Ex-vivo expanded human NK cells express activating receptors that mediate cytotoxicity of allogeneic and autologous cancer cell lines by direct recognition and antibody directed cellular cytotoxicity

Caroline J Voskens, Ryuko Watanabe, Sandra Rollins, Dario Campana, Kenichiro Hasumi, Dean L Mann, Caroline J Voskens, Ryuko Watanabe, Sandra Rollins, Dario Campana, Kenichiro Hasumi, Dean L Mann

Abstract

Background: The possibility that autologous NK cells could serve as an effective treatment modality for solid tumors has long been considered. However, implementation is hampered by (i) the small number of NK cells in peripheral blood, (ii) the difficulties associated with large-scale production of GMP compliant cytolytic NK cells, (iii) the need to activate the NK cells in order to induce NK cell mediated killing and (iv) the constraints imposed by autologous inhibitory receptor-ligand interactions. To address these issues, we determined (i) if large numbers of NK cells could be expanded from PBMC and GMP compliant cell fractions derived by elutriation, (ii) their ability to kill allogeneic and autologous tumor targets by direct cytotoxicity and by antibody-mediated cellular cytotoxicity and (iii) defined NK cell specific receptor-ligand interactions that mediate tumor target cell killing.

Methods: Human NK cells were expanded during 14 days. Expansion efficiency, NK receptor repertoire before and after expansion, expression of NK specific ligands, cytolytic activity against allogeneic and autologous tumor targets, with and without the addition of chimeric EGFR monoclonal antibody, were investigated.

Results: Cell expansion shifted the NK cell receptor repertoire towards activation and resulted in cytotoxicity against various allogeneic tumor cell lines and autologous gastric cancer cells, while sparing normal PBMC. Blocking studies confirmed that autologous cytotoxicity is established through multiple activating receptor-ligand interactions. Importantly, expanded NK cells also mediated ADCC in an autologous and allogeneic setting by antibodies that are currently being used to treat patients with select solid tumors.

Conclusion: These data demonstrate that large numbers of cytolytic NK cells can be generated from PBMC and lymphocyte-enriched fractions obtained by GMP compliant counter current elutriation from PBMC, establishing the preclinical evidence necessary to support clinical trials utilizing autologous expanded NK cells, both directly and in combination with monoclonal antibodies in future cell-based immunotherapy in select solid tumors.

Figures

Figure 1
Figure 1
Cytolytic NK cells are efficiently expanded from PBMC. In the presence of K562-IL15-41BBL (A) expanded cells become significantly enriched (P = 0.0307) in NK cells (defined by CD56+CD3- cells) after 14 days of culture. Expanded cells were evaluated for cytolytic activity using 4 hour 51Cr release assays. Ex-vivo expanded cells from PBMC (■ donor 1 and △ donor 2), but not freshly purified non-expanded NK cells (◇), efficiently lysed allogeneic tumor cell lines derived from breast (MCF-7) and prostate (LNCaP) cancers but not allogeneic or autologous PBMC derived from donor 1 (B). The mean percentage cytotoxicity is shown from triplicate wells. Error bars represent the SD. Lytic activity is likely mediated by NK cells in the expanded cell population (○) since separation in individual populations of NK cells (◇) and NKT/T cells (△) resulted in allogeneic cytolytic activity of the expanded cell population and the purified NK cell population. Little lytic activity was observed in the presence of NKT/T cells alone (C). The mean percentage cytotoxicity is shown from triplicate wells from one representative experiment. Error bars represent the SD. Experiment shown represents one of three individual experiments with three different donors.
Figure 2
Figure 2
Ex-vivo expanded NK cells recognize autologous gastric tumor cells through different activating receptor-ligand interactions. PBMC from two gastric cancer patients were ex-vivo expanded for 14 days and then tested for cytolytic activity against autologous gastric tumor cells in 4 hour 51Cr release assays. (A) Ex-vivo expanded cells from both patients (patient 1 and patient 2), but not freshly purified non-expanded NK cells from patient 2, efficiently lysed autologous gastric tumor cells (Effector to Target Ratio 20:1). To evaluate the impact of activating receptor-ligand interactions on autologous tumor cell lysis indicated blocking antibodies (10 μg/ml) were added during 4 hours of incubation. (B) Cytotoxicity was reduced in the presence of DNAM-1 (P = 0.0309) and NKp30 (P = 0.0056) for patient 1 and in the presence of NKp46 (P = 0.0003) for patient 2. In both patients autologous cytolytic activity was abrogated in the presence of all four blocking antibodies with P = 0.0111 and P = 0.0001, respectively. Statistical analysis is based on triplicate wells of four (patient 1) and two (patient 2) experiments performed, respectively. Error bars represent the SD. * P < 0.05. MoIgG1 indicates mouse IgG1.
Figure 3
Figure 3
Cetuximab significantly enhances cytolytic activity and ADCC is negatively affected by inhibition of activating receptor-ligand interactions. Ex-vivo expanded cells from cancer patient 1 were evaluated for their ability to mediate ADCC against autologous (patient 1) and allogeneic (TU-167 and H-358) EGFR expressing lung cancer cells. (A) Cytolytic activity of ex-vivo expanded cells was enhanced in the presence of Cetuximab (10 μg/ml, black bar) but not in the presence of control human IgG1 (10 μg/ml; dotted bar) or media alone (white bar). The mean percentage cytotoxicity is shown from triplicate wells from one representative experiment. Error bars represent the SD. Experiment shown represents one of two individual experiments. (B) The addition of blocking antibodies (10 μg/ml) against DNAM-1, NKp46, NKp44 and NKp30 (= all) significantly reduced (P = 0.0176) Cetuximab-mediated ADCC. Statistical analysis is based on three experiments performed. Error bars represent the SD. * P < 0.05. HuIgG1 indicates human IgG1, Ctx; Cetuximab and moIgG1; mouse IgG1.
Figure 4
Figure 4
Distribution of lineage-specific phenotypic markers on PBMC and separate cell fractions obtained after counter current elutriation. PBMC and elutriated cell fractions were stained with various lineage-specific directly-conjugated antibodies and analyzed by flow cytometry (A). Average number of cells and phenotypic distribution (%) expressing lineage-markers in elutriated cell fractions (n = 11) (B).
Figure 5
Figure 5
Ex-vivo expanded cells from elutriated cell fractions efficiently lyse allogeneic prostate cancer cells. PBMC and elutriated fractions 2, 3 and 4 from the same healthy individual were expanded ex-vivo in the presence of K562-mbIL15-41BBL and IL-2 for 14 days and then tested for in vitro cytolytic activity. Cytolytic activity was evaluated in 4 hour 51Cr release assays against (A) prostate cancer (DU-145, PC-3 and LNCaP) cells. Ex-vivo expanded cells from elutriated cell fractions 2 (◇), 3 (△) and 4 (□) lysed prostate cancer cells in a similar fashion as ex-vivo expanded cells from PBMC (○). (B) Elutriated cell fractions become enriched in NK cells (defined by CD56+CD3- cells) after 14 days of culture regardless the cellular content of these fractions. The mean percentage cytotoxicity is shown from triplicate wells from one representative experiment. Bars represent the SD. Experiment shown represents one of four individual experiments.

References

    1. Kiessling R, Klein E, Wigzell H. "Natural" killer cells in the mouse. I. Cytotoxic cells with specificity for mouse Moloney leukemia cells. Specificity and distribution according to genotype. Eur J Immunol. 1975;5:112–117. doi: 10.1002/eji.1830050208.
    1. Kiessling R, Klein E, Pross H, Wigzell H. "Natural" killer cells in the mouse. II. Cytotoxic cells with specificity for mouse Moloney leukemia cells. Characteristics of the killer cell. Eur J Immunol. 1975;5:117–121. doi: 10.1002/eji.1830050209.
    1. Herberman RB, Nunn ME, Lavrin DH. Natural cytotoxic reactivity of mouse lymphoid cells against syngeneic acid allogeneic tumors. I. Distribution of reactivity and specificity. Int J Cancer. 1975;16:216–229. doi: 10.1002/ijc.2910160204.
    1. Herberman RB, Nunn ME, Holden HT, Lavrin DH. Natural cytotoxic reactivity of mouse lymphoid cells against syngeneic and allogeneic tumors. II. Characterization of effector cells. Int J Cancer. 1975;16:230–239. doi: 10.1002/ijc.2910160205.
    1. Ljunggren HG, Malmberg KJ. Prospects for the use of NK cells in immunotherapy of human cancer. Nat Rev Immunol. 2007;7:329–339. doi: 10.1038/nri2073.
    1. Cooper MA, Fehniger TA, Caligiuri MA. The biology of human natural killer-cell subsets. Trends Immunol. 2001;22:633–640. doi: 10.1016/S1471-4906(01)02060-9.
    1. Karre K, Ljunggren HG, Piontek G, Kiessling R. Selective rejection of H-2-deficient lymphoma variants suggests alternative immune defence strategy. Nature. 1986;319:675–678. doi: 10.1038/319675a0.
    1. Ruggeri L, Capanni M, Casucci M, Volpi I, Tosti A, Perruccio K, Urbani E, Negrin RS, Martelli MF, Velardi A. Role of natural killer cell alloreactivity in HLA-mismatched hematopoietic stem cell transplantation. Blood. 1999;94:333–339.
    1. Ruggeri L, Capanni M, Urbani E, Perruccio K, Shlomchik WD, Tosti A, Posati S, Rogaia D, Frassoni F, Aversa F, Martelli MF, Velardi A. Effectiveness of donor natural killer cell alloreactivity in mismatched hematopoietic transplants. Science. 2002;295:2097–2100. doi: 10.1126/science.1068440.
    1. Shlomchik WD. Graft-versus-host disease. Nat Rev Immunol. 2007;7:340–352. doi: 10.1038/nri2000.
    1. Rosenberg SA, Lotze MT, Muul LM, Leitman S, Chang AE, Ettinghausen SE, Matory YL, Skibber JM, Shiloni E, Vetto JT. et al.Observations on the systemic administration of autologous lymphokine-activated killer cells and recombinant interleukin-2 to patients with metastatic cancer. N Engl J Med. 1985;313:1485–1492. doi: 10.1056/NEJM198512053132327.
    1. Imai C, Iwamoto S, Campana D. Genetic modification of primary natural killer cells overcomes inhibitory signals and induces specific killing of leukemic cells. Blood. 2005;106:376–383. doi: 10.1182/blood-2004-12-4797.
    1. Berg M, Lundqvist A, McCoy P Jr, Samsel L, Fan Y, Tawab A, Childs R. Clinical-grade ex vivo-expanded human natural killer cells up-regulate activating receptors and death receptor ligands and have enhanced cytolytic activity against tumor cells. Cytotherapy. 2009;11:341–355. doi: 10.1080/14653240902807034.
    1. Miller JS, Oelkers S, Verfaillie C, McGlave P. Role of monocytes in the expansion of human activated natural killer cells. Blood. 1992;80:2221–2229.
    1. Miller JS, Soignier Y, Panoskaltsis-Mortari A, McNearney SA, Yun GH, Fautsch SK, McKenna D, Le C, Defor TE, Burns LJ, Orchard PJ, Blazar BR, Wagner JE, Slungaard A, Weisdorf DJ, Okazaki IJ, McGlave PB. Successful adoptive transfer and in vivo expansion of human haploidentical NK cells in patients with cancer. Blood. 2005;105:3051–3057. doi: 10.1182/blood-2004-07-2974.
    1. Sedlmayr P, Rabinowich H, Winkelstein A, Herberman RB, Whiteside TL. Generation of adherent lymphokine activated killer (A-LAK) cells from patients with acute myelogenous leukaemia. Br J Cancer. 1992;65:222–228.
    1. Fujisaki H, Kakuda H, Shimasaki N, Imai C, Ma J, Lockey T, Eldridge P, Leung WH, Campana D. Expansion of highly cytotoxic human natural killer cells for cancer cell therapy. Cancer Res. 2009;69:4010–4017. doi: 10.1158/0008-5472.CAN-08-3712.
    1. Berger TG, Strasser E, Smith R, Carste C, Schuler-Thurner B, Kaempgen E, Schuler G. Efficient elutriation of monocytes within a closed system (Elutra) for clinical-scale generation of dendritic cells. J Immunol Methods. 2005;298:61–72. doi: 10.1016/j.jim.2005.01.005.
    1. Lin W, Voskens CJ, Zhang X, Schindler DG, Wood A, Burch E, Wei Y, Chen L, Tian G, Tamada K, Wang LX, Schulze DH, Mann D, Strome SE. Fc-dependent expression of CD137 on human NK cells: insights into "agonistic" effects of anti-CD137 monoclonal antibodies. Blood. 2008;112:699–707. doi: 10.1182/blood-2007-11-122465.
    1. Kabelitz D, Wesch D, He W. Perspectives of gammadelta T cells in tumor immunology. Cancer Res. 2007;67:5–8. doi: 10.1158/0008-5472.CAN-06-3069.
    1. Lanier LL. NK cell recognition. Annu Rev Immunol. 2005;23:225–274. doi: 10.1146/annurev.immunol.23.021704.115526.
    1. Becknell B, Caligiuri MA. Interleukin-2, interleukin-15, and their roles in human natural killer cells. Adv Immunol. 2005;86:209–239. full_text.
    1. Brooks AG, Posch PE, Scorzelli CJ, Borrego F, Coligan JE. NKG2A complexed with CD94 defines a novel inhibitory natural killer cell receptor. J Exp Med. 1997;185:795–800. doi: 10.1084/jem.185.4.795.
    1. Braud VM, Allan DS, O'Callaghan CA, Soderstrom K, D'Andrea A, Ogg GS, Lazetic S, Young NT, Bell JI, Phillips JH, Lanier LL, McMichael AJ. HLA-E binds to natural killer cell receptors CD94/NKG2A, B and C. Nature. 1998;391:795–799. doi: 10.1038/35869.
    1. Shiroishi M, Tsumoto K, Amano K, Shirakihara Y, Colonna M, Braud VM, Allan DS, Makadzange A, Rowland-Jones S, Willcox B, Jones EY, van der Merwe PA, Kumagai I, Maenaka K. Human inhibitory receptors Ig-like transcript 2 (ILT2) and ILT4 compete with CD8 for MHC class I binding and bind preferentially to HLA-G. Proc Natl Acad Sci USA. 2003;100:8856–8861. doi: 10.1073/pnas.1431057100.
    1. Morton DL, Goodnight JE Jr. Clinical trials of immunotherapy: present status. Cancer. 1978;42:2224–2233. doi: 10.1002/1097-0142(197811)42:5<2224::AID-CNCR2820420521>;2-F.
    1. Disis ML, Bernhard H, Jaffee EM. Use of tumour-responsive T cells as cancer treatment. Lancet. 2009;373:673–683. doi: 10.1016/S0140-6736(09)60404-9.
    1. Kurai J, Chikumi H, Hashimoto K, Yamaguchi K, Yamasaki A, Sako T, Touge H, Makino H, Takata M, Miyata M, Nakamoto M, Burioka N, Shimizu E. Antibody-dependent cellular cytotoxicity mediated by cetuximab against lung cancer cell lines. Clin Cancer Res. 2007;13:1552–1561. doi: 10.1158/1078-0432.CCR-06-1726.
    1. Raben D, Helfrich B, Chan DC, Ciardiello F, Zhao L, Franklin W, Baron AE, Zeng C, Johnson TK, Bunn PA Jr. The effects of cetuximab alone and in combination with radiation and/or chemotherapy in lung cancer. Clin Cancer Res. 2005;11:795–805.
    1. Riddell SR, Appelbaum FR. Graft-versus-host disease: a surge of developments. PLoS Med. 2007;4:e198. doi: 10.1371/journal.pmed.0040198.
    1. North J, Bakhsh I, Marden C, Pittman H, Addison E, Navarrete C, Anderson R, Lowdell MW. Tumor-primed human natural killer cells lyse NK-resistant tumor targets: evidence of a two-stage process in resting NK cell activation. J Immunol. 2007;178:85–94.
    1. Lakshmikanth T, Burke S, Ali TH, Kimpfler S, Ursini F, Ruggeri L, Capanni M, Umansky V, Paschen A, Sucker A, Pende D, Groh V, Biassoni R, Hoglund P, Kato M, Shibuya K, Schadendorf D, Anichini A, Ferrone S, Velardi A, Karre K, Shibuya A, Carbone E, Colucci F. NCRs and DNAM-1 mediate NK cell recognition and lysis of human and mouse melanoma cell lines in vitro and in vivo. J Clin Invest. 2009;119:1251–1263. doi: 10.1172/JCI36022.
    1. Ungefroren H, Voss M, Jansen M, Roeder C, Henne-Bruns D, Kremer B, Kalthoff H. Human pancreatic adenocarcinomas express Fas and Fas ligand yet are resistant to Fas-mediated apoptosis. Cancer Res. 1998;58:1741–1749.
    1. Alici E, Sutlu T, Bjorkstrand B, Gilljam M, Stellan B, Nahi H, Quezada HC, Gahrton G, Ljunggren HG, Dilber MS. Autologous antitumor activity by NK cells expanded from myeloma patients using GMP-compliant components. Blood. 2008;111:3155–3162. doi: 10.1182/blood-2007-09-110312.
    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;107:159–166. doi: 10.1182/blood-2005-04-1351.
    1. Erdmann M, Dorrie J, Schaft N, Strasser E, Hendelmeier M, Kampgen E, Schuler G, Schuler-Thurner B. Effective clinical-scale production of dendritic cell vaccines by monocyte elutriation directly in medium, subsequent culture in bags and final antigen loading using peptides or RNA transfection. J Immunother. 2007;30:663–674. doi: 10.1097/CJI.0b013e3180ca7cd6.
    1. Zobywalski A, Javorovic M, Frankenberger B, Pohla H, Kremmer E, Bigalke I, Schendel DJ. Generation of clinical grade dendritic cells with capacity to produce biologically active IL-12p70. J Transl Med. 2007;5:18. doi: 10.1186/1479-5876-5-18.

Source: PubMed

3
订阅