Abstract
Immune checkpoint blockade therapy has been successful in treating some types of cancers but has not shown clinical benefits for treating leukemia 1. This result suggests that leukemia exploits unique escape mechanisms. Certain immune inhibitory receptors that are expressed by normal immune cells are also present on leukemia cells. It remains unknown whether these receptors can initiate immune-related primary signaling in tumor cells. Here we show that LILRB4, an ITIM-containing receptor and a monocytic leukemia marker, supports tumor cell infiltration into tissues and suppresses T cell activity via ApoE/LILRB4/SHP-2/uPAR/Arginase-1 signaling axis in acute myeloid leukemia (AML) cells. Blocking LILRB4 signaling using knockout and antagonistic antibody approaches impeded AML development. Thus, LILRB4 orchestrates tumor invasion pathways in monocytic leukemia cells by creating an immune-suppressive microenvironment. LILRB4 represents a compelling target for treatment of monocytic AML.
To identify novel mechanisms for AML development and immune regulation, we analyzed the relationship between gene expression of known co-stimulating and co-inhibitory receptors and the overall survival of AML patients as documented in the TCGA database. The expression of the mRNA encoding leukocyte immunoglobulin-like receptor B4 (LILRB4), an immune inhibitory receptor restrictively expressed on monocytic cells 2–4 and monocytic AML cells (FAB M4 and M5 AML subtypes) 5 ranked on the top of the list for negative correlation with AML patient survival (Fig. 1a and Extended Data Fig. 1a-d and Supplementary Table 1). Importantly, LILRB4 levels were higher on monocytic AML cells than on normal monocytes (Fig. 1b).
A previous study reported that the extracellular domain of LILRB4 inhibited T cell activities 6. To test whether LILRB4 expressed on AML cells has T cell-suppressive function, we co-cultured LILRB4-positive leukemia cells, LILRB4-negative leukemia cells, or normal hematopoietic cells with either autologous T cells or T cells from healthy donors. Only LILRB4-positive monocytic AML cells significantly suppressed T cell proliferation (Fig. 1c and Extended Data Fig. 1e-f). We then deleted lilrb4 from human monocytic AML THP-1 and MV4–11 cells and found that the T cell suppressive ability of AML cells was significantly reduced upon lilrb4 knockout (lilrb4-KO) and was restored by forced expression of wild-type lilrb4 (as lilrb4-KO-wt), but not by a mutant lilrb4 with deleted intracellular domain (as lilrb4-KO-intΔ) (Fig. 1d and Extended Data Fig. 2a-g). Moreover, when wild-type THP-1 cells and human T cells were cultured in separate transwells, LILRB4-mediated T cell inhibition was also observed and was able to be reversed by anti-LILRB4 blocking antibodies (Extended Data Fig. 2h-p). Blocking LILRB4 resulted in increase of cytotoxicity T cells and cytokine releasing (Extended Data Fig. 2q-u). These in vitro data suggest that, instead of the extracellular domain 6, the intracellular signaling of LILRB4 in AML cells is required for suppression of T cell activity.
Next, we used humanized mouse xenograft models and an immunocompetent mouse model to investigate LILRB4 function in immune checkpoint blockade. Subcutaneous implantation of THP-1 cells, but not the lilrb4-KO THP-1 cells, resulted in AML development in human T cell reconstituted mice, which was blocked by anti-LILRB4 treatment 7 (Extended Data Fig. 3a-i). Doxycycline-induced LILRB4 deletion in an established disseminated leukemia model in humanized mice also impaired leukemia development and restored T cells (Fig. 1e-f and Extended Data Fig. 3j-l). In addition, we subcutaneously implanted human LILRB4-expressing mouse C1498 AML cells (hlilrb4-C1498) into C57BL/6 mice to establish a syngeneic immunocompetent mouse model. To exclude the anti-tumor effects from Fc effector functions, we treated tumor-bearing mice with anti-LILRB4 with the Fc glycosylation site N297A mutation 8. LILRB4 blockade effectively lowered tumor burden and prolonged survival; depleting CD8+ T cells eliminated the anti-tumor effects of the anti-LILRB4 antibody (Extended Data Fig. 3m-r). These results suggest that the tumor-supportive effect of LILRB4 depends on host T cell inhibition. The anti-LILRB4 antibody treatment generated tumor-specific memory T cells (Extended Data Fig. 3s). Similar results were obtained in disseminated hlilrb4-C1498 syngeneic mouse model (Extended Data Fig. 3x-z). Finally, the blockade of LILRB4 significantly reduced leukemia development in primary human monocytic AML-derived xenografts (Fig. 1g-i and Extended Data Fig. 4a) and increased the numbers of engraftable autologous human T cells (Extended Data Fig. 4b). Together, our in vitro and in vivo results indicate that LILRB4 signaling in monocytic AML cells suppresses T cell-mediated anti-tumor immunity.
One of the characteristic features of monocytic AML is enhanced extramedullary infiltration of tumor cells 9. We observed that the antibody blockade of LILRB4 results in significant decrease of leukemic infiltration into internal organs, including bone marrow, liver, and brain (Extended Data Fig. 3t-v). Although anti-LILRB4 antibody treatment did not reduce the size of subcutaneous C1498 tumors in C57BL/6 mice depleted of CD8+ T cells (Extended Data Fig. 3m), treatment with anti-LILRB4 antibody did lead to decreased leukemia cell infiltration into liver (Extended Data Fig. 3w). We hypothesized that, in addition to T cell inhibition, LILRB4 promotes leukemia infiltration. To test this hypothesis, we performed trans-endothelial migration and homing assays and monitored leukemia infiltration relative to LILRB4 expression on leukemia cells. Human AML THP-1 cells depleted of LILRB4 had lower trans-endothelial migration in vitro than cells that expressed LILRB4 (Fig. 2a). Deletion of lilrb4 reduced homing and engraftment of AML cells to hematopoietic organs (Fig. 2b-c), and resulted in prolonged survival of xenografted mice (Fig. 2d) and delayed body weight loss (Fig. 2e). In contrast, forced expression of human LILRB4 in mouse AML C1498 or WEHI-3 cells had the opposite effects (Figs. 2f-j and Extended Data Fig. 5a-e). Antibody-mediated LILRB4 blockade showed the same effect as lilrb4 knockout in LILRB4-expressing AML cells (Extended Data Fig. 5f-t). This effect was depended on LILRB4 expression and its intracellular signaling in leukemia cells (Fig. 2k) but not the Fc effector functions of the antibody (Extended Data Fig. 5u-v). Furthermore, LILRB4 blockade reduced infiltration ability of primary monocytic AML cells (Fig. 2l-n and Extended Data Fig. 4c-e). Our results are concordant with previous studies showing that the frequency of circulating LILRB4+ AML blasts is significantly lower than that of the LILRB4- AML blasts 5 and that LILRB4+ chronic lymphocytic leukemia cells are associated with lymphoid tissue involvement 10. Bone marrow, liver, and brain to which LILRB4+ AML cells tend to migrate are known to have certain immune privileges 11–13. Thus, LILRB4-mediated migration, which supports enhanced extramedullary infiltration of monocytic AML cells, may also contribute to immune evasion.
The anti-LILRB4 antibody blockade of immune inhibitory and migration functions of AML cells suggests that those functions of LILRB4 are regulated by extracellular mechanisms. Integrin-αvβ3, was previously identified as the ligand for gp49B1, the mouse LILRB4 orthologue 14. However, a variety of integrin-αβ complexes did not activate human LILRB4 reporter cells (Extended Data Fig. 6a-b). Surprisingly, human serum and mouse serum were capable of activating the LILRB4 reporter but not reporters for other LILRBs (Fig. 3a). Through protein liquid chromatography fractionation followed by reporter assays and mass spectrometry, we identified APOE that specifically activated the reporters of LILRB4 and mouse PirB (Fig. 3b and Extended Data Fig. 6c-j). The serum from wild-type but not APOE-null mice activated the LILRB4 reporter (Fig. 3c). In addition, both liposome-reconstituted APOE protein (APOE-POPC) and lipid-free APOE activated LILRB4 reporter cells (Fig. 3d). The binding of APOE to THP-1 cells was significantly decreased by lilrb4-KO (Fig. 3e). The specific binding of recombinant APOE to LILRB4 was confirmed using microscale thermophoresis (MST), surface plasmon resonance (SPR), and bio-layer interferometry (Octet). The dissociation constant was 210 nM as determined by MST (Fig. 3f and Extended Data Fig. 6k-l). Mutagenesis studies showed that the N-terminal domain of APOE, and P35 and W106 in the first Ig-domain and Y121 in the linker region between two Ig-domains of LILRB4 are critical for APOE-mediated activation of LILRB4 (Fig. 3g-h and Supplementary Table 2 and Extended Data Fig. 6m).
The finding that APOE activates the immune inhibitory receptor LILRB4 is in agreement with the well-documented immune-suppressive function of APOE 15,16. To determine whether T cell suppressive activity of LILRB4 depends on APOE, we examined proliferation of T cells co-cultured with control or apoe-knockout human AML cells. AML cells deficient in APOE restored proliferation of T cells and suppressed migration of leukemia cells (Fig. 3i and Extended Data Fig. 6n-t). Moreover, the percentage of T cells in co-culture was significantly lower when the LILRB4-ectopically-expressing C1498 cells were treated with wild-type mouse serum compared to those treated with apoe-knockout mouse serum (Fig. 3j-k). Addition of liposome-reconstituted APOE to co-culture of mouse spleen cells and LILRB4-expressing AML cells decreased the T cell percentage (Fig. 3l). Furthermore, expression of LILRB4 significantly increased C1498 cells infiltrating to bone marrow and liver in wild-type mice but not in APOE-null recipients (Fig. 3m). These data indicate that APOE activates LILRB4 on human monocytic AML cells to suppress T cell proliferation and support AML cell migration.
We sought to identify the signaling downstream of LILRB4 required for T cell suppression and leukemia infiltration. Phosphatases SHP-1, SHP-2, and SHIP can be recruited to the intracellular domain of LILRB 2. The level of phosphorylation of SHP-2 but not of SHP-1 or SHIP was lower in lilrb4-KO AML cells than in wild-type cells (Fig. 4a and Extended Data Fig. 7a). Loss of SHP-2, but not loss of SHP-1 or SHIP, rescued T cell suppression by THP-1 cells (Fig. 4b and Extended Data Fig. 7b-c), and decreased short-term (20 hrs) and long-term (21 days) infiltration of THP-1 cells (Fig. 4c-d). Our results suggest that SHP-2 is a mediator of LILRB4 signaling.
Our Ingenuity Pathway Analysis showed that the activity of key transcription factors NFkB1 and RELA in the NF-κB pathway 17, which is positively regulated by SHP-2 18, was most significantly inhibited by loss of lilrb4 (Fig. 4e and Supplementary Tables 3 and 4). Consistently, the phosphorylation of IKKα/β and levels of nuclear NF-κB were decreased in lilrb4-KO AML cells (Fig. 4f-g and Extended Data Fig. 7a). Inhibition of NF-κB signaling restored T cell suppression and reduced AML cell infiltration in a LILRB4-dependent manner (Fig. 4h-i and Extended Data Fig. 7e-f). Therefore the effects of LILRB4 activation are mediated through the NF-κB pathway, which is particularly robust in monocytic AML among AML subtypes 19.
Consistent with our result that AML cells inhibit T cell proliferation in transwells (Extended Data Fig. 2o-p), the conditioned medium from wild-type THP-1 cells suppressed T cell activity but that medium from lilrb4-KO cells did not (Fig. 4j). Among proteins that were present higher in the conditioned medium of WT THP-1 cells than the lilrb4-KO counterparts (Extended Data Fig. 7g-i), uPAR is highly expressed by monocytic AML cells 20. uPAR, an NF-κB target, is well known to promote cancer invasion, metastasis, survival, and angiogenesis 21, 22. The addition of recombinant uPAR decreased proliferation of T cells co-cultured with lilrb4-KO THP-1 cells in a dose-dependent manner (Fig. 4k and Extended Data Fig. 7j). This activity of uPAR was likely mediated by downstream effectors in AML cells because uPAR does not effectively decrease T cell proliferation directly (Extended Data Fig. 7k).
The expression of Arginase-1 (ARG1), like uPAR, was significantly lower in lilrb4-KO AML cells than in wild-type cells (Extended Data Fig. 7l-m). ARG1 is up-regulated by uPAR-mediated signaling and inhibits T cell proliferation 23,24, and can be elevated by APOE 25 and NF-κB 26 for immune-suppressive functions. We hypothesized that ARG1 is a key downstream effector of LILRB4-NF-kB-uPAR signaling. ARG1 can be secreted by AML cells to inhibit T cell activity 27. Recombinant ARG1 decreased T cell proliferation in the co-culture with lilrb4-KO, apoe-KO, and shp-2-KO AML or primary AML cells (Fig. 4l and Extended Data Fig. 7n-p). Moreover, addition and overexpression of either uPAR or ARG1 rescued the migration ability of lilrb4-KO AML cells in vitro and in vivo, respectively (Extended Data Fig. 7q and Fig. 4m). Together, our results indicate that LILRB4/SHP-2/NF-κB/uPAR/ARG1 is a signaling pathway in monocytic AML cells (Extended Data Figs. 8–9) that suppresses immune activity and supports leukemia migration.
Because LILRB4 is restrictively expressed on normal monocytic cells 2 in which the LILRB4 signaling may differ from that in leukemia cells (Extended Data Fig. 9) and LILRB4 blockade did not significantly interfere with normal hematopoietic function (Extended Data Fig. 10), LILRB4 targeting may have minimal toxicity. Importantly, LILRB4 is also expressed on certain other types of cancers and myeloid-derived suppressor cells, tolerogenic dendritic cells, and tumor-associated macrophages 28,2,5,29,30. Targeting LILRB4 may thus enable combination of immunotherapy and targeted therapy in cancer treatment.
METHODS
Mice
C57 BL/6J and NOD-scid IL2Rγ null (NSG) mice were purchased from and maintained at the animal core facility of University of Texas Southwestern Medical Center (UTSW). Apoe-Knockout (apoe-KO, Apoetm1Unc) mice as previously described 31 were purchased from the Jackson Laboratory. Animal work described in this manuscript has been approved and conducted under the oversight of the UT Southwestern Institutional Animal Care and Use Committee (IACUC). For each experiment, the same sex and age-matched (4–8 weeks) mice were used and randomly allocated to each group; and For tumor size measurement and in vivo lumina imaging experiments, treatment conditions of the mice were blinded. The minimum number of mice in each group was calculated based on results from our prior relevant studies 32–36. For the subcutaneous tumor model, the tumor size was calculated by (width x width x length) cm3. The maximal tumor measurement permitted by UTSW IACUC is 2 cm in diameter of tumor. In none of the experiments were these limits exceeded (see Source Data Extended Data Figure 3). We have complied with all relevant ethical regulations with approved animal study protocols.
Cell culture
293T cells were cultured in Dulbecco’s modified Eagle’s medium (DMEM) supplemented with 10% fetal bovine serum (FBS) at 37 °C in 5% CO2 and the normal level of O2. Human umbilical vein/vascular endothelium cells (HUVECs) (ATCC, CRL-1730) were cultured in endothelial cell growth medium plus growth factor, cytokines and supplements (EGM-BulletKit, Lonza) at 37 °C in 5% CO2 and the normal level of O2. Human monocytic AML cells, THP-1 (ATCC, TIB-202), MV4–11 (ATCC, CRL-9591), and U937 (ATCC, CRL-1593.2), and mouse AML cells, WEHI-3 (ATCC, TIB-68), were cultured in Roswell Park Memorial Institute (RPMI) 1640 supplemented with 10% FBS at 37 °C in 5% CO2 and the normal level of O2. Mouse AML cells, C1498 (ATCC, TIB-49) were cultured in DMEM supplemented with 10% FBS at 37 °C in 5% CO2 and the normal level of O2. All cell lines were routinely tested using a mycoplasma-contamination kit (R&D Systems).
Primary human leukemia cells
Primary human AML and B-ALL samples were obtained from the tissue banks at UTSW and University of Texas MD Anderson Cancer Center (MDACC). Informed consent was obtained under protocols reviewed and approved by the Institutional Review Board at UTSW and MDACC (IRB STU 122013–023 by UTSW and LAB10–0682 by MDACC). The UTSW cohort included 105 AML patients representative of AML subtypes by the French-American-British (FAB) classification, acute myeloblastic leukemia with minimal maturation (M1, n=9), acute myeloblastic leukemia with maturation (M2, n=34), acute promyelocytic leukemia (M3, n=10), acute myelomonocytic leukemia (M4, n=34), acute monocytic leukemia (M5, n=25), acute erythroid leukemia (M6, n=2), and acute megakaryoblastic leukemia (M7, n=1) and patients with undifferentiated leukemia (AUL; n=1) and transient myeloproliferative disorder (TAM; n=2) (Supplementary Table 1). Samples were frozen in FBS with 10% DMSO and stored in liquid nitrogen. Primary leukemia samples used in PDX, co-culture, short-term infiltration assay and western blotting were summarized in Supplementary Table 5. We have complied with all relevant ethical regulations with approved study protocols.
Human normal monocytes and macrophages
Human normal monocytes (CD14+ cells) were isolated by the AutoMACS Pro Separation System (Miltenyi Biotech, Auburn, CA) from the mononuclear cells fraction of normal peripheral blood. Briefly, buffy coat was purchased from Interstate Blood Bank (Memphis, TN) and the mononuclear cell layer was separated by Ficoll Hypaque (17144003, GE Lifesciences) density gradient separation. Mononuclear cells were treated with red blood cell lysis buffer to remove red blood cells and then incubated with CD14 microbead-conjugated antibody (130–050-201, Miltenyi Biotech, Auburn, CA) for 15 min at 4°C. CD14 positive cells were then isolated using the positive selection program according to the manufacturer’s protocol. One million CD14+ cells were plated in macrophage culture media, Iscove’s modified Dulbecco’s medium (IMDM) (12440053, Thermo fisher) supplemented with 10% human AB serum (MT35060CI, Fisher Scientific), 1% NEAA (11–140-050, Fisher), 2 μM L-alanine-L-glutamine (SH3003402, Fisher), per each well of a 6-well plate and cultured for 7 days. After incubation, most of the cells were adherent to the plastic surface and stained positive for CD14 and other markers specific for macrophages.
TCGA analyses
Data were obtained from the TCGA acute myeloid leukemia database (version: August 16, 2016). The patients were classified into AML subtypes (FAB classification) M0 (undifferentiated acute myeloblastic leukemia) (n=16), M1 (n=42), M2 (n=39), M3 (n=16), M4 (n=35), M5 (n=18), M6 (n=2), M7 (n=3); two cases were not classified by subtype. The mRNA levels of indicated genes were determined by RNA-seq (polyA+ IlluminaHiSeq). RESM-normalized counts are reported, and data were analyzed and visualized with UCSC Xena (xena.ucsc.edu). For analysis of overall survival, 160 patients with available survival data were separated into three groups based on whether they had high, moderate, or low gene expression and then analyzed by Xena Kaplan Meier plot (http://xena.ucsc.edu/survival-plots/).
Flow cytometry
Primary antibodies including anti-human CD45-PE (BD Pharmingen, HI30, 1:100), CD45-FITC (BD Pharmingen, HI30, 1:100), CD45-APC (BD Pharmingen, HI30, 1:100), anti-human CD34-FITC (BD Pharmingen, 55582, 1:100), anti-human CD19-PE (eBioscience, HIB19, 1:100), anti-human CD20-PE (BD Pharmingen, 555623, 1:100), anti-human CD11b-APC (eBioscience, ICRF44, 1:100), anti-human LILRB4-APC (eBioscience, ZM4.1, 1:100), anti-human LILRB4-PE (Biolegend, ZM4.1, 1:100), anti-human CD14-APC (eBioscience, 61D3, 1:100), anti-human CD33-APC (Biolegend, P67.6, 1:100), anti-human CD4-APC (eBioscience, RPA-T4, 1:100), anti-human CD3-FITC (BioLegend, HIT3a, 1:100), anti-human CD3-Pacific blue (BD Pharmingen, SP34–2, 1:100) anti-human CD8-PE (BD Pharmingen, 555367, 1:100), anti-human CD28-APC (eBioscience, CD28.2, 1:100), anti-human CD40L-APC (eBioscience, 24–31, 1:100), anti-human PD1-APC (Biolegend, EH12.2H7, 1:100), anti-human TIM3-APC (eBioscience, F38–2E2, 1:100), anti-human TIGIT-APC (eBioscience, MBSA43, 1:100), anti-human LAG3-APC (eBioscience, 3DS223H, 1:100), anti-human FasL-PE (eBioscience, 24–31, 1:100), anti-uPAR-APC (Biolegend, VIM5, 1:100), anti-mouse CD3-APC (BioLegend, 17A2, 1:200), anti-mouse CD8a-PE (BioLegend, 53–6.7, 1:200), anti-mouse CD45-PE (BD Pharmingen, 30-F11, 1:200), anti-mouse CD49b-APC (eBioscience, DX5, 1:200), anti-mouse CD49f-PE (eBioscience, GoH3, 1:200), anti-mouse CD11b-APC (BioLegend, M1/71, 1:200), anti-mouse CD11b-PE (BioLegend, M1/71, 1:200), anti-mouse CD11c-APC (eBioscience, N418, 1:200), anti-mouse F4/80-APC (BioLegend, BM8, 1:200), anti-His-tag-APC (R&D systems, AD1.1.10, 1:400), and IgG isotype-control-APC (eBioscience, P3.6.2.8.1, 1:400) antibodies were used. Cells were run on either Calibur for analysis or FACSAria for analysis and sorting. Flow data were analysed by Flowjo software. For analysis of human hematopoietic engraftment in NSG mice, a previously published protocol was followed 33,35,37. PI staining was used to exclude dead cells in analysis and sorting. For intracellular staining, we followed the two-step protocol for fixation/methanol from eBioscience. Briefly, human primary AML cells were stained for the surface expression of LILRB4 (anti-LILRB4-Alexa Fluor 647, Biolegend, ZM4.1, 1:100) and CD33 (anti-human CD33-FITC, Biolegend, HIM3–4, 1:100) and fixable cell viability dye eFluor 450 (Bioscience, Cat#65–0863-14, 1:100) followed by fixation (IC fixation buffer, eBioscience, Cat#00–8222) and methanol treatment. After that, cells were stained for intracellular antigens by anti-p-SHP-2 (Y580)-PE (Cell signaling, Cat#13328S, 1:100), anti-pIKKα/β (S176/180) (16A6) (Cell signaling, Cat#2697, 1:100), anti-NFκB (S529)-PE (eBioscience, B33B4WP, 1:100), anti-uPAR-PE (Biolegend, VIM5, 1:100), anti-Arginase-1 (D4E3M) (Cell signaling, Cat#93668, 1:100), rabbit IgG Isotype control-PE (Cell signaling, Cat#5742, 1:100), mouse IgG Isotype control-PE (eBioscience, m2a-15F8, 1:100) and anti-rabbit IgG-PE (Jackson Immunoresearch Lab, Cat#111–116-144, 1:400) for flow cytometry analysis.
Virus construction and infection
For retrovirus packaging, plasmid constructs XZ201-IRES-GFP and XZ201-human lilrb4 (hlilrb4)-IRES-GFP were mixed with PCL-ECO (2:1), followed by transfection into 293T cells using Lipofectamine 2000 (Invitrogen). For lentivirus packaging, CRISPER/Cas-9 based gRNA (guide RNA) constructs and other constructs for gene overexpression including – pLentiLox3.7-luciferase-IRES-GFP, ZsGreen-hlilrb4 and ZsGreen-hlilrb4-intΔ, pLVX-plaur-IRES-tdTomato, pLVX-arg1-IRES-tdTomato were mixed with psPAX2 and pMD2.G (Addgene) at a ratio of 4:3:1 and transfected into 293T cells using Lipofectamine 2000 (Invitrogen). Virus-containing supernatant was collected 48–72 hrs post-transfection and used for infection as described previously 38.
CRISPR/Cas9-based gene knockout in AML cells
Human AML cells were infected with doxycycline-inducible Cas9-expressing lentivirus (pCW-Cas9, Addgene 50661). After 1 μg/ml puromycin selection, the survived cells were infected with sgRNA-expressing lentivirus, produced by the plasmid modified from pSLQ1651 (Addgene 51024) by replacing the puro-mcherry with GFP for sorting. Scramble control sgRNA (sgRNA 5’- GAACGACTAGTTAGGCGTGTA −3’), lilrb4 targeting sgRNA (sgRNA1 5’- TGTTACTATCGCAGCCCTGT −3’; sgRNA2 5’- GTAGGTCCCCCCGTGCACTG −3’; sgRNA3 5’-CCTGTGACCTCAGTGCACGG −3’), apoe targeting sgRNA (sgRNA1 5’- CTTTTGGGATTACCTGCGC −3’; sgRNA2 5’- AACTGGCACTGGGTCGCTTT −3’), shp-1 targeting sgRNA (sgRNA1 5’- TAAGACCTACATCGCCAGCC −3’; sgRNA2 5’- GAAGAACTTGCACCAGCGTC −3’), shp-2 targeting sgRNA (sgRNA1 5’- GAGACTTCACACTTTCCGTT −3’; sgRNA2 5’- TACAGTACTACAACTCAAGC −3’), ship targeting sgRNA (sgRNA1 5’- CACGCAGAGCGCGTATGCCC −3’; sgRNA2 5’- TGGCAACATCACCCGCTCCA −3’) which were designed by an online tool (http://crispr.mit.edu), were cloned into the sgRNA plasmid, individually. After treated with 1 μg/ml doxycycline (Sigma, Cat#PHR1789) for 1 week, these cells were staining with anti-LILRB4 antibody and the LILRB4 negative cells were sorted as lilrb4-knockout cells. For apoe-, shp-1-, shp-2- and ship-knockout cells, GFP+ cells were sorted into a 96-well plate as single cell per well. After cell expanded, knockout cells were verified by western blotting. For in vivo induction of CRISPR/Cas9 to achieve gene knockout, we fed mice with doxycycline as described 39. Briefly, 7 days after Cas9/lilrb4-sgRNA-transfected THP-1 cell implantation, mice were treat with 2 mg/mouse of doxycycline via gavage daily for 5 days to achieve Cas9 expression in engrafted leukemia cells. The knockout was validated by flow cytometry.
Leukemia cell and T cell co-culture assay
In the co-culture assay, human T cells (5 × 104 per well) isolated from health donor peripheral blood (PB009–1-0, Allcells) were mixed with irradiated (28 Gy) indicated human leukemia cells in a U-bottom 96 well-plate. For non-contact co-culture of T cells with leukemia cells, leukemia cells were cultured in the upper chamber of transwell inserts (pore size, 3 μM, #09–761-80, Thermo Fisher) in U-bottom 96 well-plate. T cells isolated from healthy donors were placed in the lower chambers of a 96-well transwell plate. Irradiated indicated leukemia cells (E:T ratio = 2:1 if not indicated) were added to the upper chambers and treated with indicated antibodies, proteins and reagents. After culture with anti-CD3/CD28-coated beads (11161D, Thermo Fisher) and 50 U/ml rhIL-2 for 5~7 days, representative cells were photographed using an inverted microscope, and T cells were stained with anti-CD3 antibodies and analyzed by flow cytometry.
For primary AML or B-ALL samples, patient leukemia cells were sorted as CD33+ and CD19+ for AML and B-ALL, respectively. These leukemia cells were cultured with autologous CD3+ T cells from the same patient or allogeneic T cells from health donor (E:T ratio = 2:1). After culture with anti-CD3/CD28-coated beads (11161D, Thermo Fisher) and 50 U/ml rhIL-2 for 14 days, representative cells were photographed using an inverted microscope, and T cells were stained with anti-CD3, anti-CD4 and anti-CD8 antibodies and analyzed by flow cytometry.
For cytotoxicity assay, human CD8+ T cells (5 × 104 per well) isolated from PBMCs of a healthy donor were stimulated with anti-CD3/CD28/CD137-coated beads (11163D, Thermo Fisher) for 2 days in a 96-well plate. Then, indicated 5 × 103 leukemia cells and 50 to 500 μg/ml anti-LILRB4 antibodies or control IgG were added. Cell numbers were determined on day 7 in triplicate wells. Or indicated leukemia cells in indicated E:T ratios were cultured with T cells for 4~6 hrs in triplicate wells. Anti-CD3 and anti-CD8 were used to detect human CTL cells; indicated live THP-1 cells were positive for GFP and negative for PI. Cell supernatants from co-cultures of stimulated CTL cells and THP-1 cells treated with anti-LILRB4 or IgG were used to examine cytokine production using human cytokine arrays (AAH-CYT-6, RayBiotech).
For mouse leukemia/T cells co-culture, spleen cells from wild-type C57bl/6 were co-cultured with 2.5×104 irradiated (28 Gy) mouse leukemia C1498 cells in a U-bottom 96 well-plate for 60 hrs. Anti-CD3/CD28-coated beads (11452D, Thermo Fisher), 50 U/ml recombinant human IL-2, and 5% serum from wild-type C57bl/6 mice or that from apoe-KO mice were added to the medium. In some experiments, 50 μg/ml lipid-bound APOE proteins (APOE-POPC) were added to the medium. The lipidation of APOE recombinant protein was conducted as described 40.
Transendothelial migration assays
To measure the ability of AML cells to migrate through endothelial cells, 3 × 105 HUVEC cells were cultured on the transwell membrane (pore size is 8 μm). After 3 days, 1 × 105 indicated leukemia cells were seeded in the upper chamber. In indicated experiments, leukemia cell were treated with antibodies or proteins in the upper chamber. After 18 h, cells in lower chamber were counted.
Short-term infiltration assay of leukemia cells and homing assay of hematopoietic stem/progenitor cells (HSPCs)
Cells (5 × 106 cells per mouse) were injected intravenously into NSG mice. Animals were treated with 10 mg/kg of anti-LILRB4 antibodies or control IgG immediately after injection of leukemia cells. Mice were sacrificed after 20 hrs. Peripheral blood, bone marrow, liver, and spleen were harvested, and single-cell suspensions were examined by flow cytometry. CFSE, GFP or indicated markers such as anti-human CD45 and anti-human CD33 was used to detect target leukemia cells in indicated experiments. Numbers of leukemia cells in recipient liver, spleen, and bone marrow are reported as a ratio relative to cell numbers in peripheral blood.
To test the infiltration ability of mouse leukemia cells, 5 × 106 C1498-GFP-hLILRB4 cells or C1498-GFP were injected intravenously into wild-type C57BL/6J or APOE-null mice. Mice were sacrificed after 20 h. GFP was used to detect leukemia cells by flow cytometry. The number of leukemia cells in recipient liver, spleen, and bone marrow were normalized to numbers in peripheral blood and are reported as a ratio.
To test HSPCs homing ability, 1 × 107 human cord blood mononuclear cells were injected intravenously into an NSG mouse. Mice were treated with 10 mg/kg of anti-LILRB4 antibodies or control IgG immediately after injection of mononuclear cells and were sacrificed after 20 hrs. Anti-human CD45 and anti-human CD34 were used to detect human HSPCs by flow cytometry. Similarly, to test the infiltration ability of normal human monocytes, 5 × 106 CD14-positive selected monocyte from health donor PBMC were labeled by CFSE and injected intravenously into an NSG mouse. Mice were treated with 10 mg/kg of anti-LILRB4 antibodies or control IgG immediately after injection of monocytes and were sacrificed after 20 hrs. CFSE-positive cells were analyzed by flow cytometry.
Innate immune cell depletion
NK cell depletion was done by i.p. injection of 50 μl anti-asialo GM1 antibodies (CL8955, Cedarlane) 3 days before leukemia cell implantation, which resulted in >90% depletion of CD45+CD49b+ NK cells in the circulation of NSG mice. Macrophages were depleted by treating NSG mice with clodronate (dichloromethylene bisphosphonate) liposomes (SKU8909, Clodrosome) (200 μl of stock solution 3 days before leukemia cell implantation), resulting in >70% depletion of CD45+CD11b+F4/80+ macrophages in the circulation of NSG mice. NSG mice were rendered neutropenic by i.p. injection of 200μg anti-Ly-6G mAb (BP0075–1, Bioxcell) on days −3, −2, −1, and 0 post leukemia cell implantation, resulting in >80% depletion of CD45.1+CD11b+CD11c- neutrophils in the circulation of NSG mice.
Human AML xenograft
Xenografts were performed essentially as described 2,3,6,7. Briefly, 6–8 week-old NSG mice were used for transplantation. 1 × 106 human leukemia cells were resuspended in 200 μl PBS for each mouse i.v. injection. Mice were immediately given 10 mg/kg of anti-LILRB4 antibodies or control IgG intravenously. Three to four weeks after transplantation, the peripheral blood, bone marrow, spleen, and liver were assessed for the engraftment. Leukemia growth was monitored over time by luminescence imaging (Max, 3×108 p/sec/cm2/sr; Min, 5×106 p/sec/cm2/sr). For survival curve experiments, the death of mice was recorded when the moribund animals were euthanized. For primary patient-derived xenograft (PDX), each NSG mouse was given 5 to 10 × 106 human primary peripheral blood or bone marrow mononuclear cells, which contain leukemia cells and other normal compartments such as normal hematopoietic stem progenitor cells and autologous T cells, via tail-vein injection. Mice were immediately given 10 mg/kg of anti-LILRB4 antibodies or control IgG intravenously and were treated twice a week until euthanization. For AML#11, mice were given 10 mg/kg of anti-LILRB4 antibodies or control IgG intravenously 7 days after leukemia cell implantation and were treated twice a week until euthanization. Leukemia growth was monitored over time by flow cytometry of human cells in peripheral blood. More than 1% of human leukemia cells in mouse tissue were considered successful engraftment of primary AML cells. One to four months after transplantation, the peripheral blood, bone marrow, spleen, and liver were assessed for the engraftment.
For the hPBMC-humanized model, 1 × 107 human PBMCs were injected intravenously into each NSG mouse. Three weeks after implantation, mice had 30 to 50% engraftment of human T cells. At 3 weeks post implantation, 1 × 106 human AML THP-1 cells, including wild-type, lilrb4-KO THP-1 cells or THP-1 cells stably express luciferase (THP-1-Luc-GFP cells), were subcutaneously implanted. Mice were immediately given 10 mg/kg of anti-LILRB4 antibodies or control IgG intravenously and were treated twice a week until euthanization. Tumor growth was monitored over time by luminescence imaging (Max, 1×108 p/sec/cm2/sr; Min, 5×106 p/sec/cm2/sr). Tumor sizes were determined by caliper measure (width x width x length). For inducible lilrb4-knockout experiment, 1 × 106 Cas9/lilrb4-sgRNA-transfected THP-1 cells were injected in each NSG mouse by i.v., immediately followed by i.v. injection of 0.5 × 106 isolated human normal T cells from health donors. 7 days after THP-1 and T cell implantation, mice were treat with 2mg/mouse of doxycycline via gavage daily for 5 days to achieve Cas9 expression in engrafted THP-1 cells. At 3 weeks post implantation, the peripheral blood, bone marrow, spleen, and liver were assessed for the engraftment.
For the human Cord blood (hCB)-xenograft model, 2 × 104 human CD34+ hCB cells were injected intravenously into each NSG mouse. Six weeks after implantation, mice had 10 to 50% engraftment of human cells. 1 × 106 THP-1 cells that stably express luciferase (THP-1-Luc-GFP cells) were intravenously implanted. Mice were immediately given 10 mg/kg of anti-LILRB4 antibodies or control IgG intravenously. Tumor growth was monitored over time by luminescence imaging (Max, 1×108 p/sec/cm2/sr; Min, 5×106 p/sec/cm2/sr). Lineages of human normal blood cells were analyzed by flow cytometry.
Mouse AML allograft
The procedure of mouse AML allograft was similar to that of human AML xenograft. Briefly, 6–8 week-old wild-type C57bl/6 mice were used for transplantation. 1 × 106 mouse leukemia cells expressing human LILRB4 were resuspended in 200 μl PBS for each mouse intravenously or subcutaneously implantation. Mice were given 10 mg/kg of anti-LILRB4-N297A antibodies or control IgG intravenously 7 days after leukemia cell implantation and were treated twice a week until euthanization. Three weeks after transplantation, the peripheral blood, bone marrow, spleen, and liver were assessed for the engraftment. For subcutaneously implant mice, tumor sizes were determined by caliper measure (width x width x length). For survival curve experiments, the death of mice was recorded when the moribund animals were euthanized. For CD8+ T depletion, 10mg/kg anti-CD8 antibodies (YTS 169.4.2, Bioxcell) were i.v. injected 3 days after leukemia cell implantation and were treated for additional two time every 3 days. To determine whether anti-LILRB4 antibody treatment generates tumor-specific memory T cells against the tumor or against LILRB4, we conducted adoptive transfer of spleen cells (5 × 106 /mouse) from anti-LILRB4 treated mice into normal recipient C57bl/6 mice. Four out of five transplanted mice rejected the control C1498-GFP mouse leukemia cells, and these mice were not susceptible to rechallenge with 3-fold higher numbers (3 × 106 /mouse) of C1498-GFP leukemia cells. While none of 5 mice with adoptive transfer of spleen cells from naïve mice reject the control C1498-GFP mouse leukemia cells.
Chimeric receptor reporter assay
We constructed a stable chimeric receptor reporter cell system as described 3,4 to test the ability of a ligand to bind to the ECD of individual LILRBs, PirB, gp49B1 and LILRB4 site mutants and to trigger the activation or inhibition of the chimerically fused intracellular domain of paired immunoglobulin-like receptor β, which signals through the adaptor DAP-12 to activate the NFAT promoter. If an agonist or antagonist binds the ECD and activates or suppresses the chimeric signaling domain, an increase or decrease, respectively, in GFP expression is observed. A competition assay was used to screen LILRB4 blocking antibodies. Briefly, APOE proteins (CI02, Novoprotein; 10 μg/ml) or human AB serum (10%, diluted in PBS) were pre-coated on 96-well plate at 37 °C for 3 hrs. After two washes with PBS, 2×104 LILRB4 reporter cells were seeded in each well; meanwhile, indicated anti-LILRB4 antibodies were added into culture media. After 16 hrs, the percentage of GFP+ reporter cells was analyzed by flow cytometry. The threshold of activation is 2 times of negative control treatment.
Fast protein liquid chromatography (FPLC) and Mass Spectrum
10% human AB serum in PBS was loaded onto a 16/60 Superdex 200 gel filtration column and eluted with PBS and 2mM EDTA. Eighty Fractions (40 ml) were collected, and each fraction (0.5 ml) was analyzed by chimeric receptor reporter assay. The active fractions (#26~30) were loaded onto PAGE-gel and processed to LC-MS/MS analysis (Orbitrap Elite) for protein identification in UTSW proteomics core. Recombinant or purified proteins used for validation were ZA2G (MBS145455, MyBioSource), AMBP (13141-H08H1, Sino Biological Inc), TTHY (12091-H08H, Sino Biological Inc), PEDF (11104-H08H, Sino Biological Inc), A2MG (MBS173010, MyBioSource), HEMO (MBS143111, MyBioSource), ANGT (MBS173525, MyBioSource), A1AT (MBS173006, MyBioSource), S100A9 (pro-814, Prospecbio), HORN (EBP08267, Biotrend USA), VTDB (CSB-EP009306HU, Biotrend USA), LRG1 (pro-141, Prospecbio), A1BG (RPE570Hu01, Cloud-Clone Corp), CRSP3 (RD172262100, BioVendor), APOA1 (16–16-120101-LEL, Athens Research & Technology), APOA2 (16–16-120102, Athens Research & Technology), APOA4 (16–16-120104, Athens Research & Technology), APOB (16–16-120200, Athens Research & Technology), APOC1 (16–16-120301, Athens Research & Technology), APOC2 (16–16-120302, Athens Research & Technology), APOC3 (16–16-120303, Athens Research & Technology), hAPOE (16–16-120500, Athens Research & Technology), mAPOE (CJ05, Novoprotein), APOE2 (350–12, Peprotech), APOE3 (350–02, Peprotech), APOE4 (350–04, Peprotech), PODXL2 (1524-EG-050, R&D systems), CD44 (12211-H08H, Sino Biological Inc), HCK (PV6128, Thermo Fisher), VEGFR3 (10806-H08H, Sino Biological Inc), NRG3 (16071-H08H, Sino Biological Inc), PI16 (H00221476-P01, Novusbio), hMAG (8940-MG-050, R&D systems), mMAG (8580-MG-100, R&D systems), CNTF (303-CR-050, R&D systems), ANGPTL-7 (914-AN-025/CF, R&D systems), integrin-α1β1 (7064-AB-025, R&D systems), integrin-α2β1 (5698-AB-050, R&D systems), integrin-α2β3 (7148-AB-025, R&D systems), integrin-α3β1 (2840-A3–050, R&D systems), integrin-α4β1 (5668-A4–050, R&D systems), integrin-α4β7 (5397-A3–050, R&D systems), integrin-α5β1 (3230-A5–050, R&D systems), integrin-α5β3 (3050-AV-050, R&D systems), integrin-α5β5 (2528-AV-050, R&D systems), integrin-α5β6 (CT039-H2508H, Sino Biological Inc), mintegrin-α5β6 (CT051-M2508H, Sino Biological Inc), integrin-α5β8 (4135-AV-050, R&D systems), integrin-α6β4 (5497-A6–050, R&D systems), integrin-α8β1 (CT016-H2508H, Sino Biological Inc), integrin-α9β1 (5438-A9–050, R&D systems), integrin-α10β1 (5895-AB-050, R&D systems), integrin-α11β1 (6357-AB-050, R&D systems), integrin-αEβ7 (5850-A3–050, R&D systems), integrin-αXβ2 (CT017-H2508H, Sino Biological Inc) and normal mouse serum (NS03L, Millipore sigma).
Bio-layer interferometry
Binding interaction analyses between LILRB4-Fc with APOE2, APOE3, and APOE4 were performed on the Octet RED96 (ForteBio, Pall Corporation). All interaction studies were performed with the protein A dip-and-read biosensors (ForteBio). All binding experiments were performed using the Octet Red and kinetics buffer at 30 °C. LILRB4-Fc coated biosensors (25 μg/ml LILRB4-Fc was loaded for 420 s) were washed in kinetics buffer before monitoring of association (300 s) and dissociation (600 s) of APOEs. Background wavelength shifts were measured from reference sensors that were loaded only with LILRB4-Fc.
Surface plasmon resonance (SPR)
Biacore 2000 and CM5 chips were used to analyze binding of recombinant APOEs to the LILRB4 extracellular domain fused to hFc, using a method as we described 2. Recombinant protein A (Pierce) was pre-immobilized in two flow cells using the amine-coupling kit from GE. LILRB4-hFc was injected into one of the flow cells to be captured by the protein A. Each binding sensorgram from the sample flow cell, containing a captured LILRB4-hFc, was corrected for the protein A coupled cell control. Following each injection of an antigen solution, which induced the binding reaction, and the dissociation period during which the running buffer was infused, the protein A surface was regenerated by the injection of the regeneration solution containing 10 mM Na3PO4 (pH 2.5) and 500 mM NaCl. All captured LILRB4-hFc, with and without APOE bound, was completely removed, and another cycle begun. All measurements were performed at 25°C with a flow rate of 30 μL/min.
Microscale thermophoresis (MST)
MST experiments were performed on a Monolith NT.115 system (NanoTemper Technologies) using 80% LED and 20% IR-laser power. Laser on and off times were set at 30 s and 5 s, respectively. Recombinant LILRB4-ECD protein (SinoBio) was labeled with 4488-NHS (NanoTemper Technologies) and applied at a final concentration of 5.9 nM. A two-fold dilution series was prepared for unlabeled His-APOE (CI06, Novoprotein) in PBS, and each dilution point was similarly transferred to LILRB4-ECD solution. The final concentrations of His-APOE ranged from 0.36 nM to 12 μM. Samples were filled into standard-treated capillaries (NanoTemper Technologies) for measurement.
Western blotting and co-immunoprecipitation
Whole cells were lysed in Laemmli sample buffer (Sigma-Aldrich) supplemented with protease inhibitor cocktail (Roche Diagnostics). Samples were separated on SDS–PAGE gels (Bio-Rad) and transferred on nitrocellulose membranes (Bio-Rad) for protein detection. Primary antibodies including Anti-SHP-1 (Cell signaling, 3759, 1:1000), anti-phospho-SHP-1 Tyr564 (Cell signaling, 8849, 1:500), anti-phospho-SHP-1 Tyr564 (Invitrogen, PA537708, 1:500), anti-SHP-2 (Cell signaling, 3397, 1:1000), anti-phospho-SHP-2 Tyr580 (Cell signaling, 3703, 1:500), anti-SHIP1 (Cell signaling, 2727, 1:1000), anti-phospho-SHIP1 Tyr1020 (Cell signaling, 3941, 1:500), anti-NFκB p65 (Cell signaling, 8242, 1:1000), anti-IKKα (Cell signaling, 11930, 1:1000), anti-IKKβ (Cell signaling, 8943, 1:1000), anti-phospho-IKKα/β Ser176/180 (Cell signaling, 2697, 1:500), anti-IκBα (Cell signaling, 4814, 1:1000), anti-phospho-IκBα Ser32 (Cell signaling, 2859, 1:500), anti-Lamin-B2 (Cell signaling, 12255, 1:1000) and anti-Arginase-1 (Cell signaling, 9819, 1:1000), anti-uPAR (Invitrogen, MON R-4–02, 1:500), anti-LILRB4 (Santa cruz, sc-366213, 1:200), anti-APOE (Creative diagnostics, DCABH-2367, 1:250), anti-β-actin (Sigma-Aldrich, A2066, 1:1000) and anti-α-tubulin (Sigma-Aldrich, MABT205, 1:1000), as well as horseradish peroxidase (HRP) conjugated secondary antibodies (Cell signaling, 7074, 1:1,000, and 7076, 1:1,000) and chemi-luminescent substrate (Invitrogen), were used. Specific cellular compartment fractionations were carried out using the NE-PER nuclear/cytoplasmic extraction kit (Thermo fisher, 78833) or the plasma membrane protein extraction kit (Abcam, ab65400). Proteins from plasma membrane fraction were further incubated with anti-LILRB4 antibodies and dynabeads protein A (Thermo fisher, 10001D) for further immunoprecipitation and western blotting.
Immunohistochemistry
Hematoxylin staining and immunostaining were performed on paraffin sections of tumors. Antibodies used were against LILRB4 (lab produced, 1:100), CD3 (Abcam, ab16669, 1:100), PD-1 (Thermo Fisher, J116, 14–9989-82, 1:100) and Arginase-1 (Cell signaling, 9819S, 1:100). The images were visualized using the Hamamatsu NanoZoomer 2.0-HT (Meyer instruments Inc., Houston, TX) and viewed in NPDview2 software (Hamamatsu, Japan).
Cytokine antibody array and arginase activity assay
To examine the secreted protein from leukemia cells, condition media were applied to a human cytokine antibody array (AAH-CYT-1000, RayBio) for the semi-quantitative detection of 120 human proteins. Image J (NIH) was used for quantification. Arginase activity was determined in condition media of indicated leukemia cells by a QuantiChrom Arginase assay kit (DARG-100, BioAssay system).
RNA-seq analysis
RNA was purified from sorted cells with Qiagen RNeasy Mini Kit and then reverse-transcribed with SuperScript III Reverse Transcriptase (Invitrogen) according to the manufacturer’s instructions. RNA-seq was performed at the UTSW Genomics and Microarray Core Facility. The cDNA was sonicated using a Covaris S2 ultrasonicator, and libraries were prepared with the KAPA High Throughput Library Preparation Kit. Samples were end-repaired, and the 3′ ends were adenylated and barcoded with multiplex adapters. PCR-amplified libraries were purified with AmpureXP beads and validated on the Agilent 2100 Bioanalyzer. Before being normalized and pooled, samples were quantified by Qubit (Invitrogen) and then run on an Illumina Hiseq 2500 instrument using PE100 SBS v3 reagents to generate 51-bp single-end reads. Before mapping, reads were trimmed to remove low-quality regions in the ends. Trimmed reads were mapped to the human genome (HM19) using TopHat v2.0.1227 with the UCSC iGenomes GTF file from Illumina.
Methods for data normalization and analysis are based on the use of “internal standards” that characterize some aspects of the system’s behavior, such as technical variability, as presented elsewhere. Genes with log2 (fold change) > 2, P < 0.01 and RPKM > 0.1 were deemed to be significantly differentially expressed between the two conditions and were used for pathway analysis and upstream transcription factor analysis. Pathway analysis was conducted using the DAVID (https://david.ncifcrf.gov/tools.jsp). Upstream transcription-factor analysis was conducted using QIAGEN’s Ingenuity tool (http://www.ingenuity.com/).
Molecular docking of LILRB4 with APOE
Docking of LILRB4 with APOE was performed on ZDOCKpro module of the Insight II package. The general protocol for running ZDOCK includes two consecutive steps of calculation described as geometry search and energy search, running in program ZDOCK and RDOCK, respectively. LILRB4 crystal structure (3P2T) and APOE3 structure (2L7B) were obtained from PDB database. Top 50 ZDOCK poses were submitted to RDOCK refinement. Poses with high score both in ZDOCK and RDOCK were selected as candidate complex for LILRB4/APOE interaction analysis (Supplementary Table 2).
Statistical analyses
Representative data from four independent experiments or indicated independent samples are presented as dot plots (means ± s.e.m.) or as box-and-whisker plots (median values (line), 25th–75th percentiles (box outline) and minimum and maximum values (whiskers)). Statistical significance for two samples-comparison was calculated by two-tailed Student’s t-test. Statistical significance for survival was calculated by the log–rank test. The multivariate analysis of TCGA data was analyzed by Cox regression. The difference was considered statistically significant if p < 0.05. n.s., not significant; p values are represented as precise values. The Pearson’s correlation analyses were performed with the RStudio software (the R Foundation).
Code availability
The custom code for Pearson’s correlation analysis in RStudio is showed below.
setwd(“~/file paths”) draw.graph = function(sam) { file = paste(sam, “.tsv”, sep =““) df1 = read.table(“file name A.tsv”, sep = “\t”, header = T) df2 = read.table(file, sep = “\t”, header =T) df = merge(df1, df2, by.x = “sample”, by.y = “sample”) df = df[!is.na(df[,2]),] r = cor.test(df[,2], df[,3]) p = cor.test(df[,2], df[,3])$p.value jpeg(filename = paste(sam,”.jpeg”, sep = ““), width = 280, height = 280, units = “px”, pointsize = 12, quality = 75) #bg = “white”, res = 300) plot(df[[2]], df[[3]], xlab = “Title”, ylab = sam, pch = 16, cex = 1, col = “red”, main = paste(“r = “,round(r$estimate,2), “\n”,”p = “,round(p,8), sep = ““)) reg = lm(df[[2]]~df[[3]]) abline(reg) dev.off() } sample = c(“file name B”) for (s in sample) { draw.graph(s) }
Extended Data
Supplementary Material
Acknowledgments:
We thank the National Cancer Institute (1R01CA172268 and 5P30CA142543), the Leukemia & Lymphoma Society (1024–14 and TRP-6024–14), the March of Dimes Foundation (1-FY14–201), the Cancer Prevention and Research Institute of Texas (RP140402, DP150056, RP180435, and PR150551), the Robert A. Welch Foundation (I-1834 and AU-0042–20030616), the National Natural Science Foundation of China (81570093, 81422001, and 81721004), the National Basic Research Program of China (2014CB965000), and the China Scholarship Council (201608330307) for generous support. We also thank Dr. Georgina Salazar for editing of the manuscript.
Footnotes
Data Availability Statement
The TCGA datasets analyzed are available in UCSC Xena Browser (https://xena.ucsc.edu). The RNA-seq datasets generated in the current study have been deposited in NCBI SRA database with the SRA accession number SRP155049 (https://www.ncbi.nlm.nih.gov/sra/SRP155049).
The Board of Regents of the University of Texas System has filed patent applications with PCT Application Nos. PCT/US2016/020838, which covers anti-LILRB antibodies and their uses in detecting and treating cancer, and PCT/US2017/044171, which covers the methods for identifying LILRB-blocking antibodies. Authors C.C.Z., M.D., Z.A., N.Z., X.G., and J.Z. are listed as inventors of PCT/US2016/020838. Authors C.C.Z., Z.A., N.Z., M.D., J.K. and X.G. are listed as inventors of PCT/US2017/044171. Both patent applications have been exclusively licensed to Immune-Onc Therapeutics, Inc. by the Board of Regents of the University of Texas System. Authors T.H. and X.C.L. are employees and hold equities of Immune-Onc Therapeutics, Inc.
References
- 1.Curran EK, Godfrey J & Kline J Mechanisms of Immune Tolerance in Leukemia and Lymphoma. Trends Immunol, doi:10.1016/j.it.2017.04.004 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Kang X et al. Inhibitory leukocyte immunoglobulin-like receptors: Immune checkpoint proteins and tumor sustaining factors. Cell Cycle 15, 25–40, doi:10.1080/15384101.2015.1121324 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Hirayasu K & Arase H Functional and genetic diversity of leukocyte immunoglobulin-like receptor and implication for disease associations. Journal of human genetics, doi:10.1038/jhg.2015.64 (2015). [DOI] [PubMed] [Google Scholar]
- 4.Trowsdale J, Jones DC, Barrow AD & Traherne JA Surveillance of cell and tissue perturbation by receptors in the LRC. Immunol Rev 267, 117–136, doi:10.1111/imr.12314 (2015). [DOI] [PubMed] [Google Scholar]
- 5.Dobrowolska H et al. Expression of immune inhibitory receptor ILT3 in acute myeloid leukemia with monocytic differentiation. Cytometry B Clin Cytom 84, 21–29, doi:10.1002/cyto.b.21050 (2013). [DOI] [PubMed] [Google Scholar]
- 6.Vlad G et al. Membrane and soluble ILT3 are critical to the generation of T suppressor cells and induction of immunological tolerance. Int Rev Immunol 29, 119–132, doi:10.3109/08830180903281185 (2010). [DOI] [PubMed] [Google Scholar]
- 7.Mosier DE, Gulizia RJ, Baird SM & Wilson DB Transfer of a functional human immune system to mice with severe combined immunodeficiency. Nature 335, 256–259, doi:10.1038/335256a0 (1988). [DOI] [PubMed] [Google Scholar]
- 8.Ha S et al. Isolation and characterization of IgG1 with asymmetrical Fc glycosylation. Glycobiology 21, 1087–1096, doi:10.1093/glycob/cwr047 (2011). [DOI] [PubMed] [Google Scholar]
- 9.Straus DJ et al. The acute monocytic leukemias: multidisciplinary studies in 45 patients. Medicine (Baltimore) 59, 409–425 (1980). [DOI] [PubMed] [Google Scholar]
- 10.Colovai AI et al. Expression of inhibitory receptor ILT3 on neoplastic B cells is associated with lymphoid tissue involvement in chronic lymphocytic leukemia. Cytometry B Clin Cytom 72, 354–362, doi:10.1002/cyto.b.20164 (2007). [DOI] [PubMed] [Google Scholar]
- 11.Crispe IN et al. Cellular and molecular mechanisms of liver tolerance. Immunol Rev 213, 101–118, doi:10.1111/j.1600-065X.2006.00435.x (2006). [DOI] [PubMed] [Google Scholar]
- 12.Carson MJ, Doose JM, Melchior B, Schmid CD & Ploix CC CNS immune privilege: hiding in plain sight. Immunol Rev 213, 48–65, doi:10.1111/j.1600-065X.2006.00441.x (2006). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Fujisaki J et al. In vivo imaging of Treg cells providing immune privilege to the haematopoietic stem-cell niche. Nature 474, 216–219, doi:10.1038/nature10160 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Castells MC et al. gp49B1-alpha(v)beta3 interaction inhibits antigen-induced mast cell activation. Nat Immunol 2, 436–442, doi:10.1038/87749 (2001). [DOI] [PubMed] [Google Scholar]
- 15.Grainger DJ, Reckless J & McKilligin E Apolipoprotein E modulates clearance of apoptotic bodies in vitro and in vivo, resulting in a systemic proinflammatory state in apolipoprotein E-deficient mice. J Immunol 173, 6366–6375 (2004). [DOI] [PubMed] [Google Scholar]
- 16.Ali K, Middleton M, Pure E & Rader DJ Apolipoprotein E suppresses the type I inflammatory response in vivo. Circ Res 97, 922–927, doi:10.1161/01.res.0000187467.67684.43 (2005). [DOI] [PubMed] [Google Scholar]
- 17.DiDonato JA, Mercurio F & Karin M NF-kappaB and the link between inflammation and cancer. Immunol Rev 246, 379–400, doi:10.1111/j.1600-065X.2012.01099.x (2012). [DOI] [PubMed] [Google Scholar]
- 18.You M, Flick LM, Yu D & Feng GS Modulation of the nuclear factor kappa B pathway by Shp-2 tyrosine phosphatase in mediating the induction of interleukin (IL)-6 by IL-1 or tumor necrosis factor. J Exp Med 193, 101–110 (2001). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Baumgartner B et al. Increased IkappaB kinase activity is associated with activated NF-kappaB in acute myeloid blasts. Leukemia 16, 2062–2071, doi:10.1038/sj.leu.2402641 (2002). [DOI] [PubMed] [Google Scholar]
- 20.Bene MC et al. CD87 (urokinase-type plasminogen activator receptor), function and pathology in hematological disorders: a review. Leukemia 18, 394–400, doi:10.1038/sj.leu.2403250 (2004). [DOI] [PubMed] [Google Scholar]
- 21.Su SC, Lin CW, Yang WE, Fan WL & Yang SF The urokinase-type plasminogen activator (uPA) system as a biomarker and therapeutic target in human malignancies. Expert Opin Ther Targets 20, 551–566, doi:10.1517/14728222.2016.1113260 (2016). [DOI] [PubMed] [Google Scholar]
- 22.Wang Y et al. Identification of a novel nuclear factor-kappaB sequence involved in expression of urokinase-type plasminogen activator receptor. Eur J Biochem 267, 3248–3254 (2000). [DOI] [PubMed] [Google Scholar]
- 23.Hu J et al. uPAR induces expression of transforming growth factor beta and interleukin-4 in cancer cells to promote tumor-permissive conditioning of macrophages. Am J Pathol 184, 3384–3393, doi:10.1016/j.ajpath.2014.08.003 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Ilkovitch D & Lopez DM Urokinase-mediated recruitment of myeloid-derived suppressor cells and their suppressive mechanisms are blocked by MUC1/sec. Blood 113, 4729–4739, doi:10.1182/blood-2008-08-176438 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Baitsch D et al. Apolipoprotein E induces antiinflammatory phenotype in macrophages. Arterioscler Thromb Vasc Biol 31, 1160–1168, doi:10.1161/atvbaha.111.222745 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Hagemann T et al. “Re-educating” tumor-associated macrophages by targeting NF-kappaB. J Exp Med 205, 1261–1268, doi:10.1084/jem.20080108 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Mussai F et al. Acute myeloid leukemia creates an arginase-dependent immunosuppressive microenvironment. Blood 122, 749–758, doi:10.1182/blood-2013-01-480129 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.de Goeje PL et al. Immunoglobulin-like transcript 3 is expressed by myeloid-derived suppressor cells and correlates with survival in patients with non-small cell lung cancer. Oncoimmunology 4, e1014242, doi:10.1080/2162402x.2015.1014242 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Chang CC et al. Tolerization of dendritic cells by T(S) cells: the crucial role of inhibitory receptors ILT3 and ILT4. Nat Immunol 3, 237–243, doi:10.1038/ni760 (2002). [DOI] [PubMed] [Google Scholar]
- 30.Suciu-Foca N et al. Soluble Ig-like transcript 3 inhibits tumor allograft rejection in humanized SCID mice and T cell responses in cancer patients. J Immunol 178, 7432–7441 (2007). [DOI] [PubMed] [Google Scholar]
- 31.Piedrahita JA, Zhang SH, Hagaman JR, Oliver PM & Maeda N Generation of mice carrying a mutant apolipoprotein E gene inactivated by gene targeting in embryonic stem cells. Proc Natl Acad Sci U S A 89, 4471–4475 (1992). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Zheng J et al. Inhibitory receptors bind ANGPTLs and support blood stem cells and leukaemia development. Nature 485, 656–660, doi:10.1038/nature11095 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Kang X et al. The ITIM-containing receptor LAIR1 is essential for acute myeloid leukaemia development. Nat Cell Biol 17, 665–677, doi:10.1038/ncb3158 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Deng M et al. A motif in LILRB2 critical for Angptl2 binding and activation. Blood 124, 924–935, doi:10.1182/blood-2014-01-549162 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Zheng J et al. Ex vivo expanded hematopoietic stem cells overcome the MHC barrier in allogeneic transplantation. Cell Stem Cell 9, 119–130, doi:10.1016/j.stem.2011.06.003 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Lu Z et al. Fasting selectively blocks development of acute lymphoblastic leukemia via leptin-receptor upregulation. Nat Med 23, 79–90, doi:10.1038/nm.4252 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Zhang CC, Kaba M, Iizuka S, Huynh H & Lodish HF Angiopoietin-like 5 and IGFBP2 stimulate ex vivo expansion of human cord blood hematopoietic stem cells as assayed by NOD/SCID transplantation. Blood 111, 3415–3423, doi:blood-2007-11-122119 [pii] 10.1182/blood-2007-11-122119 [doi] (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Zheng J, Huynh H, Umikawa M, Silvany R & Zhang CC Angiopoietin-like protein 3 supports the activity of hematopoietic stem cells in the bone marrow niche. Blood 117, 470–479, doi:10.1182/blood-2010-06-291716 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Cawthorne C, Swindell R, Stratford IJ, Dive C & Welman A Comparison of doxycycline delivery methods for Tet-inducible gene expression in a subcutaneous xenograft model. J Biomol Tech 18, 120–123 (2007). [PMC free article] [PubMed] [Google Scholar]
- 40.Denisov IG, Grinkova YV, Lazarides AA & Sligar SG Directed self-assembly of monodisperse phospholipid bilayer Nanodiscs with controlled size. J Am Chem Soc 126, 3477–3487, doi:10.1021/ja0393574 (2004). [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.