Skip to main content
Oncoimmunology logoLink to Oncoimmunology
. 2014 Apr 15;3:e28147. doi: 10.4161/onci.28147

Are natural killer cells superior CAR drivers?

Hans Klingemann 1,2,*
PMCID: PMC4203506  PMID: 25340009

Abstract

T lymphocytes engineered to express a chimeric antigen receptor (CAR) are being celebrated as a major breakthrough of anticancer immunotherapy. Natural killer cells have not received similar attention as CAR effectors, although the use of these relatively short-lived cytotoxic cells is associated with several advantages.

Keywords: CAR, NK cells, NK-92 cells, T lymphocytes, cancer, on-target/off-tumor effect


The latter American Society of Hematology (ASH) meeting, which took place last December in New Orleans, was dominated by the enthusiasm on the anti-leukemia effects of T lymphocytes engineered to express a CD19-targeting chimeric antigen receptor (CAR).1 Fourteen out of 16 pediatric patients with relapsed or advanced acute lymphoblastic leukemia (ALL) entered a remission in response to the adoptive transfer of these cells. A similar outcome was documented for adults with chronic lymphocytic leukemia.

CAR-expressing T cells are usually generated from autologous T cells, but T lymphocytes from allogeneic donors are also being explored in this sense, especially upon relapse after stem cell transplantation.2 CAR-bearing T cells are usually activated with anti-CD3/CD28 beads and expanded in culture flasks (such as the WaveR system) in the presence of interleukin (IL)-2. CARs against an expanding array of cell surface-exposed tumor-associated antigens (TAAs) have been and continue to be engineered.3 Since the majority of these TAAs are not tumor specific, CAR-expressing T cells can cross-react with healthy cells, mediating an “on-target/off-tumor” side effect. For example, T cells expressing a CD19-targeting CAR can cause a profound and long-lasting B-cell deficiency as they eliminate normal B cells.4 T lymphocytes bearing a CAR specific for interleukin 3 receptor, α (ILR3A, also known as CD123) kill not only leukemic cells but also bone marrow cells that express the same receptor, leading to prolonged and profound marrow suppression.5 In some cases this on-target/off-tumor side effect can be fatal, as it happened in a patient with metastatic colon carcinoma who received T cells engineered to express a HER2 targeting CAR. In this case, the side effects of CAR-expressing T cells on low level HER2 expressing lung epithelium led to fatal pulmonary complications combined with a massive cytokine release.

It has been suggested that the antineoplastic activity of CAR-expressing T cells is related to and dependent on their persistence in the patient circulation and malignant tissue. If this were indeed the case, the on-target/off-tumor effects would also persist. For CD19-redirecetd T cells, this would entail a prolonged depletion of normal B cells and hence long-term defects in humoral immunity.

As recent clinical trials have suggested, antigen loss cancer variants can emerge as a result of the selective pressure imposed by immunotherapeutic interventions, often driving disease relapse.1 In this setting, TAA-specific T cells would continue to mediate on-target/off tumor effects, such as the suppression of normal B cells or bone marrow precursors. A potential solution to this issue is provided by the transduction of T cells with CAR-coding mRNAs, usually resulting in the loss of expression over a few days.7 Indeed, most CAR-expressing T cells currently tested in clinical trials are obtained with lentiviral constructs, which integrate into the genome and hence ensure persistent transgene expression.

Natural killer (NK) cells may represent alternative cytotoxic effectors for CAR-driven cytolysis. Allogeneic NK cells are expected to induce an immune response and be rejected after a few days, and even autologous NK cells should disappear relatively rapidly from the circulation, owing to their limited lifespan. NK cells have additional advantages over T cells (Table 1). In particular, while T lymphocytes only kill their targets by a CAR-specific mechanism, NK cells are endowed with spontaneous cytotoxic activity and can trigger the demise of target cells in a TAA-unrestricted manner via specific natural cytotoxicity receptors (NCRs), including NCR3 (also known as NKp30), NCR2 (also known as NKp44), NCR1 (also known as NKp46), and killer cell lectin-like receptor subfamily K, member 1 (KLRK1, best known as NKG2D). NK cells also express the Fc fragment of IgG, low affinity III, receptor (FcγRIII), that binds the Fc fragment of antibodies to elicit antibody-dependent cell-mediated cytotoxicity (ADCC). This specific feature of NK cells would enable the combination of 2 targeted therapies recognizing different (or the same) TAA(s), namely CAR-expressing NK cells and a TAA-specific monoclonal antibody.

Table 1. Comparison of CAR- expressing T, natural killer and NK-92 cells.

Parameter T cells NK cells NK-92 cells
Collection Leukopheresis Leukopheresis Continuously growing cell line consisting of “pure” (100%) activated NK cells
Preparation Activation of cells with anti-CD3/CD28 beads
Allogeneic donor: MHC match required
NK cells represent only 10% of all lymphocytes.
Autologous: Enrichment needed (selection for CD56+ cells).
Allogeneic donor: MHC-matched donor or depletion of alloreactive T-cells to prevent GvH reactions
No processing necessary prior to CAR engineering
Expansion Flasks, bags or WaveR expansion system Requires engineered feeders (example: K562 cells expressing IL-15 and TNFSF9) plus IL-2 (in flasks, bags or bioreactors) Expansion in serum free-medium without feeders, but IL-2 only (in flasks, bags or bioreactors)
Transduction Lentiviral systems transduce about 1/3 of T cells Low transfection efficiency even with viral vectors Transfection efficiency of about 50%, compatible with sorting
Cytotoxic mechanisms CAR-restricted killing
In case of antigen loss on tumors, CAR-expressing T cells become ineffective
Multiple receptors can trigger CAR-independent and FcR-dependent cytotoxicity Multiple receptors can trigger CAR-independent and FcR-dependent cytotoxicity
Adverse events Can cause “off target” effects
Survive for prolonged periods in the patient circulation
Can induce a storm of pro-inflammatory cytokines
Limited life span in patients
No concern about persisting CAR-associated side effects
Limited life span in patients
No concern about persisting CAR-associated side effects
Miscellaneous Suicide genes are required to control life span in vivo No need for suicide gene No need for suicide gene
Clinical results Phase I studies have shown clinical benefit Proof of clinical benefit pending Proof of clinical benefit pending
Off-the-shelf CAR-specific cellular product? Autologous cells, required preparation on a per patient basis Possible to have donor NK cells cryopreserved, but recovery is poor after upon thawing Possible to have NK-92 cryopreserved and expanded upon thawing (before infusion)

Abbreviations: CAR, chimeric antigen receptor; FcR, Fc receptor; GvH, graft-vs.-host; IL, interleukin; NK, natural killer; TNFSF9, tumor necrosis factor superfamily, member 9.

Additional features of NK cells could make them better and potentially safer CAR drivers than T cells. For instance, NK cells produce a host of cytokines that are different from those produced by T cells, including interferon γ (IFNγ) and granulocyte macrophage colony-stimulating factor (GM-CSF). The cytokine storm initiated by the infusion of CAR-expressing T cells is indeed largely mediated by their pro-inflammatory cytokines such as tumor necrosis factor α (TNFα), IL-1, and IL-6. It is also known that NK cells are “serial killers.” Thus, time-lapse videomicroscopy studies have shown that NK as well as NK-92 cells (a continuously-growing, highly-active, NK cell-derived cell line) diligently move from one target to the next one, killing on as many as 7–10 cells.8 Evidence for such a serial killing by T cells is lacking at this point.

Nonetheless, there are some obstacles for the use of circulating NK cells for CAR-based immunotherapy. Like T lymphocytes, NK cells are obtained (from patients or donors) by leukopheresis, which can be time-consuming, costly and occasionally requiring central venous access. Since only about 10% of circulating lymphocytes are NK cells, some extent of selection/enrichment is necessary, which is generally performed by positive CD56-based, magnetic immunoselection. In case of allogeneic donors, peripheral blood mononuclear cells also must be depleted of T cells to prevent graft-vs.-host reactions. Moreover, a feeder layer and additional cytokines are required to maximize the expansion of circulating NK cells in vitro. A feeder layer consisting of the leukemia K562 cells engineered to express tumor necrosis factor superfamily, member 9 (TNFSF9, best known as 4–1BBL) and IL-15 seems to be very effective, but a product testing before infusion must ensure that all malignant cells have been completely removed.9 Finally, the transfection efficiency of circulating NK cells is variable and generally not very high, even when viral vectors are employed.10

Although the challenge of introducing CAR-coding genes into sufficient numbers of circulating NK cells may be overcome at some point, NK-92 cells present an open cellular platform for CAR-based immunotherapy.11 The transfection efficiency of NK-92 cells is about 50%, even with non-viral methods.10 Besides being technically more simple and under less-constraining regulations, avoiding viral vectors eliminates the risks of oncogene activation and insertional mutagenesis. Upon sorting, CAR-expressing NK-92 cells can be enriched to obtain a population near-to-exclusively composed of NK-92 carrying the CAR of interest.

So far, NK-92 cells have been efficiently transduced with a number of different CAR-coding constructs (Table 2) and pre-clinical studies in xenotransplanted immunodeficient mice have demonstrated the potential therapeutic effects of this approach. Several centers are gearing up to test whether CAR-expressing NK cells can keep up with their T-cell counterparts. Eventually, we might even discover that both these cell types have their place in the multimodal approach that is required to eliminate cancer and control its recurrence.

Table 2. CAR-coding genes transfected/transduced so far into natural killer cells.

Target Indication(s) Blood NK cells NK-92 cells Refs.
CD19 Lymphoid malignancies X X 10,1215
CD20 Lymphoid malignancies X X 1618
CD38 Multiple myeloma   X 19
ERBB2 Breast carcinoma
Head and neck cancer
Ovarian carcinoma
Glioblastoma
X X 11,20
GD2 Neuroblastoma   X 21
EPCAM Breast carcinoma
Pancreatic cancer
  X 22
EBNA3C EBV infections   X 23
CS1 Multiple myeloma   X 24
LMAN1 Melanoma
Neuroblastoma
  X 25

Abbreviations: EBNA3C, Epstein-Barr nuclear antigen 3C; ERBB2, v-erb-b2 avian erythroblastic leukemia viral oncogene homolog 2; EPCAM, epithelial cell adhesion molecule; GD2, ganglioside GD2; LMAN1, lectin, mannose-binding, 1

Disclosure of Potential Conflicts of Interest

H.K. is Founder of Conkwest Inc. and its Chief Medical and Scientific Officer

Citation: Klingemann H. Are natural killer cells superior CAR drivers?. OncoImmunology 2014; 3:e28147; 10.4161/onci.28147

References

  • 1.Grupp SA, Frey NV, Aplenc R, Barrett DM, Chew A, Kalos M, Levine BL, Litchman M, Maude SL, Rheingold SR, et al. T Cells engineered with a Chimeric Antigen Receptor (CAR) targeting CD19 (CTL019) produce significant in vivo proliferation, complete responses and long-term persistence without GvHd in children and adults with relapsed, refractory ALL. Blood. 2013;122:67. [Google Scholar]
  • 2.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;122:4129–39. doi: 10.1182/blood-2013-08-519413. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Sadelain M, Brentjens R, Rivière I. The promise and potential pitfalls of chimeric antigen receptors. Curr Opin Immunol. 2009;21:215–23. doi: 10.1016/j.coi.2009.02.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Kalos M, Levine BL, Porter DL, Katz S, Grupp SA, Bagg A, June CH. T cells with chimeric antigen receptors have potent antitumor effects and can establish memory in patients with advanced leukemia. Sci Transl Med. 2011;3:95ra73. doi: 10.1126/scitranslmed.3002842. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Gill S, Tasian SK, Ruella M, Shestova O, Li Y, Porter DL, Carroll M, Danet-Desnoyers G, Scholler J, Grupp SA, et al Anti-CD123 Chimeric Antigen Receptor T cells (CART-123) provide a novel myeloablative conditioning regimen that eradicates human acute myeloid leukemia in preclinical models. Blood. 2013;122:143. doi: 10.1182/blood-2013-09-529537. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Morgan RA, Yang JC, Kitano M, Dudley ME, Laurencot CM, Rosenberg SA. Case report of a serious adverse event following the administration of T cells transduced with a chimeric antigen receptor recognizing ERBB2. Mol Ther. 2010;18:843–51. doi: 10.1038/mt.2010.24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Zhao Y, Moon E, Carpenito C, Paulos CM, Liu X, Brennan AL, Chew A, Carroll RG, Scholler J, Levine BL, et al. Multiple injections of electroporated autologous T cells expressing a chimeric antigen receptor mediate regression of human disseminated tumor. Cancer Res. 2010;70:9053–61. doi: 10.1158/0008-5472.CAN-10-2880. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Bhat R, Watzl C. Serial killing of tumor cells by human natural killer cells--enhancement by therapeutic antibodies. PLoS One. 2007;2:e326. doi: 10.1371/journal.pone.0000326. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.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;69:4010–7. doi: 10.1158/0008-5472.CAN-08-3712. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Boissel L, Betancur M, Wels WS, Tuncer H, Klingemann H. Transfection with mRNA for CD19 specific chimeric antigen receptor restores NK cell mediated killing of CLL cells. Leuk Res. 2009;33:1255–9. doi: 10.1016/j.leukres.2008.11.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Uherek C, Tonn T, Uherek B, Becker S, Schnierle B, Klingemann HG, Wels W. Retargeting of natural killer-cell cytolytic activity to ErbB2-expressing cancer cells results in efficient and selective tumor cell destruction. Blood. 2002;100:1265–73. [PubMed] [Google Scholar]
  • 12.Li L, Liu LN, Feller S, Allen C, Shivakumar R, Fratantoni J, Wolfraim LA, Fujisaki H, Campana D, Chopas N, et al. Expression of chimeric antigen receptors in natural killer cells with a regulatory-compliant non-viral method. Cancer Gene Ther. 2010;17:147–54. doi: 10.1038/cgt.2009.61. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Imai C, Mihara K, Andreansky M, Nicholson IC, Pui C-H, Geiger TL, Campana D. Chimeric receptors with 4-1BB signaling capacity provoke potent cytotoxicity against acute lymphoblastic leukemia. Leukemia. 2004;18:676–84. doi: 10.1038/sj.leu.2403302. [DOI] [PubMed] [Google Scholar]
  • 14.Shimasaki N, Fujisaki H, Cho D, Masselli M, Lockey T, Eldridge P, Leung W, Campana D. A clinically adaptable method to enhance the cytotoxicity of natural killer cells against B-cell malignancies. Cytotherapy. 2012;14:830–40. doi: 10.3109/14653249.2012.671519. [DOI] [PubMed] [Google Scholar]
  • 15.Romanski A, Uherek C, Bug G, Muller T, Rossig C, Kampfmann M, Krossok N, Hoelzer D, Seifried E, Wels W, et al Re-targeting of an NK cell line (NK-92) with specificity for CD19 efficiently kills human B-precursor leukemic cells. Blood. 2004;104:751a. [Google Scholar]
  • 16.Chu Y, Yahr A, Ayello J, van de Ven C, Zhou X, Cairo MS. Anti-CD20 Chimeric Antigen Receptor (CAR) modified expanded Natural Killer (NK) cells significantly mediate Burkitt Lymphoma (BL) regression and improve survival in human BL xenografted NSG mice. Blood. 2013;122:3263. [Google Scholar]
  • 17.Boissel L, Betancur-Boissel M, Lu W, Krause DS, Van Etten RA, Wels WS, Klingemann H. Retargeting NK-92 cells by means of CD19- and CD20-specific chimeric antigen receptors compares favorably with antibody-dependent cellular cytotoxicity. Oncoimmunology. 2013;2:e26527. doi: 10.4161/onci.26527. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Müller T, Uherek C, Maki G, Chow KU, Schimpf A, Klingemann HG, Tonn T, Wels WS. Expression of a CD20-specific chimeric antigen receptor enhances cytotoxic activity of NK cells and overcomes NK-resistance of lymphoma and leukemia cells. Cancer Immunol Immunother. 2008;57:411–23. doi: 10.1007/s00262-007-0383-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Yang S, Xin A, Brown RD, Ho J, Gibson J, Joshua DE, Wels W, Sze DM. Development of retargeted CD38-specific NK-92 cell line for potential anti-myeloma immunotherapy. Blood. 2005;106:5104. [abstr] [Google Scholar]
  • 20.Kruschinski A, Moosmann A, Poschke I, Norell H, Chmielewski M, Seliger B, Kiessling R, Blankenstein T, Abken H, Charo J. Engineering antigen-specific primary human NK cells against HER-2 positive carcinomas. Proc Natl Acad Sci U S A. 2008;105:17481–6. doi: 10.1073/pnas.0804788105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Esser R, Müller T, Stefes D, Kloess S, Seidel D, Gillies SD, Aperlo-Iffland C, Huston JS, Uherek C, Schönfeld K, et al. NK cells engineered to express a GD2 -specific antigen receptor display built-in ADCC-like activity against tumour cells of neuroectodermal origin. J Cell Mol Med. 2012;16:569–81. doi: 10.1111/j.1582-4934.2011.01343.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Sahm C, Schönfeld K, Wels WS. Expression of IL-15 in NK cells results in rapid enrichment and selective cytotoxicity of gene-modified effectors that carry a tumor-specific antigen receptor. Cancer Immunol Immunother. 2012;61:1451–61. doi: 10.1007/s00262-012-1212-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.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. doi: 10.1038/cgt.2011.66. [DOI] [PubMed] [Google Scholar]
  • 24.Chu J, Deng Y, Benson DM, He S, Hughes T, Zhang J, Peng Y, Mao H, Yi L, Ghoshal K, et al. CS1-specific chimeric antigen receptor (CAR)-engineered natural killer cells enhance in vitro and in vivo antitumor activity against human multiple myeloma. Leukemia. 2013 doi: 10.1038/leu.2013.279. Forthcoming. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Koehne G, Guo HF, Trivedi D, Williams RY, O’Reilly RO, Cheung V. Redirecting NK-cell cytolytic activity to solid tumors using chimeric scFv receptor gene-modified adoptive immunotherapy. ASCO Proc 2001; 22: 175a [Google Scholar]

Articles from Oncoimmunology are provided here courtesy of Taylor & Francis

RESOURCES