Tumor- and cytokine-primed human natural killer cells exhibit distinct phenotypic and transcriptional signatures

May Sabry, Agnieszka Zubiak, Simon P Hood, Poppy Simmonds, Helena Arellano-Ballestero, Eily Cournoyer, Meghavi Mashar, A Graham Pockley, Mark W Lowdell, May Sabry, Agnieszka Zubiak, Simon P Hood, Poppy Simmonds, Helena Arellano-Ballestero, Eily Cournoyer, Meghavi Mashar, A Graham Pockley, Mark W Lowdell

Abstract

An emerging cellular immunotherapy for cancer is based on the cytolytic activity of natural killer (NK) cells against a wide range of tumors. Although in vitro activation, or "priming," of NK cells by exposure to pro-inflammatory cytokines, such as interleukin (IL)-2, has been extensively studied, the biological consequences of NK cell activation in response to target cell interactions have not been thoroughly characterized. We investigated the consequences of co-incubation with K562, CTV-1, Daudi RPMI-8226, and MCF-7 tumor cell lines on the phenotype, cytokine expression profile, and transcriptome of human NK cells. We observe the downregulation of several activation receptors including CD16, CD62L, C-X-C chemokine receptor (CXCR)-4, natural killer group 2 member D (NKG2D), DNAX accessory molecule (DNAM)-1, and NKp46 following tumor-priming. Although this NK cell phenotype is typically associated with NK cell dysfunction in cancer, we reveal the upregulation of NK cell activation markers, such as CD69 and CD25; secretion of pro-inflammatory cytokines, including macrophage inflammatory proteins (MIP-1) α /β and IL-1β/6/8; and overexpression of numerous genes associated with enhanced NK cell cytotoxicity and immunomodulatory functions, such as FAS, TNFSF10, MAPK11, TNF, and IFNG. Thus, it appears that tumor-mediated ligation of receptors on NK cells may induce a primed state which may or may not lead to full triggering of the lytic or cytokine secreting machinery. Key signaling molecules exclusively affected by tumor-priming include MAP2K3, MARCKSL1, STAT5A, and TNFAIP3, which are specifically associated with NK cell cytotoxicity against tumor targets. Collectively, these findings help define the phenotypic and transcriptional signature of NK cells following their encounters with tumor cells, independent of cytokine stimulation, and provide insight into tumor-specific NK cell responses to inform the transition toward harnessing the therapeutic potential of NK cells in cancer.

Conflict of interest statement

UCL has patented the use of CTV-1 and other tumor cells for the exvivo priming of NK cells for therapy. Some of the research presented in this manuscript was funded by INmuneBio Inc. and by Novamune Ltd., which are companies that develop clinical products involving NK-priming technology with the intention of taking them into clinical trials. MWL is a co-founder and chief scientific officer of INmuneBio Inc. and MS is a consultant to INmuneBio Inc. The following patent held by UCL is pertinent to this work: PCT/GB2010/051135, “Preserved Compositions of Activated NK Cells and Methods of Using the Same.” This does not alter our adherence to PLOS ONE policies on sharing data and materials.

Figures

Fig 1. Loss of NK cell activation…
Fig 1. Loss of NK cell activation receptor expression following tumor-priming.
(A) Freshly isolated NK cells were incubated in medium alone, with K562, CTV-1, Daudi, RPMI-8226, or MCF-7 cells or with IL-2/12/15/18 (individually or in combination) overnight at 37°C. Thereafter, cells were washed. NK cell activation receptor expression was analyzed using flow cytometry. The percentage change of NK cell expression was calculated for each receptor after stimulation and results are presented as the mean of 3–5 different donors. Red indicates an increase, and green indicates a decrease in receptor expression, according to the key on the right, relative to NK cells in medium alone. (B) Freshly isolated NK cells were incubated in medium alone or with CTV-1 cells overnight at 37°C. Cells were washed, the surface expression of 20 different NK cell activation receptors was analyzed. Bars represent the means ± SD of 5 different donors. The percentages of NK cell expression of different receptors after stimulation was compared with those in cells treated with medium alone using the paired t-test. Statistical significance is indicated as: *P <0.05; **P <0.01; ***P <0.001.
Fig 2. Influence of CTV-1 cell priming…
Fig 2. Influence of CTV-1 cell priming on pro-inflammatory cytokine secretion by NK cells.
(A) Freshly isolated NK cells were incubated alone or with CTV-1 cells for 6 hours at 37°C. Supernatants were harvested, and the concentrations of 25 different cytokines and chemokines were determined using a multiplex immunoassay. Bars represent means ± SD of 4 different donors. Post-stimulation cytokine secretion by NK cells was compared with that of cells incubated in medium alone using the paired t-test. Statistical significance is indicated as: *P <0.05; **P <0.01; ***P <0.001. (B) NK cells were incubated with CTV-1 cells at 37°C. Supernatants were harvested at different time points, as indicated, and the concentrations of IFN-γ, TNF-α, MIP-1α, MIP-1β, and RANTES were determined by a multiplex immunoassay. Values represent mean ± SD of 4 different donors.
Fig 3. Influence of tumor- or cytokine-priming…
Fig 3. Influence of tumor- or cytokine-priming on cytokine secretion by NK cells.
NK cells were incubated in medium alone, with K562, or CTV-1 cells for 6 hours or with IL-2 or IL-12 overnight at 37°C. Supernatants were harvested, and the concentrations of IFN-γ, TNF-α, MIP-1α, MIP-1β, RANTES, IL-R2α, IL-1α, and IL-1β were determined by a multiplex immunoassay. Bars represent means ± SD of 4 different donors. Post-stimulation NK cell cytokine secretion was compared with that in cells incubated in medium alone using the paired t-test. Statistical significance is indicated as: *P <0.05; **P <0.01; ***P <0.001.
Fig 4. Differential influence of tumor cell…
Fig 4. Differential influence of tumor cell or cytokine priming on the transcriptome of NK cells.
(A) Schematic of the workflow in obtaining NK cell gene expression profiles after tumor cell or cytokine priming. Human NK cells were isolated from peripheral blood and incubated in medium alone, mitomycin-C-treated K562, or CTV-1 for 6 h or with IL-2 overnight. NK cells were enriched from co-cultures, and RNA extraction and gene expression profiling were performed using RNA-Seq or the NanoString nCounter FLEX platform. (B) Hierarchical clustering was applied to the gene expression values, and the top differentially expressed genes are shown in a heat map with rows representing the relative expression of each gene across 12 samples. Red corresponds to higher gene expression, and blue corresponds to lower gene expression. Similarity in gene expression patterns between different samples are reflected in the resulting dendrogram. Z-Score is calculated using the following formula: (gene expression value in sample of interest)—(mean expression across all samples) / SD. Venn diagrams generated by the intersection of list of genes upregulated (C) or downregulated (D) by at least 1 Log2fold (P

Fig 5. Top cell signaling network induced…

Fig 5. Top cell signaling network induced in tumor-primed NK (TpNK) cells as generated by…

Fig 5. Top cell signaling network induced in tumor-primed NK (TpNK) cells as generated by Ingenuity pathway analysis.
Nodes represent genes that are either upregulated (red), downregulated (green), or absent from our original differential expression analysis (white), but play an important role in the tumor-specific signaling network according to Ingenuity Pathway Analysis. The intensity of node color signifies the extent of change in expression of the respective gene relative to NK cells incubated in medium alone. Node shapes and the relationships between different genes are outlined in the figure legend.
Fig 5. Top cell signaling network induced…
Fig 5. Top cell signaling network induced in tumor-primed NK (TpNK) cells as generated by Ingenuity pathway analysis.
Nodes represent genes that are either upregulated (red), downregulated (green), or absent from our original differential expression analysis (white), but play an important role in the tumor-specific signaling network according to Ingenuity Pathway Analysis. The intensity of node color signifies the extent of change in expression of the respective gene relative to NK cells incubated in medium alone. Node shapes and the relationships between different genes are outlined in the figure legend.

References

    1. Bryceson YT, March ME, Ljunggren HG, Long EO. Synergy among receptors on resting NK cells for the activation of natural cytotoxicity and cytokine secretion. Blood. 2006;107(1):159–66. 2005-04-1351 [pii]; 10.1182/blood-2005-04-1351
    1. Morvan MG, Lanier LL. NK cells and cancer: you can teach innate cells new tricks. Nat Rev Cancer. 2016;16(1):7–19. 10.1038/nrc.2015.5 .
    1. North J, Bakhsh I, Marden C, Pittman H, Addison E, Navarrete C, et al. Tumor-primed human natural killer cells lyse NK-resistant tumor targets: evidence of a two-stage process in resting NK cell activation. JImmunol. 2007;178(1):85–94. 178/1/85 [pii].
    1. Sabry M, Tsirogianni M, Bakhsh IA, North J, Sivakumaran J, Giannopoulos K, et al. Leukemic priming of resting NK cells is killer Ig-like receptor independent but requires CD15-mediated CD2 ligation and natural cytotoxicity receptors. JImmunol. 2011;187(12):6227–34. jimmunol.1101640 [pii]; 10.4049/jimmunol.1101640
    1. Meazza R, Azzarone B, Orengo AM, Ferrini S. Role of common-gamma chain cytokines in NK cell development and function: perspectives for immunotherapy. J Biomed Biotechnol. 2011;2011:861920 10.1155/2011/861920
    1. Wang F, Tian Z, Wei H. Genomic expression profiling of NK cells in health and disease. Eur J Immunol. 2015;45(3):661–78. Epub 2014/12/06. 10.1002/eji.201444998 .
    1. Bottcher A, Ostwald J, Koczan D, Knecht R, Kramp B, Dommerich S. Gene expression profiling of circulating natural killer cells in head and neck squamous cell carcinoma. Cancer Genomics Proteomics. 2013;10(5):197–207. .
    1. Gillard-Bocquet M, Caer C, Cagnard N, Crozet L, Perez M, Fridman WH, et al. Lung tumor microenvironment induces specific gene expression signature in intratumoral NK cells. Front Immunol. 2013;4:19 10.3389/fimmu.2013.00019
    1. Srpan K, Ambrose A, Karampatzakis A, Saeed M, Cartwright ANR, Guldevall K, et al. Shedding of CD16 disassembles the NK cell immune synapse and boosts serial engagement of target cells. J Cell Biol. 2018;217(9):3267–83. Epub 2018/07/04. 10.1083/jcb.201712085
    1. Fauriat C, Long EO, Ljunggren HG, Bryceson YT. Regulation of human NK-cell cytokine and chemokine production by target cell recognition. Blood. 2010;115(11):2167–76. blood-2009-08-238469 [pii]; 10.1182/blood-2009-08-238469
    1. Jewett A, Bonavida B. Target-induced anergy of natural killer cytotoxic function is restricted to the NK-target conjugate subset. Cell Immunol. 1995;160(1):91–7. Epub 1995/01/01. .
    1. Cavalcanti M, Jewett A, Bonavida B. Irreversible cancer cell-induced functional anergy and apoptosis in resting and activated NK cells. Int J Oncol. 1999;14(2):361–6. Epub 1999/01/26. 10.3892/ijo.14.2.361 .
    1. Jewett A, Bonavida B. Target-induced inactivation and cell death by apoptosis in a subset of human NK cells. J Immunol. 1996;156(3):907–15. Epub 1996/02/01. .
    1. Nieto-Velazquez NG, Torres-Ramos YD, Munoz-Sanchez JL, Espinosa-Godoy L, Gomez-Cortes S, Moreno J, et al. Altered Expression of Natural Cytotoxicity Receptors and NKG2D on Peripheral Blood NK Cell Subsets in Breast Cancer Patients. Transl Oncol. 2016;9(5):384–91. 10.1016/j.tranon.2016.07.003
    1. Kono K, Ressing ME, Brandt RM, Melief CJ, Potkul RK, Andersson B, et al. Decreased expression of signal-transducing zeta chain in peripheral T cells and natural killer cells in patients with cervical cancer. ClinCancer Res. 1996;2(11):1825–8.
    1. Fauriat C, Mallet F, Olive D, Costello RT. Impaired activating receptor expression pattern in natural killer cells from patients with multiple myeloma. Leukemia. 2006;20(4):732–3. 10.1038/sj.leu.2404096 .
    1. Romee R, Foley B, Lenvik T, Wang Y, Zhang B, Ankarlo D, et al. NK cell CD16 surface expression and function is regulated by a disintegrin and metalloprotease-17 (ADAM17). Blood. 2013;121(18):3599–608. blood-2012-04-425397 [pii]; 10.1182/blood-2012-04-425397
    1. Warren HS, Altin JG, Waldron JC, Kinnear BF, Parish CR. A carbohydrate structure associated with CD15 (Lewis x) on myeloid cells is a novel ligand for human CD2. JImmunol. 1996;156(8):2866–73.
    1. Casu B, Dondero A, Regis S, Caliendo F, Petretto A, Bartolucci M, et al. Novel Immunoregulatory Functions of IL-18, an Accomplice of TGF-beta1. Cancers (Basel). 2019;11(1). Epub 2019/01/16. 10.3390/cancers11010075
    1. Foltz JA, Moseman JE, Thakkar A, Chakravarti N, Lee DA. TGFbeta Imprinting During Activation Promotes Natural Killer Cell Cytokine Hypersecretion. Cancers (Basel). 2018;10(11). Epub 2018/11/08. 10.3390/cancers10110423
    1. Cuturi MC, Anegon I, Sherman F, Loudon R, Clark SC, Perussia B, et al. Production of hematopoietic colony-stimulating factors by human natural killer cells. JExpMed. 1989;169(2):569–83.
    1. Warren HS, Kinnear BF, Phillips JH, Lanier LL. Production of IL-5 by human NK cells and regulation of IL-5 secretion by IL-4, IL-10, and IL-12. JImmunol. 1995;154(10):5144–52.
    1. Pockley AG, Henderson B. Extracellular cell stress (heat shock) proteins-immune responses and disease: an overview. Philos Trans R Soc Lond B Biol Sci. 2018;373(1738). Epub 2017/12/06. 10.1098/rstb.2016.0522
    1. Hodge DL, Schill WB, Wang JM, Blanca I, Reynolds DA, Ortaldo JR, et al. IL-2 and IL-12 alter NK cell responsiveness to IFN-gamma-inducible protein 10 by down-regulating CXCR3 expression. J Immunol. 2002;168(12):6090–8. 10.4049/jimmunol.168.12.6090 .
    1. Liu Y, Beyer A, Aebersold R. On the Dependency of Cellular Protein Levels on mRNA Abundance. Cell. 2016;165(3):535–50. Epub 2016/04/23. 10.1016/j.cell.2016.03.014 .
    1. Yokoyama WM, Kim S, French AR. The dynamic life of natural killer cells. Annu Rev Immunol. 2004;22:405–29. 10.1146/annurev.immunol.22.012703.104711 .
    1. Konjevic G, Mirjacic MK, Vuletic A, Jovic V, Jurisic V, Babovic N, et al. Low expression of CD161 and NKG2D activating NK receptor is associated with impaired NK cell cytotoxicity in metastatic melanoma patients. ClinExpMetastasis. 2007;24(1):1–11. 10.1007/s10585-006-9043-9
    1. Healy CG, Simons JW, Carducci MA, DeWeese TL, Bartkowski M, Tong KP, et al. Impaired expression and function of signal-transducing zeta chains in peripheral T cells and natural killer cells in patients with prostate cancer. Cytometry. 1998;32(2):109–19. 10.1002/(SICI)1097-0320(19980601)32:2<109::AID-CYTO6>;2-G [pii].
    1. Costello RT, Sivori S, Marcenaro E, Lafage-Pochitaloff M, Mozziconacci MJ, Reviron D, et al. Defective expression and function of natural killer cell-triggering receptors in patients with acute myeloid leukemia. Blood. 2002;99(10):3661–7. 10.1182/blood.v99.10.3661
    1. El-Sherbiny YM, Meade JL, Holmes TD, McGonagle D, Mackie SL, Morgan AW, et al. The requirement for DNAM-1, NKG2D, and NKp46 in the natural killer cell-mediated killing of myeloma cells. Cancer Res. 2007;67(18):8444–9. 67/18/8444 [pii]; 10.1158/0008-5472.CAN-06-4230
    1. Ogasawara K, Hamerman JA, Hsin H, Chikuma S, Bour-Jordan H, Chen T, et al. Impairment of NK cell function by NKG2D modulation in NOD mice. Immunity. 2003;18(1):41–51. Epub 2003/01/18. .
    1. Quatrini L, Molfetta R, Zitti B, Peruzzi G, Fionda C, Capuano C, et al. Ubiquitin-dependent endocytosis of NKG2D-DAP10 receptor complexes activates signaling and functions in human NK cells. Sci Signal. 2015;8(400):ra108 Epub 2015/10/29. 10.1126/scisignal.aab2724 .
    1. Castriconi R, Cantoni C, Della Chiesa M, Vitale M, Marcenaro E, Conte R, et al. Transforming growth factor beta 1 inhibits expression of NKp30 and NKG2D receptors: consequences for the NK-mediated killing of dendritic cells. Proc Natl Acad Sci U S A. 2003;100(7):4120–5. Epub 2003/03/21. 10.1073/pnas.0730640100
    1. Lee JC, Lee KM, Kim DW, Heo DS. Elevated TGF-beta1 secretion and down-modulation of NKG2D underlies impaired NK cytotoxicity in cancer patients. J Immunol. 2004;172(12):7335–40. Epub 2004/06/10. 10.4049/jimmunol.172.12.7335 .
    1. Warren HS, Altin JG, Waldron JC, Kinnear BF, Parish CR. A carbohydrate structure associated with CD15 (Lewis x) on myeloid cells is a novel ligand for human CD2. J Immunol. 1996;156(8):2866–73. .
    1. Peruzzi G, Femnou L, Gil-Krzewska A, Borrego F, Weck J, Krzewski K, et al. Membrane-type 6 matrix metalloproteinase regulates the activation-induced downmodulation of CD16 in human primary NK cells. J Immunol. 2013;191(4):1883–94. Epub 2013/07/16. 10.4049/jimmunol.1300313
    1. Gati A, Da RS, Guerra N, Escudier B, Moretta A, Chouaib S, et al. Analysis of the natural killer mediated immune response in metastatic renal cell carcinoma patients. IntJCancer. 2004;109(3):393–401. 10.1002/ijc.11730
    1. Penack O, Gentilini C, Fischer L, Asemissen AM, Scheibenbogen C, Thiel E, et al. CD56dimCD16neg cells are responsible for natural cytotoxicity against tumor targets. Leukemia. 2005;19(5):835–40. 2403704 [pii]; 10.1038/sj.leu.2403704
    1. Grzywacz B, Kataria N, Verneris MR. CD56(dim)CD16(+) NK cells downregulate CD16 following target cell induced activation of matrix metalloproteinases. Leukemia. 2007;21(2):356–9; author reply 9. 10.1038/sj.leu.2404499 .
    1. Peduto L. ADAM9 as a potential target molecule in cancer. Curr Pharm Des. 2009;15(20):2282–7. Epub 2009/07/16. .
    1. Fehniger TA, Miller JS, Stuart RK, Cooley S, Salhotra A, Curtsinger J, et al. A Phase 1 Trial of CNDO-109-Activated Natural Killer Cells in Patients with High-Risk Acute Myeloid Leukemia. Biol Blood Marrow Transplant. 2018;24(8):1581–9. Epub 2018/03/30. 10.1016/j.bbmt.2018.03.019 .
    1. Kottaridis PD, North J, Tsirogianni M, Marden C, Samuel ER, Jide-Banwo S, et al. Two-Stage Priming of Allogeneic Natural Killer Cells for the Treatment of Patients with Acute Myeloid Leukemia: A Phase I Trial. PLoS One. 2015;10(6):e0123416 Epub 2015/06/11. 10.1371/journal.pone.0123416
    1. Casetti L, Martin-Lanneree S, Najjar I, Plo I, Auge S, Roy L, et al. Differential contributions of STAT5A and STAT5B to stress protection and tyrosine kinase inhibitor resistance of chronic myeloid leukemia stem/progenitor cells. Cancer Res. 2013;73(7):2052–8. Epub 2013/02/13. 10.1158/0008-5472.CAN-12-3955 .
    1. Wei S, Gamero AM, Liu JH, Daulton AA, Valkov NI, Trapani JA, et al. Control of lytic function by mitogen-activated protein kinase/extracellular regulatory kinase 2 (ERK2) in a human natural killer cell line: identification of perforin and granzyme B mobilization by functional ERK2. J Exp Med. 1998;187(11):1753–65. Epub 1998/06/10. 10.1084/jem.187.11.1753
    1. Jeffrey KL, Brummer T, Rolph MS, Liu SM, Callejas NA, Grumont RJ, et al. Positive regulation of immune cell function and inflammatory responses by phosphatase PAC-1. Nat Immunol. 2006;7(3):274–83. 10.1038/ni1310 .
    1. Pal M, Schwab L, Yermakova A, Mace EM, Claus R, Krahl AC, et al. Tumor-priming converts NK cells to memory-like NK cells. Oncoimmunology. 2017;6(6):e1317411 Epub 2017/07/07. 10.1080/2162402X.2017.1317411
    1. Park KU, Jin P, Sabatino M, Feng J, Civini S, Khuu H, et al. Gene expression analysis of ex vivo expanded and freshly isolated NK cells from cancer patients. J Immunother. 2010;33(9):945–55. 10.1097/CJI.0b013e3181f71b81
    1. Fehniger TA, Cai SF, Cao X, Bredemeyer AJ, Presti RM, French AR, et al. Acquisition of murine NK cell cytotoxicity requires the translation of a pre-existing pool of granzyme B and perforin mRNAs. Immunity. 2007;26(6):798–811. 10.1016/j.immuni.2007.04.010 .

Source: PubMed

3
Subscribe