Infusions of Allogeneic Natural Killer Cells as Cancer Therapy Wing Leung Clin Cancer Res 2014;20:3390-3400.

Updated version

Cited Articles

E-mail alerts Reprints and Subscriptions Permissions

Access the most recent version of this article at: http://clincancerres.aacrjournals.org/content/20/13/3390

This article cites by 96 articles, 44 of which you can access for free at: http://clincancerres.aacrjournals.org/content/20/13/3390.full.html#ref-list-1

Sign up to receive free email-alerts related to this article or journal. To order reprints of this article or to subscribe to the journal, contact the AACR Publications Department at [email protected]. To request permission to re-use all or part of this article, contact the AACR Publications Department at [email protected].

Downloaded from clincancerres.aacrjournals.org on July 17, 2014. © 2014 American Association for Cancer Research.

Clinical Cancer Research

Review

Infusions of Allogeneic Natural Killer Cells as Cancer Therapy Wing Leung1,2

Abstract Natural killer (NK) cells are normal white blood cells capable of killing malignant cells without prior sensitization. Allogeneic NK cell infusions are attractive for cancer therapy because of non–cross-resistant mechanisms of action and minimal overlapping toxicities with standard cancer treatments. Although NK therapy is promising, many obstacles will need to be overcome, including insufficient cell numbers, failure of homing to tumor sites, effector dysfunction, exhaustion, and tumor cell evasion. Capitalizing on the wealth of knowledge generated by recent NK cell biology studies and the advancements in biotechnology, substantial progress has been made recently in improving therapeutic efficiency and reducing side effects. A multipronged strategy is essential, including immunogenetic-based donor selection, refined NK cell bioprocessing, and novel augmentation techniques, to improve NK function and to reduce tumor resistance. Although data from clinical trials are currently limited primarily to hematologic malignancies, broader applications to a wide spectrum of adult and pediatric cancers are under way. The unique properties of human NK cells open up a new arena of novel cell-based immunotherapy against cancers that are resistant to contemporary therapies. Clin Cancer Res; 20(13); 3390–400. 2014 AACR.

Disclosure of Potential Conflicts of Interest W. Leung is a co-inventor of a patent-pending diagnostic method assigned to St. Jude Children’s Research Hospital and licensed to Insight Genetics.

CME Staff Planners' Disclosures The members of the planning committee have no real or apparent conflict of interest to disclose.

Learning Objectives Upon completion of this activity, the participant should have a better understanding of the biology of human natural killer (NK) cells relevant to cancer immunotherapy, the general steps of allogeneic NK-based treatments, and the avenues of current and future strategies being explored in NK cell therapy.

Acknowledgment of Financial or Other Support This activity does not receive commercial support.

Introduction For many cancers, cure rates remain unacceptably low. Advances in genetics and immunology have led to biologic therapies with non–cross-resistant mechanisms of action and no overlapping toxicities. Cancer immunotherapy is possible with an array of effector mechanisms involving innate and adaptive immunity (1). Various cytokines, antibodies, and cellular therapies have been FDA approved or are in late-phase clinical trials. Author's Affiliations: 1Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children's Research Hospital; and 2Department of Pediatrics, University of Tennessee, Memphis, Tennessee Corresponding Author: Wing Leung, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105-3678. Phone: 901595-2617; Fax: 901-521-9005; E-mail: [email protected] doi: 10.1158/1078-0432.CCR-13-1766 2014 American Association for Cancer Research.

3390

Infusions of purified natural killer (NK) cells are latecomers in cellular immunotherapy, compared with dendritic cells (DC), lymphokine-activated killer (LAK) cells, tumor-infiltrating lymphocytes, and conventional cytotoxic T cells. Many investigators consider that the cytolytic activity of LAK cells commonly used in the 1980s actually represents the activity of IL2-activated NK cells, although the products contained polyclonal T cells. Biologically, murine NK cells have been known to kill malignant cells without T-cell assistance or prior sensitization since their initial description in 1975 (2–5); however, the therapeutic potential of human NK cells was not revealed until their biology of target cell recognition was elucidated in the 1990s and the antileukemia effect was observed in HLAhaploidentical hematopoietic cell transplantation (HCT) in the early 2000s (6, 7). This review covers NK cell biology relevant to cancer immunotherapy and outlines avenues being explored in clinical trials.

Clin Cancer Res; 20(13) July 1, 2014

Downloaded from clincancerres.aacrjournals.org on July 17, 2014. © 2014 American Association for Cancer Research.

NK Infusions as Cancer Therapy

NK Cell Biology NK cells were first named and characterized by Kiessling at the Karolinska Institute (Stockholm, Sweden) and by Herberman at the National Cancer Institute (Bethesda, MD; refs. 2–5). The term "natural" referred to their natural existence in rodents and "killer" to their spontaneous killing of lymphoma and leukemia cells in nonimmune animals. They were called "N-cells" or "K cells" by some investigators (3, 8). These cells were unique, as they lacked markers characteristic of B and T lymphocytes and were nonphagocytic, low-adhesive, and cytotoxic to IgG-coated targets. NK cells are generated in bone marrow and develop normally in athymic mice and in humans with DiGeorge syndrome. Induction of cytotoxicity is possible in NK cells within minutes, in contrast with the slower conventional induction of the T-cell immune response (9). Approximately 5% to 15% of human blood lymphocytes are NK cells (CD56þ, CD3/14/19), 90% of which are CD56dim, with most of them being CD16þ, and they are responsible for early innate immunity and antibodydependent cell-mediated cytotoxicity (ADCC) against infection or cancer through IFNg, perforin, granzyme, FasL, or TRAIL (10). Approximately 10% of human blood NK cells are CD56bright, and they participate in late (>16 hours) inflammatory response by secreting IFNg, TNFa, G-CSF, GM-CSF, and IL3. Interactions between DCs and NK cells are essential for priming adaptive immunity. NK cells may also directly interact with T and B cells through CD40 ligand and OX40 (11, 12). Regulatory receptors Human NK cells are regulated by a sophisticated network of surface receptors (Fig. 1), allowing them to distinguish normal from abnormal cells within seconds. Target cell lysis occurs when the activating signal dominates the inhibitory signal (Fig. 2). Early studies revealed similarities between murine NK cells and cells responsible for so-called "hybrid resistance" to parental hematopoietic grafts (13, 14). The proposal that the resistance was related to receptors recognizing the absence of MHC class I arose from observations that murine lymphoma cells that were low in MHC expression were NK-susceptible (15). This led to the "missing-self" hypothesis in the 1980s (16). Breakthroughs in human NK receptor biology were seen in the early 1990s, when the KIR gene family was found to recognize HLA class I (17–19). Identification of other NK receptors, such as NCRs, NKG2D, DNAM, 2B4, and CD94/ NKG2A, collectively points to a sophisticated network that controls human NK activity.

Step 1: KIR Typing for Donor Selection and Prognostication Three general steps in NK-based therapy are summarized in Fig. 3. KIR typing is a critical first step because KIR is highly polymorphic. KIR typing is not only useful for allogeneic donor selection but also for prognostication in

www.aacrjournals.org

autologous NK therapy (20). Among known NK receptors, the KIR receptor family is one of the primary determinants of NK response. Subsets of T cells also expressed KIR (21). Clinical KIR typing includes genotyping for gene content and A/B haplotype categorization, phenotyping for gene expression and number of KIRþ cells, and allelotyping for functional strength. The first level of KIR diversity is gene content (22, 23). Only approximately 5% of people have all 15 gene family members; others lack one or more genes. The diversity of gene content is based primarily on diversity in B haplotypes, which typically contain more activating KIRs. The two hallmark genes for A haplotypes are KIR2DL3 and KIR3DL1. They are segregated with KIR2DL2 and KIR3DS1 as alleles in the centromeric (Cen) and telomeric (Tel) motifs, respectively (Fig. 4A). On the basis of this pattern, a simplified typing and scoring method for B haplotype content can be derived (Fig. 4B), viz., KIR2DL3þ/ KIR2DL2, Cen-A/A; KIR2DL3þ/KIR2DL2þ, Cen-A/B; KIR2DL3/KIR2DL2þ, Cen-B/B; KIR3DL1þ/KIR3DS1, Tel-A/A; KIR3DL1þ/KIR3DS1þ, Tel-A/B; and KIR3DL1/ KIR3DS1þ, Tel-B/B. Because KIRs are encoded in chromosome 19 and ligand HLAs are in chromosome 6, these gene families are segregated independently; thus, HLA genotypes cannot be used to predict KIR content, and autologous KIR-HLA mismatch is possible. The second level of diversity is gene expression (24). By real-time PCR and flow cytometry, more than 10-fold variability in expression has been observed among donors. Nowadays, the easiest method to quantify NK cells expressing only one inhibitory KIR gene (i.e., negative for all other MHC inhibitory receptors) is multicolor flow cytometry (24–26). The number of single KIRþ cells is in direct proportion to activity against target cells without the ligand (receptor–ligand mismatched). Although the number of single KIRþ cells correlates with the presence of selfligand (25), the variability among donors renders predictions based on the ligand alone unacceptable for clinical use. The third level of diversity is allele polymorphism, which has been observed in all inhibitory KIR genes (22, 23). For example, in the KIR2DL1 family, 25 alleles have been observed, and each has different strengths in inhibiting NK cells and different durability of surface expression after interaction with ligands (27). These differences correlate with the intensity of inhibitory signaling through SHP2 and b arrestin-2, resulting in differences in immune synapse formation. Mechanistically, the arginine residue at position 245 in the transmembrane domain is important in inhibition. Alleles with arginine in position 245 are stronger than those with cysteine in that position (27). On the basis of this molecular determinant, single-nucleotide polymorphism assays have been developed for rapid, high-throughput clinical typing (28). The same assay can be simultaneously used for KIR ligand typing. Notably, this assay requires less than 0.1 mg of DNA and can be performed within 1 day. Using this assay, investigations showed that patients who received a donor graft containing the stronger KIR allele had

Clin Cancer Res; 20(13) July 1, 2014

Downloaded from clincancerres.aacrjournals.org on July 17, 2014. © 2014 American Association for Cancer Research.

3391

Leung

IFNα

IL1 IL15

IL2 IL18

IL12 IL21

NK cell KIR2DL1/2/3 (HLA-C)

KIR2DS1/S2 (HLA-C)

KIR3DL1 (HLA-B)

KIR2DS4 (HLA-A, -C)

KIR3DL2 (HLA-A)

KIR2DL4 (HLA-G, HS)

CD94/NKG2A (HLA-E)

Inhibitory

Activating

LIR-1 (HLA-A–G)

CD94/NKG2C (HLA-E) KIR3DL2 (CpG)

KLRG-1 (cadherins)

CD16 (IgG)

CEACAM1 (CEACAM5)

NKG2D (MIC, ULBP) NCRs (B7-H6, NKp44L)

TIGIT (PVR, PVRL2) Coreceptor

DNAM-1 (PVR, Nectin-2) Siglec –3, –7, –9 (sialic acid) LAIR-1 (collagen) 2B4 (CD48) CD300A (PS) NKp80 NTBA (NTBA) (AICL)

Figure 1. Surface receptors and their ligands. Cytokine receptors are shown on top of a human NK cell. Other receptors are broadly classified and color-coded on the basis of their primary function (inhibitory receptors in red, activating receptors in green, inhibitory coreceptors in red-black stripes, and activating coreceptors in green-black stripes). Their ligands are shown within parentheses. Many other known receptors are not shown, including chemotactic receptors (CCR-2, -5, -7: CXCR-1, -3, -4, -6; CX3CR1; and Chem23R), adhesion receptors (CD2 and b1 and b2 integrins), and activating coreceptors (CD96, CS1, and TLR).

© 2014 American Association for Cancer Research CCR Reviews

fewer relapses, better survival, and less transplant-related mortality (29). These effects were observed regardless of primary disease [acute myelogenous leukemia (AML) vs.

NK cell A

Target cell

acute lymphoblastic leukemia (ALL)], total body irradiation, T-cell depletion, and donor type. Statistically, there is an interaction effect with HLA-C receptor–ligand mismatch:

Characteristics of target cell

Outcome

Normal MHC class I No activating ligands

No response

No MHC class I No activating ligands

No response

No MHC class I activating ligands

Attack target

Normal MHC class I activating ligands

Dynamic equilibrium

– +

B

C

D

© 2014 American Association for Cancer Research

Figure 2. Dynamic equilibrium. After cell-to-cell contact, NK cell integrates signals from its surface receptors in seconds, resulting in either target-cell attack or no response and continual immunosurveillance of other cells. A, healthy cells express normal amount of MHC class I ligands with no activating "stress" ligands. B, downregulation or absence of MHC ligand for cognate inhibitory receptors is insufficient to trigger NK cells. This happens in the physiologic setting with red blood cells and autologous KIR receptor– ligand mismatch cells, and in pathologic conditions with adult lymphoblastic leukemia. C, sufficient activating ligands must be expressed on target cells to induce NK cell activity. D, if self-MHC ligands are expressed in normal amount, the reactivity of the NK cell is ultimately dependent on the balance of activating and inhibitory signals. Successful NK cell therapy relies on clinical strategies that optimize activation and avoid inhibition by cancer cells.

CCR Reviews

3392

Clin Cancer Res; 20(13) July 1, 2014

Clinical Cancer Research

Downloaded from clincancerres.aacrjournals.org on July 17, 2014. © 2014 American Association for Cancer Research.

NK Infusions as Cancer Therapy

A Donor selection, including KIR typing 105 0

Donor

0

Best

104 3

2D L1 2D L 2D 2 L 3D 3 L N 1 eg H 2O

10

2

10

0

61.2 0 102

B

38.8 3

10

4

10

5

10

NK cell purification and processing ex vivo

C Patient

NK cell augmentation in vivo

QA and QC Aug Tumor

Tumor Tumor

NK NK Tumor

Aug

NK NK

Tumor

Tumor Tumor

NK

Tumor

NK

Aug Aug NK

© 2014 American Association for Cancer Research CCR Reviews Figure 3. Three basic steps of allogeneic NK cell therapy. A, the first step includes donor selection based on health history and examination, infectious biomarkers, and KIR typing, because KIRs are highly polymorphic. All three levels of KIR typing should be done, including genotyping, phenotyping, and allelotyping (depicted from left to right are representative outputs from flow cytometry, RT-PCR, and allelotyping analyses). B, the second step is NK cell purification and processing to enrich NK cells and to improve NK cell functions. The process needs vigorous steps for quality assurance (QA) and quality control (QC). C, after cell infusion, antitumor activity could be enhanced in vivo using various augmentation agents (Aug).

Patients with the best survival were those with a stronger KIR allele and receptor–ligand mismatch from donors. The second-best group was those with a stronger KIR allele but no receptor–ligand mismatch. The worst survival was in those with the weaker allele, regardless of mismatch. Donor KIR typing is not necessary in some KIR mismatch models (Fig. 4C). The first model was proposed by the Perugia group (7); the ligand mismatch model requires typing HLA in both donor and recipient. A donor is mismatched if a ligand is present in the donor but absent in the recipient. This ligand mismatch model does not require donor or recipient KIR typing. The second model was proposed by the Nantes group (30); in this model, KIR is typed in both donor and recipient. If a receptor is present in

www.aacrjournals.org

the donor but absent in the recipient, the donor is mismatched. Because B haplotypes contain more genes than A haplotypes, mismatch typically signifies a B haplotype in the donor. The third model is the receptor–ligand mismatch model proposed by the Memphis group (31). This model requires donor KIR typing and recipient HLA typing. A donor is mismatched if an inhibitory receptor is present in the donor but the ligand is absent in the recipient. Notably, the receptor–ligand mismatch model is the only model applicable to autologous transplantation and antibody therapy; studies have shown that the model is useful for predicting relapse in both therapeutic settings (32–34). Not all potential donors require all three levels of KIR typing (Fig. 4D).

Clin Cancer Res; 20(13) July 1, 2014

Downloaded from clincancerres.aacrjournals.org on July 17, 2014. © 2014 American Association for Cancer Research.

3393

Leung

A

Cen A

3DL3

3DL3

Tel A

2DL3

2DS2

2DL2

2DL5B

2DS3/5

2DL1

3DP1

2DL4

3DL1

2DL1

3DP1

2DL4

3DS1

2DS4

2DL5A

Cen B

2DS3/5

3DL2

2DS1

3DL2

Tel B

B KIR3DL1

KIR2DL3 Telomeric B score

Centromeric B score Positive Positive

1

2

0

Does not exist

KIR2DL2 Negative

Positive

Negative

Positive

1

2

Negative

0

Very rare

Negative

Total B score = Centromeric B score + Telomeric B score =

KIR3DS1 (________)

*Total B scores range from 0–4

C Mismatch model

Relationship

Ligand mismatch

Donor KIRL vs. recipient KIRL

Test ordered

Definition of mismatch

Donor HLA recipient HLA

Missing KIRL in a recipient for a KIRL that is present in donor

Donor KIR recipient KIR

Missing KIR in a recipient for a KIR that is present in donor

Example Donor

Receptor mismatch

Applicable to antibody therapy

No

No

No

No

Yes

Yes

Recipient

C2

C2

C1

C2

Donor Donor KIR vs. recipient KIR

Applicable to autologous cell therapy

Recipient

2DS/Bx Donor Receptor– ligand mismatch

Donor KIR vs. recipient KIRL

Donor KIR recipient HLA

Recipient

Missing KIRL in a recipient for a KIR that is present in donor

C2

2DL2

D

C245+ R245– Not selected

Donor KIR2DL1 allelotyping C245– R245+ BW4+ C1+ C2+

Recipient HLA typing

Donor KIR genotyping

Select donor with highest B score

Donor KIR genotyping

Select donor with highest B score

Missing ≥ 1 ligand Donor KIR phenotyping by flow cytometry

Receptor–ligand match

Receptor–ligand mismatch Select donor with largest amount of receptor–ligand mismatched cells © 2014 American Association for Cancer Research CCR Reviews

3394

Clin Cancer Res; 20(13) July 1, 2014

Clinical Cancer Research

Downloaded from clincancerres.aacrjournals.org on July 17, 2014. © 2014 American Association for Cancer Research.

NK Infusions as Cancer Therapy

Step 2: NK Cell Processing and Quality Assurance NK cell purification The second step for therapy is NK cell processing and preinfusion quality assessment, including cell count, viability, sterility, phenotype, function, and purity (35). Studies in the 1980s with autologous LAK cells consisted primarily of expanded polyclonal T cells with low NK percentages (36). The easiest way to select highly purified NK cells is immunomagnetic cell separation (37). After separation, the product typically contains more than 90% NK cells with very few T cells, B cells, and monocytes. Thus, the risks of GVHD, regulatory T cell (Treg) suppression, cytokine competition, Epstein-Barr virus–lymphoproliferative disease, passenger lymphocyte syndrome, cytokine storm, and suppression by myeloid suppressor cells or blood DCs are minimized (38–40). The NK phenotype is unaltered, and cells retain extensive proliferative capacity in vivo with potent antitumor response (41). Importantly, purified NK cells can be infused in small volumes, and studies have shown that purified cells are safe, do not cause GVHD, and are detectable in the recipient’s blood for approximately 2 to 4 weeks after infusion with preferential expansion of KIR-mismatched NK cells (42). NK cell bioprocessing ex vivo Donor NK cells can be manipulated ex vivo using a combination of strategies to optimize efficacy and specificity (Fig. 5). One challenge has been NK exhaustion, as adoptively transferred cells rapidly lose activating receptor expression and IFNg production capability through downregulation of transcription factors T-bet and eomesodermin (43). Furthermore, a highly acidic tumor environment may render NK cells nonfunctional (44). A common strategy to prime and activate cells is using soluble factors such as IL2, -12, -15, -18, and -21 or type I IFN (45). Studies have shown that IFNa and IL2 synergistically augment NK cells against solid tumors. Similarly, IL12 with either IL2 or IL18 can be used to overcome resistance (31). In cytokine-based culture systems, massive expansion of mature NK cells is possible. In a study of 7 patients with newly diagnosed, untreated multiple myeloma, the number of NK cells expanded on average by 1,600-fold after 20 days of culture with considerable increase in cytotoxicity (46). Alternatively, NK cells could be stimulated using artificial presenting cells with membrane-bound molecules such as IL15, IL21, and 41BB ligand (47, 48). Animal studies have shown that these cells are potent against Ewing sarcoma, rhabdomyosarcoma, and neuroblastoma and showed prolonged survival without GVHD.

Another way to increase NK potency is using chimeric antigen receptor (CAR). Receptor insertion can be mediated using retroviral, lentiviral, mRNA, or Sleeping Beauty transposon/transposase systems (49–51). Investigations have shown that NK cells transduced with anti-CD19 CAR or NKG2D are potent against B-lineage and osteosarcoma cells in vivo and in vitro (49, 52). CARþ cells have been generated against GD2 in neuroblastoma and CD33/CD123 in myeloid leukemia (51, 53, 54). Clinical trials are ongoing, including NCT00995137 and NCT01974479, for B-lineage hematologic malignancies. For centers without gene modification capabilities, NK cells could be activated by culturing in the presence of unmodified CD56 cells and stimulating cytokines. Recently, investigators showed that this method expanded and activated NK cells from donors and patients with neuroblastoma (55). These cells have great efficacy toward neuroblastoma in vitro and in vivo through NCR, DNAM-1, perforin, and granzyme B without risk of GVHD. One obstacle in NK therapy is insufficient homing to tumor sites. Recently, investigators used CCR7þ cells to transfer CCR7 onto NK cells via trogocytosis, resulting in better homing of NK cells to the tumor site in a mouse model and in Transwell migration experiments (56). Another way to facilitate adhesion of NK cells to tumor cells is using immunocytokines such as antibody against GD2 conjugated to IL2 (57). This chimeric protein attaches to GD2 on neuroblastoma cells on one side and to IL2 receptor on NK cells on the other. In addition to mature NK cells, human embryonic stem cells (hESC), induced pluripotent stem cells, and NK leukemia cell lines may be used as starting cells (58–60). hESCderived NK cells carry the CD94þCD117low/ phenotype, which has potent antitumor activity (58). Cytokine-based culture systems, in particular, enhance the expansion of NKG2D/NCRþ NK cells from umbilical cord blood (59). Because of the technical difficulty in producing cellular products at treatment centers, the National Heart, Lung, and Blood Institute (NHLBI; Bethesda, MD) sponsored the Production Assistance for Cellular Therapies program (PACT; ref. 61). Using this approach, apheresis cells have been sent to remote processing centers and purified, and activated NK cells were sent to the transplant centers for infusion into patients (62).

Step 3: NK Augmentation In Vivo Combination with therapeutic antibody or chimeric proteins One mechanism of antibodies in cancer therapy is ADCC. NK cells carry high-affinity Fc receptors that mediate this function. Conceptually, to overcome resistance, it would be

Figure 4. KIR haplotype map, B scoring, mismatch model, and typing algorithm. A, simplified genomic maps of A and B haplotypes. KIR genes that are present in some but not all cen-B motifs are color-coded in green and the two loci that are used for simplified B-scoring are in red. B, calculation of total B scores. C, all three mismatch models are biologically sound but are fundamentally different in principles and in clinical usages, including the relationship in consideration, the blood tests required, the definition of mismatches, and the applicability to antibody therapy and autologous settings. D, donor typing and selection algorithms.

www.aacrjournals.org

Clin Cancer Res; 20(13) July 1, 2014

Downloaded from clincancerres.aacrjournals.org on July 17, 2014. © 2014 American Association for Cancer Research.

3395

Leung

NK cells derived from ESC/IPS ESC IPS

Selection of KIREx vivo NKp favorable development and donor mNK expansion

mNK cell purification and activation

NKp

HSC

mNK Augmentation strategies • ↑ Survival (IL21) • ↑ Cytokine priming (IFNα, IL2, IL12, IL18) • ↑ Homing/adhesion (togocytosis, ICs) • ↑ Specificity/potency (mAb, BiKE, CAR) • ↑ CD16 (ADAM17 inhibitor) • ↑ Activating receptor (NKG2D, DNAM, NCR) • ↑ Lysis (lenalidomide, TKI, decitabine, CTLA4) • ↓ Inhibition (anti-KIR, anti-NKG2a, anti-A2A, anti-PD1 mAb)

Sensitization strategies

Tumor

• HDAC inhibitor • ADAM inhibitor • Proteasome inhibitor • Demethylation • RXRγ agonist • DNA damage • JAK inhibitor

Tumor

iDC

aNK

DC editing and maturation

Lysis B cell

IFNγ

CD4+ Th

CD8+

mDC

CTL Tumor Ag uptake and presentation

Antitumor antibody

Figure 5. Clinically feasible approaches to optimize NK cell therapy. Most protocols use mature NK cells (mNK) as starting cells, while a few use stem or progenitor cells such as ESC, induced pluripotent stem cells (IPS), and hematopoietic stem cells (HSC) to generate NK precursors (NKp). NK cells can be augmented through various strategies to obtain large numbers of activated NK cells (aNK). After infusion, the NK cells may kill cancer cells directly or mediate the development of adaptive immunity via interactions with immature DCs (iDC), B cells, T-helper cells (Th), cytotoxic T lymphocytes (CTL), and mature DCs (mDC). Tumor cells can be sensitized by various agents to render them more susceptible to NK cells. Bold arrows indicate cell intrinsic changes. Thin arrows indicate cell extrinsic interactions. Ag, antigen.

Tumor-specific adaptive immunity

© 2014 American Association for Cancer Research CCR Reviews

attractive to administer NK therapy concurrently with antibodies such as those against CD19, CD20, CD22, CD33, CD123, HER2, EGFR, and GD2 (63, 64). Typing for recipient and donor FcgRIII 158V/F polymorphism is important in optimizing ADCC (65). Stimulating NK cells with an agonistic monoclonal antibody specific for CD137 or with blocking antibodies against KIR/NKG2A may increase cell killing (66–68). Thus, using a second antibody that activates donor NK cells may improve the efficacy of antibodies against tumor-associated antigens. Another method to activate CD16 is a bispecific or trispecific killer cell engager (BiKE or TriKE). A fully humanized CD16-CD33 BiKE has been created that strongly activates NK cells against CD33þ AML blasts (69). Pretreatment with ADAM17 inhibitor prevented CD16 shedding and overcame inhibition of class I MHC-recognizing inhibitory receptors. Incubating cancer cells with BiKE and TriKE increased NK cytolytic activity against tumor targets and production of IFNg, TNFa, GM-CSF, IL8, MIP-1a, and RANTES (70).

3396

Clin Cancer Res; 20(13) July 1, 2014

Combination with supplemental medication After NK infusion, many regimens include low-dose IL2 injections. Newer agents are being investigated. Lenalidomide enhances ADCC in rituximab treatment of non-Hodgkin lymphoma and B-cell chronic lymphocytic leukemia through enhanced granzyme B and FasL expression (71). The effects of lenalidomide are partly related to CD4þ T-cell production of IL2 (72). Imatinib triggers DC-mediated NK activation (73), whereas dasatinib enhances NK expansion via eomesodermin (74). Ligation of CD86 on NK cells with CTLA4-Ig fusion protein or blockade of A2A adenosine receptor interaction with CD73 increases tumor killing (75, 76). During NK therapy, concurrent suppressive medications should be avoided, including agents such as azacytidine and sorafenib, which substantially impair PI3K and ERK phosphorylation and hamper NK reactivity (77, 78). Sunitinib does not inhibit NK cells, and decitabine may augment NK reactivity (78).

Clinical Cancer Research

Downloaded from clincancerres.aacrjournals.org on July 17, 2014. © 2014 American Association for Cancer Research.

NK Infusions as Cancer Therapy

Combination with tumor cell modulation After CD16, NKG2D is the second most potent activating receptor expressed on NK cells. Unfortunately, NKG2D ligand expression in most cancer cells is low (79). Recently, various methods have been used to upregulate NKG2D ligand expression or prevent its shedding from the cell surface, including epigenetic modulation with histone deacetylase (HDAC) inhibitors (which may induce glycogen synthase kinase-3 activity; ref. 80), proteosome inhibitors, and demethylating agents. These agents may also work together to induce expression of Fas and TRAIL-R2 (DR5; refs. 81, 82). Recently, high-throughput screening of 5,600 bioactive compounds revealed spironolactone as a novel RXRg agonist that activates the ATM–CHK2 pathway to upregulate transcription of all classes of NKG2D ligands for cancer prevention and control (83). In another study using a lentiviral shRNA library targeting more than 10,000 human genes, silencing of JAK1 and JAK2 increased the susceptibility of a variety of tumor cell types to NKmediated lysis (84).

Clinical Considerations NK cell therapy is applicable to various forms of cancer (85, 86), but its current use has been focused primarily in hematologic malignancies (20, 63, 87, 88). NK cells can be used for patients with refractory leukemia and can be given before conventional HCT for induction of remission, after HCT as consolidation, or in place of HCT. In an early feasibility study, IL2-stimulated NK cells were infused post-HCT into 3 pediatric patients who had persistent leukemia blasts at the time of transplant (89). No significant toxicity was observed, and complete remission and complete donor chimerism were observed within 1 month after HCT. In a subsequent prospective phase II study, preemptive immunotherapy with purified NK cells after haploidentical HCT was investigated in 16 patients (90). The patients received 29 NK infusions 3, 40, and 100 days after transplantation. The median dose of NK cells per product was 1.21  107/kg. With a median follow-up of 5.8 years, 4 of the 16 patients with high-risk leukemia remained alive. In 10 patients with advanced multiple myeloma receiving autologous HCT, haploidentical T cell–depleted, KIR ligand–mismatched NK cells were infused (91). No GVHD or failure of autologous stem cell reconstitution was observed. Encouragingly, 50% of the patients achieved near complete remission. These results set the stage for future studies of KIR ligand–mismatched NK therapy in the autologous setting.

NK therapy without HCT For allogeneic NK therapy in a nontransplant setting, transient suppression of the host immune system is required to allow time for donor-derived NK expansion and therapeutic effect. Even in the autologous setting, induction of transient leukopenia may promote homeostatic expansion of NK cells and reduce suppressive effects from Tregs and myeloid suppressor cells (32, 33, 92). One common outpatient regimen includes fludarabine and low-dose cyclophosphamide, and this regimen is well tolerated even in children, with 10 of 10 patients surviving free of leukemia (42). The rare occurrence of complications results in the cost of fresh NK therapy being less than 10% of that for conventional HCT. Infusions of purified NK cells were feasible after similar conditioning in 13 elderly patients including septuagenarians (93). In a study of haploidentical related-donor NK infusion, a high-intensity inpatient regimen of high-dose cyclophosphamide and fludarabine resulted in a marked increase in endogenous IL15, expansion of donor NK cells, and induction of complete hematologic remission in 5 of 19 patients with poor-prognosis AML (94). In a subsequent phase II study for recurrent ovarian and breast cancer, 20 patients underwent the regimen of fludarabine and cyclophosphamide with or without low-dose total body irradiation (95). After cell infusion, IL2 was administered subcutaneously for 2 weeks. With a mean NK cell dose of 2.16  107 cells/kg, 9 of 13 (69%) patients without irradiation and 6 of 7 with irradiation had donor DNA detectable after NK cell infusions. In another study of 6 patients with advanced B-cell non-Hodgkin lymphoma, 4 patients showed an objective clinical response (96). Unfortunately, Tregs also expanded in vivo in these two studies; therefore, future strategies should include novel techniques to suppress the expansion of Tregs.

Conclusions In summary, NK cells are promising in cancer therapy, considering their speed of action, potency in cancer cell killing, applicability to many types of cancer, lack of adverse effects, ease of preparation and administration, availability, permissibility of donor selection, affordability, and complimentary actions with other therapies. The unique properties of NK cells open a new arena of novel cell-based immunotherapy against cancers that are resistant to contemporary therapies. Received October 29, 2013; revised February 6, 2014; accepted February 25, 2014; published online July 1, 2014.

References 1.

2.

Kirkwood JM, Butterfield LH, Tarhini AA, Zarour H, Kalinski P, Ferrone S. Immunotherapy of cancer in 2012. CA Cancer J Clin 2012;62: 309–35. 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;5:117–21.

www.aacrjournals.org

3.

4.

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;16:230–9. 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;16:216–29.

Clin Cancer Res; 20(13) July 1, 2014

Downloaded from clincancerres.aacrjournals.org on July 17, 2014. © 2014 American Association for Cancer Research.

3397

Leung

5.

6.

7.

8.

9.

10.

11.

12. 13.

14.

15.

16. 17.

18.

19.

20. 21.

22.

23.

24.

25.

26.

3398

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;5:112–7. Valiante NM, Parham P. Natural killer cells, HLA class I molecules, and marrow transplantation. Biol Blood Marrow Transplant 1997;3: 229–35. Ruggeri L, Capanni M, Urbani E, Perruccio K, Shlomchik WD, Tosti A, et al. Effectiveness of donor natural killer cell alloreactivity in mismatched hematopoietic transplants. Science 2002;295:2097–100. MacLennan IC, Connell GE, Gotch FM. Effector activating determinants on IgG. II. Differentiation of the combining sites for C1q from those for cytotoxic K cells and neutrophils by plasmin digestion of rabbits IgG. Immunology 1974;26:303–10. Liu D, Xu L, Yang F, Li D, Gong F, Xu T. Rapid biogenesis and sensitization of secretory lysosomes in NK cells mediated by targetcell recognition. Proc Natl Acad Sci U S A 2005;102:123–7. De Maria A, Bozzano F, Cantoni C, Moretta L. Revisiting human natural killer cell subset function revealed cytolytic CD56(dim)CD16þ NK cells as rapid producers of abundant IFN-gamma on activation. Proc Natl Acad Sci U S A 2010;108:728–32. Li S, Yan Y, Lin Y, Bullens DM, Rutgeerts O, Goebels J, et al. Rapidly induced, T-cell independent xenoantibody production is mediated by marginal zone B cells and requires help from NK cells. Blood 2007; 110:3926–35. Croft M. Control of immunity by the TNFR-related molecule OX40 (CD134). Annu Rev Immunol 2010;28:57–78. Cudkowicz G, Bennett M. Peculiar immunobiology of bone marrow allografts. II. Rejection of parental grafts by resistant F 1 hybrid mice. J Exp Med 1971;134:1513–28. Cudkowicz G, Stimpfling JH. Hybrid resistance to parental marrow grafts: association with the K region of H-2. Science 1964;144: 1339–40. Karre K, Ljunggren HG, Piontek G, Kiessling R. Selective rejection of H2-deficient lymphoma variants suggests alternative immune defence strategy. Nature 1986;319:675–8. Ljunggren HG, Karre K. In search of the 'missing self': MHC molecules and NK cell recognition. Immunol Today 1990;11:237–44. Colonna M, Samaridis J. Cloning of immunoglobulin-superfamily members associated with HLA-C and HLA-B recognition by human natural killer cells. Science 1995;268:405–8. D'Andrea A, Chang C, Franz-Bacon K, McClanahan T, Phillips JH, Lanier LL. Molecular cloning of NKB1. A natural killer cell receptor for HLA-B allotypes. J Immunol 1995;155:2306–10. Wagtmann N, Biassoni R, Cantoni C, Verdiani S, Malnati MS, Vitale M, et al. Molecular clones of the p58 NK cell receptor reveal immunoglobulin-related molecules with diversity in both the extra- and intracellular domains. Immunity 1995;2:439–49. Leung W. Use of NK cell activity in cure by transplant. Br J Haematol 2011;155:14–29. Chan WK, Rujkijyanont P, Neale G, Yang J, Bari R, Das Gupta N, et al. Multiplex and genome-wide analyses reveal distinctive properties of KIRþ and CD56þ T cells in human blood. J Immunol 2013;191: 1625–36. Pyo CW, Guethlein LA, Vu Q, Wang R, Abi-Rached L, Norman PJ, et al. Different patterns of evolution in the centromeric and telomeric regions of group A and B haplotypes of the human killer cell Ig-like receptor locus. PLoS ONE 2010;5:e15115. Parham P, Moffett A. Variable NK cell receptors and their MHC class I ligands in immunity, reproduction and human evolution. Nat Rev Immunol 2013;13:133–44. Leung W, Iyengar R, Triplett B, Turner V, Behm FG, Holladay MS, et al. Comparison of killer Ig-like receptor genotyping and phenotyping for selection of allogeneic blood stem cell donors. J Immunol 2005; 174:6540–5. Shilling HG, Young N, Guethlein LA, Cheng NW, Gardiner CM, Tyan D, et al. Genetic control of human NK cell repertoire. J Immunol 2002;169:239–47. Pende D, Marcenaro S, Falco M, Martini S, Bernardo ME, Montagna D, et al. Anti-leukemia activity of alloreactive NK cells in KIR ligand-

Clin Cancer Res; 20(13) July 1, 2014

27.

28.

29.

30.

31.

32.

33.

34.

35.

36.

37.

38.

39.

40. 41.

42.

43.

44.

45.

46.

mismatched haploidentical HSCT for pediatric patients: evaluation of the functional role of activating KIR and redefinition of inhibitory KIR specificity. Blood 2009;113:3119–29. Bari R, Bell T, Leung WH, Vong QP, Chan WK, Das Gupta N, et al. Significant functional heterogeneity among KIR2DL1 alleles and a pivotal role of arginine 245. Blood 2009;114:5182–90. Bari R, Leung M, Turner VE, Embrey C, Rooney B, Holladay M, et al. Molecular determinant-based typing of KIR alleles and KIR ligands. Clin Immunol 2011;138:274–81. Bari R, Rujkijyanont P, Sullivan E, Kang G, Turner V, Gan K, et al. Effect of donor KIR2DL1 allelic polymorphism on the outcome of pediatric allogeneic hematopoietic stem-cell transplantation. J Clin Oncol 2013;31:3782–90. Gagne K, Brizard G, Gueglio B, Milpied N, Herry P, Bonneville F, et al. Relevance of KIR gene polymorphisms in bone marrow transplantation outcome. Hum Immunol 2002;63:271–80. Leung W, Iyengar R, Turner V, Lang P, Bader P, Conn P, et al. Determinants of antileukemia effects of allogeneic NK cells. J Immunol 2004;172:644–50. Leung W, Handgretinger R, Iyengar R, Turner V, Holladay MS, Hale GA. Inhibitory KIR-HLA receptor-ligand mismatch in autologous haematopoietic stem cell transplantation for solid tumour and lymphoma. Br J Cancer 2007;97:539–42. Venstrom JM, Zheng J, Noor N, Danis KE, Yeh AW, Cheung IY, et al. KIR and HLA genotypes are associated with disease progression and survival following autologous hematopoietic stem cell transplantation for high-risk neuroblastoma. Clin Cancer Res 2009;15:7330–4. Delgado DC, Hank JA, Kolesar J, Lorentzen D, Gan J, Seo S, et al. Genotypes of NK cell KIR receptors, their ligands, and Fcgamma receptors in the response of neuroblastoma patients to Hu14.18-IL2 immunotherapy. Cancer Res 2010;70:9554–61. Koehl U, Brehm C, Huenecke S, Zimmermann SY, Kloess S, Bremm M, et al. Clinical grade purification and expansion of NK cell products for an optimized manufacturing protocol. Front Oncol 2013;3:118. Rosenberg SA, Lotze MT, Muul LM, Leitman S, Chang AE, Ettinghausen SE, 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;313:1485–92. Iyengar R, Handgretinger R, Babarin-Dorner A, Leimig T, Otto M, Geiger TL, et al. Purification of human natural killer cells using a clinical-scale immunomagnetic method. Cytotherapy 2003;5:479–84. Smyth MJ, Teng MW, Swann J, Kyparissoudis K, Godfrey DI, Hayakawa Y. CD4þCD25þ T regulatory cells suppress NK cell-mediated immunotherapy of cancer. J Immunol 2006;176:1582–7. Perez-Martinez A, Iyengar R, Gan K, Chotsampancharoen T, Rooney B, Holladay M, et al. Blood dendritic cells suppress NK cell function and increase the risk of leukemia relapse after hematopoietic cell transplantation. Biol Blood Marrow Transplant 2011;17:598–607. Shook DR, Leung W. Natural killer cell therapy for cancer: delivering on a promise. Transfusion 2013;53:245–8. Leung W, Iyengar R, Leimig T, Holladay MS, Houston J, Handgretinger R. Phenotype and function of human natural killer cells purified by using a clinical-scale immunomagnetic method. Cancer Immunol Immunother 2005;54:389–94. Rubnitz JE, Inaba H, Ribeiro RC, Pounds S, Rooney B, Bell T, et al. NKAML: a pilot study to determine the safety and feasibility of haploidentical natural killer cell transplantation in childhood acute myeloid leukemia. J Clin Oncol 2010;28:955–9. Gill S, Vasey AE, De Souza A, Baker J, Smith AT, Kohrt HE, et al. Rapid development of exhaustion and down-regulation of eomesodermin limit the antitumor activity of adoptively transferred murine natural killer cells. Blood 2012;119:5758–68. Helmlinger G, Yuan F, Dellian M, Jain RK. Interstitial pH and pO2 gradients in solid tumors in vivo: high-resolution measurements reveal a lack of correlation. Nat Med 1997;3:177–82. Moretta A, Ciccone E, Pantaleo G, Tambussi G, Bottino C, Melioli G, et al. Surface molecules involved in the activation and regulation of T or natural killer lymphocytes in humans. Immunol Rev 1989;111:145–75. Alici E, Sutlu T, Bjorkstrand B, Gilljam M, Stellan B, Nahi H, et al. Autologous antitumor activity by NK cells expanded from myeloma

Clinical Cancer Research

Downloaded from clincancerres.aacrjournals.org on July 17, 2014. © 2014 American Association for Cancer Research.

NK Infusions as Cancer Therapy

47.

48.

49.

50.

51.

52.

53.

54.

55.

56.

57.

58.

59.

60.

61.

62.

63. 64.

65.

66.

patients using GMP-compliant components. Blood 2008;111: 3155–62. Fujisaki H, Kakuda H, Shimasaki N, Imai C, Ma J, Lockey T, et al. Expansion of highly cytotoxic human natural killer cells for cancer cell therapy. Cancer Res 2009;69:4010–7. Denman CJ, Senyukov VV, Somanchi SS, Phatarpekar PV, Kopp LM, Johnson JL, et al. Membrane-bound IL-21 promotes sustained ex vivo proliferation of human natural killer cells. PLoS ONE 2012;7:e30264. 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;106:376–83. Shimasaki N, Fujisaki H, Cho D, Masselli M, Lockey T, Eldridge P, et al. A clinically adaptable method to enhance the cytotoxicity of natural killer cells against B-cell malignancies. Cytotherapy 2012;14:830–40. Tettamanti S, Magnani CF, Biondi A, Biagi E. Acute myeloid leukemia and novel biological treatments: monoclonal antibodies and cellbased gene-modified immune effectors. Immunol Lett 2013;155:43–6. Chang YH, Connolly J, Shimasaki N, Mimura K, Kono K, Campana D. A chimeric receptor with NKG2D specificity enhances natural killer cell activation and killing of tumor cells. Cancer Res 2013;73:1777–86. Altvater B, Landmeier S, Pscherer S, Temme J, Schweer K, Kailayangiri S, et al. 2B4 (CD244) signaling by recombinant antigen-specific chimeric receptors costimulates natural killer cell activation to leukemia and neuroblastoma cells. Clin Cancer Res 2009;15:4857–66. Esser R, Muller T, Stefes D, Kloess S, Seidel D, Gillies SD, 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. Rujkijyanont P, Chan WK, Eldridge PW, Lockey T, Holladay M, Rooney B, et al. Ex vivo activation of CD56(þ) immune cells that eradicate neuroblastoma. Cancer Res 2013;73:2608–18. Somanchi SS, Somanchi A, Cooper LJ, Lee DA. Engineering lymph node homing of ex vivo–expanded human natural killer cells via trogocytosis of the chemokine receptor CCR7. Blood 2012;119: 5164–72. Koehn TA, Trimble LL, Alderson KL, Erbe AK, McDowell KA, Grzywacz B, et al. Increasing the clinical efficacy of NK and antibody-mediated cancer immunotherapy: potential predictors of successful clinical outcome based on observations in high-risk neuroblastoma. Front Pharmacol 2012;3:91. Woll PS, Grzywacz B, Tian X, Marcus RK, Knorr DA, Verneris MR, et al. Human embryonic stem cells differentiate into a homogeneous population of natural killer cells with potent in vivo antitumor activity. Blood 2009;113:6094–101. Spanholtz J, Tordoir M, Eissens D, Preijers F, van der Meer A, Joosten I, et al. High log-scale expansion of functional human natural killer cells from umbilical cord blood CD34-positive cells for adoptive cancer immunotherapy. PLoS ONE 2010;5:e9221. Tonn T, Schwabe D, Klingemann HG, Becker S, Esser R, Koehl U, et al. Treatment of patients with advanced cancer with the natural killer cell line NK-92. Cytotherapy 2013;15:1563–70. Reed W, Noga SJ, Gee AP, Rooney CM, Wagner JE, McCullough J, et al. Production Assistance for Cellular Therapies (PACT): four-year experience from the United States National Heart, Lung, and Blood Institute (NHLBI) contract research program in cell and tissue therapies. Transfusion 2009;49:786–96. Koepsell SA, Kadidlo DM, Fautsch S, McCullough J, Klingemann H, Wagner JE, et al. Successful "in-flight" activation of natural killer cells during long-distance shipping. Transfusion 2013;53:398–403. Leung W. Immunotherapy in acute leukemia. Semin Hematol 2009;46: 89–99. Alderson KL, Sondel PM. Clinical cancer therapy by NK cells via antibody-dependent cell-mediated cytotoxicity. J Biomed Biotechnol 2011;2011:379123. Cartron G, Dacheux L, Salles G, Solal-Celigny P, Bardos P, Colombat P, et al. Therapeutic activity of humanized anti-CD20 monoclonal antibody and polymorphism in IgG Fc receptor FcgammaRIIIa gene. Blood 2002;99:754–8. Kohrt HE, Houot R, Weiskopf K, Goldstein MJ, Scheeren F, Czerwinski D, et al. Stimulation of natural killer cells with a CD137-specific

www.aacrjournals.org

67.

68.

69.

70.

71.

72.

73.

74.

75.

76.

77.

78.

79.

80.

81.

82.

83.

84.

85.

antibody enhances trastuzumab efficacy in xenotransplant models of breast cancer. J Clin Invest 2012;122:1066–75. Vey N, Bourhis JH, Boissel N, Bordessoule D, Prebet T, Charbonnier A, et al. A phase 1 trial of the anti-inhibitory KIR mAb IPH2101 for AML in complete remission. Blood 2012;120:4317–23. Benson DM Jr, Hofmeister CC, Padmanabhan S, Suvannasankha A, Jagannath S, Abonour R, et al. A phase 1 trial of the anti-KIR antibody IPH2101 in patients with relapsed/refractory multiple myeloma. Blood 2012;120:4324–33. Wiernik A, Foley B, Zhang B, Verneris MR, Warlick E, Gleason MK, et al. Targeting natural killer cells to acute myeloid leukemia in vitro with a CD16  33 bispecific killer cell engager and ADAM17 inhibition. Clin Cancer Res 2013;19:3844–55. Gleason MK, Verneris MR, Todhunter DA, Zhang B, McCullar V, Zhou SX, et al. Bispecific and trispecific killer cell engagers directly activate human NK cells through CD16 signaling and induce cytotoxicity and cytokine production. Mol Cancer Ther 2012;11:2674–84. Wu L, Adams M, Carter T, Chen R, Muller G, Stirling D, et al. lenalidomide enhances natural killer cell and monocyte-mediated antibodydependent cellular cytotoxicity of rituximab-treated CD20þ tumor cells. Clin Cancer Res 2008;14:4650–7. Hsu AK, Quach H, Tai T, Prince HM, Harrison SJ, Trapani JA, et al. The immunostimulatory effect of lenalidomide on NK-cell function is profoundly inhibited by concurrent dexamethasone therapy. Blood 2011;117:1605–13. Borg C, Terme M, Taieb J, Menard C, Flament C, Robert C, et al. Novel mode of action of c-kit tyrosine kinase inhibitors leading to NK celldependent antitumor effects. J Clin Invest 2004;114:379–88. Tanaka J, Sugita J, Shiratori S, Shigematsu A, Imamura M. Dasatinib enhances the expansion of CD56þCD3 NK cells from cord blood. Blood 2012;119:6175–6. Peng Y, Luo G, Zhou J, Wang X, Hu J, Cui Y, et al. CD86 is an activation receptor for NK cell cytotoxicity against tumor cells. PLoS ONE 2013;8: e83913. Beavis PA, Divisekera U, Paget C, Chow MT, John LB, Devaud C, et al. Blockade of A2A receptors potently suppresses the metastasis of CD73þ tumors. Proc Natl Acad Sci U S A 2013;110:14711–6. Krusch M, Salih J, Schlicke M, Baessler T, Kampa KM, Mayer F, et al. The kinase inhibitors sunitinib and sorafenib differentially affect NK cell antitumor reactivity in vitro. J Immunol 2009;183:8286–94. Schmiedel BJ, Arelin V, Gruenebach F, Krusch M, Schmidt SM, Salih HR. Azacytidine impairs NK cell reactivity while decitabine augments NK cell responsiveness toward stimulation. Int J Cancer 2011;128: 2911–22. Diermayr S, Himmelreich H, Durovic B, Mathys-Schneeberger A, Siegler U, Langenkamp U, et al. NKG2D ligand expression in AML increases in response to HDAC inhibitor valproic acid and contributes to allorecognition by NK-cell lines with single KIR-HLA class I specificities. Blood 2008;111:1428–36. Skov S, Pedersen MT, Andresen L, Straten PT, Woetmann A, Odum N. Cancer cells become susceptible to natural killer cell killing after exposure to histone deacetylase inhibitors due to glycogen synthase kinase-3-dependent expression of MHC class I–related chain A and B. Cancer Res 2005;65:11136–45. Hallett WH, Ames E, Motarjemi M, Barao I, Shanker A, Tamang DL, et al. Sensitization of tumor cells to NK cell-mediated killing by proteasome inhibition. J Immunol 2008;180:163–70. Lundqvist A, Abrams SI, Schrump DS, Alvarez G, Suffredini D, Berg M, et al. Bortezomib and depsipeptide sensitize tumors to tumor necrosis factor-related apoptosis-inducing ligand: a novel method to potentiate natural killer cell tumor cytotoxicity. Cancer Res 2006;66:7317–25. Leung WH, Vong QP, Lin W, Janke L, Chen T, Leung W. Modulation of NKG2D ligand expression and metastasis in tumors by spironolactone via RXRgamma activation. J Exp Med 2013;210:2675–92. Bellucci R, Nguyen HN, Martin A, Heinrichs S, Schinzel AC, Hahn WC, et al. Tyrosine kinase pathways modulate tumor susceptibility to natural killer cells. J Clin Invest 2012;122:2369–83. Klingemann HG. Cellular therapy of cancer with natural killer cells— where do we stand? Cytotherapy 2013;15:1185–94.

Clin Cancer Res; 20(13) July 1, 2014

Downloaded from clincancerres.aacrjournals.org on July 17, 2014. © 2014 American Association for Cancer Research.

3399

Leung

86. Koepsell SA, Miller JS, McKenna DH Jr. Natural killer cells: a review of manufacturing and clinical utility. Transfusion 2013;53:404–10. 87. Farnault L, Sanchez C, Baier C, Le Treut T, Costello RT. Hematological malignancies escape from NK cell innate immune surveillance: mechanisms and therapeutic implications. Clin Dev Immunol 2012; 2012:421702. 88. Farhan S, Lee DA, Champlin RE, Ciurea SO. NK cell therapy: targeting disease relapse after hematopoietic stem cell transplantation. Immunotherapy 2012;4:305–13. 89. Koehl U, Sorensen J, Esser R, Zimmermann S, Gruttner HP, Tonn T, et al. IL-2 activated NK cell immunotherapy of three children after haploidentical stem cell transplantation. Blood Cells Mol Dis 2004; 33:261–6. 90. Stern M, Passweg JR, Meyer-Monard S, Esser R, Tonn T, Soerensen J, et al. Pre-emptive immunotherapy with purified natural killer cells after haploidentical SCT: a prospective phase II study in two centers. Bone Marrow Transplant 2013;48:433–8. 91. Shi J, Tricot G, Szmania S, Rosen N, Garg TK, Malaviarachchi PA, et al. Infusion of haplo-identical killer immunoglobulin-like receptor ligand

3400

Clin Cancer Res; 20(13) July 1, 2014

92.

93.

94.

95.

96.

mismatched NK cells for relapsed myeloma in the setting of autologous stem cell transplantation. Br J Haematol 2008;143:641–53. Terme M, Ullrich E, Delahaye NF, Chaput N, Zitvogel L. Natural killer cell-directed therapies: moving from unexpected results to successful strategies. Nat Immunol 2008;9:486–94. Curti A, Ruggeri L, D'Addio A, Bontadini A, Dan E, Motta MR, et al. Successful transfer of alloreactive haploidentical KIR ligand-mismatched natural killer cells after infusion in elderly high risk acute myeloid leukemia patients. Blood 2011;118:3273–9. Miller JS, Soignier Y, Panoskaltsis-Mortari A, McNearney SA, Yun GH, Fautsch SK, et al. Successful adoptive transfer and in vivo expansion of human haploidentical NK cells in patients with cancer. Blood 2005; 105:3051–7. Geller MA, Cooley S, Judson PL, Ghebre R, Carson LF, Argenta PA, et al. A phase II study of allogeneic natural killer cell therapy to treat patients with recurrent ovarian and breast cancer. Cytotherapy 2011;13:98–107. Bachanova V, Burns LJ, McKenna DH, Curtsinger J, PanoskaltsisMortari A, Lindgren BR, et al. Allogeneic natural killer cells for refractory lymphoma. Cancer Immunol Immunother 2010;59:1739–44.

Clinical Cancer Research

Downloaded from clincancerres.aacrjournals.org on July 17, 2014. © 2014 American Association for Cancer Research.

Infusions of allogeneic natural killer cells as cancer therapy.

Natural killer (NK) cells are normal white blood cells capable of killing malignant cells without prior sensitization. Allogeneic NK cell infusions ar...
1MB Sizes 5 Downloads 4 Views