720
Views
3
CrossRef citations to date
0
Altmetric
Editorial

An allogeneic NK cell line engineered to express chimeric antigen receptors

A novel strategy of cellular immunotherapy against cancer

, , , &
Article: e27156 | Received 01 Nov 2013, Accepted 01 Nov 2013, Published online: 14 Nov 2013

In recent years, several immunotherapeutic approaches against cancer have emerged, including the adoptive transfer of T cells that have been reprogrammed to react against one or more tumor-associated antigens (TAAs). In most cases, reprogramming is achieved as T cells are engineered to express the antigen-binding domain of a TAA-specific monoclonal antibody fused to intracellular domains that are normally involved in T-cell receptor (TCR) signaling and/or co-stimulation.Citation1-Citation3 In small cohorts of patients affected by hematological malignancies including both low-grade and aggressive B-cell neoplasms, significant, long-lasting clinical responses were observed upon the administration of these chimeric antigen receptor (CAR)-expressing T cells.Citation4,Citation5

To circumvent the use of autologous T cells, which requires labor-intensive steps of isolation and expansion in vitro, Klingermann’s group developed an allogeneic natural killer (NK) cell line (NK-92 cells) that express CARs comprising single-chain variable fragments from murine CD19- or CD20-specific antibodies.Citation6 NK cells are advantageous as compared with T cells as they recognize malignant cells in a HLA-unrestricted manner and can lyse them in the absence of pre-sensitization.Citation7 NK cells exert cytotoxic functions by multiple mechanisms, including (1) the direct release of granzyme and perforin upon the physical interaction with target cells, (2) the so-called “antibody-dependent cell-mediated cytotoxicity” (ADCC), a process whereby NK cells kill their targets once these are opsonized by antibodies, following the binding of constant fragments (Fcs) to Fc fragment of IgG, low affinity III, receptors (FcγRIIIs), (3) the secretion of TH1 cytokines, and (4) via the granzyme/perforin pathway upon activation by dendritic cells.Citation8 However, the infusion of unmodified NK cells, both as a standalone intervention and in combination with immunostimulatory cytokines, failed to induce any significant diseases regression in patients affected by multiple solid tumors.Citation9

In a recent issue of OncoImmunology, Boissel et al. demonstrated that NK-92 cells engineered to express CD20-specific CARs exhibit improved cytotoxicity against primary chronic lymphocytic leukemia (CLL) cells in vitro as compared with parental NK-92 cells pulsed with various anti-CD20 monoclonal antibodies. Of note, Boissel et al. did not use the same anti-CD20 monoclonal antibodies to compare the cytotoxicity of CAR-expressing NK-92 cells to that of NK-92 cells primed for ADCC, which may have introduced a bias. Indeed, the binding affinity of monoclonal antibodies for their targets is known to affect cytotoxicity.Citation10 However, these results confirm and extend previous reports demonstrating the superiority of CAR-expressing NK cells over NK cells pulsed with TAA-specific antibodies at lysing cancer cells.Citation11 Taken together, these observations suggest that administration of monoclonal antibodies should be less effective than that of (NK or T) cells engineered to express a CAR based on the same molecule. Although no comparison of the clinical activity of these two immunotherapeutic strategies has been performed, T cells modified o express CD19-specific CARs have been shown to be clinically effective in patients that are resistant to a chimeric bispecific antibody targeting CD3 and CD19 (blinatumomab).Citation4 Unlike antibodies, CAR-modified cells have the potential to replicate in vivo, and the long-term persistence of these cells might underlie sustained disease control, eliminating the need for repeated infusions.Citation12

The adoptive transfer of NK-92 cells expressing CD19-targeting CARs effectively eradicated human SUP-B15, but not TMD-5, leukemia cells growing in immunodeficient mice.Citation6 Various hypotheses can be put forward to explain this differential activity, including variations in the levels of expression of TAAs (in this case, CD19) on the surface of leukemia cells, the differential sensitivity of SUP-B15 and TMD-5 cells to apoptosis as triggered by CAR-modified NK-92 cells, and the existence of specific mechanisms developed by TMD-5 cells to escape the antineoplastic activity of NK cells. To test the hypothesis that TMD-5 cells escape the cytotoxicity of CAR-modified NK-92 cells because the CAR-bearing NK-92 cells fail to infiltrate the neoplastic bone marrow, Boissel et al. injected CAR-expressing NK cells directly into bone marrow, observing a significant antitumor effect locally but not at distant sites. Moreover, intravenously injected CAR-expressing NK-92 cells were found in the peripheral blood and spleen but not in the bone marrow, confirming the hypothesis that TMD-5 cells are insensitive to this therapeutic approach due to a homing issue. To explain such a homing bias at the molecular level, it would have been of interest to analyze the integrin and chemokine receptor profile of CAR-expressing NK-92 cells. The relocalization of antigen-experienced CD4+ T cells to bone marrow is dependent on integrin α2β1, a collagen receptor.Citation13 However, since CAR-bearing NK-92 cells were able to cure SUP-B15 acute lymphoid leukemia cells (which also infiltrated the bone marrow), not only the phenotype of NK-92 cells but also features of the tumor microenvironment created by TMD-5 cells might explain their resistance to this immunotherapeutic approach.

Various strategies have been developed to correct defects in the homing of effector T or NK cells within neoplastic lesions.Citation14 For example, imatinib, a targeted anticancer agent, stimulates NK cells to localize next to foci of malignant cells.Citation15 Both the trafficking to neoplastic sites and in vivo antitumor activity of T cells modified to recognize a peptide derived from Wilms’ tumor 1 (WT-1, a TAA frequently expressed by pulmonary cancers) in a HLA-A24-restricted fashion were improved when these cells were engineered to express chemokine (C-C motif) receptor 2 (CCR2), which recognizes a chemokine that is highly expressed in the lung (i.e., chemokine (C-C motif) ligand 2, CCL2).Citation16

The CARs used by Boissel et al. to engineer allogeneic NK cells did not comprise the signaling domain of co-stimulatory molecules. As domains of this type have been shown to promote the persistence of adoptively transfer effector cells in vivo, they may represent a means to improve the antineoplastic activity of CAR-expressing NK cells.Citation4,Citation5,Citation12,Citation17 Allogeneic NK cells may actually represent a weakness as compared with their autologous counterparts, because they may be rapidly rejected. However, Boissel et al. provided evidence for the expansion of allogeneic NK-92 cells in vivo.Citation6 In addition, allogeneic NK cells have previously been shown to be efficient for the therapy of acute myeloid leukemia.Citation18 The proof-of-concept for this allogeneic, CAR-based immunotherapeutic approach has been recently provide in patients affected by B-cell malignancies, as donor-derived allogeneic T cells engineered to express CD19-specific CARs were shown to induce disease regression in individuals that were insensitive to conventional donor lymphocyte infusion (DLIs) upon allogeneic hematopoietic stem cell transplantation, and were not associated with no signs of graft-vs.-host disease.Citation19 The good safety profile of CAR-expressing allogeneic NK cells in patients further supports the clinical development of this immunotherapeutic approach.Citation20

Disclosure of Potential Conflicts of Interest

No potential conflicts of interest were disclosed.

Acknowledgments

This work was supported by grants from Ligue contre le Cancer, Agence Nationale de la Recherche (ANR), Labex Immuno-Oncology, Institut National du Cancer (Carpem project), Canceropole-Region Ile de France.

Citation: An allogeneic NK cell line engineered to express chimeric antigen receptors: A novel strategy of cellular immunotherapy against cancer. OncoImmunology 2013; 2:e27156; 10.4161/onci.27156

References

  • Galluzzi L, Vacchelli E, Eggermont A, Fridman WH, Galon J, Sautès-Fridman C, Tartour E, Zitvogel L, Kroemer G. Trial Watch: Adoptive cell transfer immunotherapy. Oncoimmunology 2012; 1:306 - 15; http://dx.doi.org/10.4161/onci.19549; PMID: 22737606
  • Vacchelli E, Eggermont A, Fridman WH, Galon J, Tartour E, Zitvogel L, Kroemer G, Galluzzi L. Trial Watch: Adoptive cell transfer for anticancer immunotherapy. Oncoimmunology 2013; 2:e24238; http://dx.doi.org/10.4161/onci.24238; PMID: 23762803
  • Stauss HJ, Morris EC. Immunotherapy with gene-modified T cells: limiting side effects provides new challenges. Gene Ther 2013; 20:1029 - 32; http://dx.doi.org/10.1038/gt.2013.34; PMID: 23804078
  • Grupp SA, Kalos M, Barrett D, Aplenc R, Porter DL, Rheingold SR, Teachey DT, Chew A, Hauck B, Wright JF, et al. Chimeric antigen receptor-modified T cells for acute lymphoid leukemia. N Engl J Med 2013; 368:1509 - 18; http://dx.doi.org/10.1056/NEJMoa1215134; PMID: 23527958
  • Brentjens RJ, Davila ML, Riviere I, Park J, Wang X, Cowell LG, Bartido S, Stefanski J, Taylor C, Olszewska M, et al. CD19-targeted T cells rapidly induce molecular remissions in adults with chemotherapy-refractory acute lymphoblastic leukemia. Sci Transl Med 2013; 5:177ra38; http://dx.doi.org/10.1126/scitranslmed.3005930; PMID: 23515080
  • Boissel L, Betancur M, Lu W, Krause D, Van Etten R, Wels W, et al.. Retargeting NK-92 cells by means of CD19- and CD20-specific CARs compares favorably with antibody-dependent cellular cytotoxicity. OncoImmunology 2013;; Forthcoming
  • Terme M, Fridman WH, Tartour E. NK cells from pleural effusions are potent antitumor effector cells. Eur J Immunol 2013; 43:331 - 4; http://dx.doi.org/10.1002/eji.201243264; PMID: 23322344
  • Vivier E, Raulet DH, Moretta A, Caligiuri MA, Zitvogel L, Lanier LL, Yokoyama WM, Ugolini S. Innate or adaptive immunity? The example of natural killer cells. Science 2011; 331:44 - 9; http://dx.doi.org/10.1126/science.1198687; PMID: 21212348
  • Tartour E, Mathiot C, Fridman WH. Current status of interleukin-2 therapy in cancer. Biomed Pharmacother 1992; 46:473 - 84; http://dx.doi.org/10.1016/0753-3322(92)90005-R; PMID: 1306361
  • Turatti F, Figini M, Balladore E, Alberti P, Casalini P, Marks JD, Canevari S, Mezzanzanica D. Redirected activity of human antitumor chimeric immune receptors is governed by antigen and receptor expression levels and affinity of interaction. J Immunother 2007; 30:684 - 93; http://dx.doi.org/10.1097/CJI.0b013e3180de5d90; PMID: 17893561
  • Tassev DV, Cheng M, Cheung NK. Retargeting NK92 cells using an HLA-A2-restricted, EBNA3C-specific chimeric antigen receptor. Cancer Gene Ther 2012; 19:84 - 100; http://dx.doi.org/10.1038/cgt.2011.66; PMID: 21979579
  • Porter DL, Levine BL, Kalos M, Bagg A, June CH. Chimeric antigen receptor-modified T cells in chronic lymphoid leukemia. N Engl J Med 2011; 365:725 - 33; http://dx.doi.org/10.1056/NEJMoa1103849; PMID: 21830940
  • DeNucci CC, Mitchell JS, Shimizu Y. Integrin function in T-cell homing to lymphoid and nonlymphoid sites: getting there and staying there. Crit Rev Immunol 2009; 29:87 - 109; http://dx.doi.org/10.1615/CritRevImmunol.v29.i2.10; PMID: 19496742
  • Sandoval F, Terme M, Nizard M, Badoual C, Bureau MF, Freyburger L, Clement O, Marcheteau E, Gey A, Fraisse G, et al. Mucosal imprinting of vaccine-induced CD8⁺ T cells is crucial to inhibit the growth of mucosal tumors. Sci Transl Med 2013; 5:172ra20; http://dx.doi.org/10.1126/scitranslmed.3004888; PMID: 23408053
  • Rusakiewicz S, Semeraro M, Sarabi M, Desbois M, Locher C, Mendez R, Vimond N, Concha A, Garrido F, Isambert N, et al. Immune infiltrates are prognostic factors in localized gastrointestinal stromal tumors. Cancer Res 2013; 73:3499 - 510; http://dx.doi.org/10.1158/0008-5472.CAN-13-0371; PMID: 23592754
  • Asai H, Fujiwara H, An J, Ochi T, Miyazaki Y, Nagai K, Okamoto S, Mineno J, Kuzushima K, Shiku H, et al. Co-introduced functional CCR2 potentiates in vivo anti-lung cancer functionality mediated by T cells double gene-modified to express WT1-specific T-cell receptor. PLoS One 2013; 8:e56820; http://dx.doi.org/10.1371/journal.pone.0056820; PMID: 23441216
  • Milone MC, Fish JD, Carpenito C, Carroll RG, Binder GK, Teachey D, Samanta M, Lakhal M, Gloss B, Danet-Desnoyers G, et al. Chimeric receptors containing CD137 signal transduction domains mediate enhanced survival of T cells and increased antileukemic efficacy in vivo. Mol Ther 2009; 17:1453 - 64; http://dx.doi.org/10.1038/mt.2009.83; PMID: 19384291
  • Velardi A, Ruggeri L, Mancusi A, Aversa F, Christiansen FT. Natural killer cell allorecognition of missing self in allogeneic hematopoietic transplantation: a tool for immunotherapy of leukemia. Curr Opin Immunol 2009; 21:525 - 30; http://dx.doi.org/10.1016/j.coi.2009.07.015; PMID: 19717293
  • Kochenderfer JN, Dudley ME, Carpenter RO, Kassim SH, Rose JJ, Telford WG, Hakim FT, Halverson DC, Fowler DH, Hardy NM, et al. Donor-derived CD19-targeted T cells cause regression of malignancy persisting after allogeneic hematopoietic stem cell transplantation. Blood 2013; Forthcoming http://dx.doi.org/10.1182/blood-2013-08-519413; PMID: 24055823
  • Arai S, Meagher R, Swearingen M, Myint H, Rich E, Martinson J, Klingemann H. Infusion of the allogeneic cell line NK-92 in patients with advanced renal cell cancer or melanoma: a phase I trial. Cytotherapy 2008; 10:625 - 32; http://dx.doi.org/10.1080/14653240802301872; PMID: 18836917