
Clement
,
D. A.
Price
,
L.
Wooldridge
,
K.
Andrew
.
2012
.
The molecular determinants of CD8 co-receptor function
.
Immunology
137
:
139
148
.
70
Sullivan
,
L. C.
,
T. H. O.
Nguyen
,
C. M.
Harpur
,
S.
Stankovic
,
A. R.
Kanagarajah
,
M.
Koutsakos
,
P. M.
Saunders
,
Z.
Cai
,
J. A.
Gray
,
J. M. L.
Widjaja
, et al
2021
.
Natural killer cell receptors regulate responses of HLA-E-restricted T cells
.
Sci. Immunol.
6
:
eabe9057
.
71
Maldini
,
C. R.
,
A. C.
Messana
,
P. B.
Bendet
,
A. J.
Camblin
,
F. M.
Musenge
,
M. L.
White
,
J. J.
Rocha
,
L. J.
Coholan
,
C.
Karaca
,
F.
Li
, et al
2024
.
Immunosuppressant therapy averts rejection of allogeneic FKBP1A-disrupted CAR-T cells
.
Mol. Ther.
32
:
3485
3503
.
72
Cross-Najafi
,
A. A.
,
K.
Farag
,
A.
Isidan
,
W.
Li
,
W.
Zhang
,
Z.
Lin
,
J. R.
Walsh
,
K.
Lopez
,
Y.
Park
,
N. G.
Higgins
, et al
2023
.
Co-expression of HLA-E and HLA-G on genetically modified porcine endothelial cells attenuates human NK cell-mediated degranulation
.
Front. Immunol.
14
:
1217809
.
73
Chimienti
,
R.
,
T.
Baccega
,
S.
Torchio
,
F.
Manenti
,
S.
Pellegrini
,
A.
Cospito
,
A.
 
Amabile
,
M. T.
Lombardo
,
P.
Monti
,
V.
Sordi
, et al
2022
.
Engineering of immune checkpoints B7-H3 and CD155 enhances immune compatibility of MHC-I−/− iPSCs for β cell replacement
.
Cell Rep.
40
:
111423
.

Supplementary data