Clinical Cancer Review Research

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 Infusions of purified natural killer (NK) cells are late- For many cancers, cure rates remain unacceptably low. comers in cellular immunotherapy, compared with den- Advances in genetics and have led to biologic dritic cells (DC), lymphokine-activated killer (LAK) cells, therapies with non–cross-resistant mechanisms of action tumor-infiltrating lymphocytes, and conventional cytotoxic and no overlapping toxicities. Cancer immunotherapy is T cells. Many investigators consider that the cytolytic activ- possible with an array of effector mechanisms involving ity of LAK cells commonly used in the 1980s actually innate and adaptive immunity (1). Various cytokines, anti- represents the activity of IL2-activated NK cells, although bodies, and cellular therapies have been FDA approved or the products contained polyclonal T cells. Biologically, are in late-phase clinical trials. 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 Author's Affiliations: 1Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children's Research Hospital; and 2Department potential of human NK cells was not revealed until their of Pediatrics, University of Tennessee, Memphis, Tennessee biology of target cell recognition was elucidated in the Corresponding Author: Wing Leung, St. Jude Children's Research Hos- 1990s and the antileukemia effect was observed in HLA- pital, 262 Danny Thomas Place, Memphis, TN 38105-3678. Phone: 901- haploidentical hematopoietic cell transplantation (HCT) in 595-2617; Fax: 901-521-9005; E-mail: [email protected] the early 2000s (6, 7). This review covers NK cell biology doi: 10.1158/1078-0432.CCR-13-1766 relevant to cancer immunotherapy and outlines avenues 2014 American Association for Cancer Research. being explored in clinical trials.

3390 Clin Cancer Res; 20(13) July 1, 2014

Downloaded from clincancerres.aacrjournals.org on October 2, 2021. © 2014 American Association for Cancer Research. NK Infusions as Cancer Therapy

NK Cell Biology autologous NK therapy (20). Among known NK receptors, NK cells were first named and characterized by Kiessling the KIR family is one of the primary determinants at the Karolinska Institute (Stockholm, Sweden) and by of NK response. Subsets of T cells also expressed KIR (21). Herberman at the National Cancer Institute (Bethesda, Clinical KIR typing includes genotyping for content and A/B haplotype categorization, phenotyping for gene MD; refs. 2–5). The term "natural" referred to their natural þ existence in rodents and "killer" to their spontaneous expression and number of KIR cells, and allelotyping for killing of lymphoma and leukemia cells in nonimmune functional strength. animals. They were called "N-cells" or "K cells" by some The first level of KIR diversity is gene content (22, 23). investigators (3, 8). These cells were unique, as they lacked Only approximately 5% of people have all 15 gene family markers characteristic of B and T lymphocytes and were members; others lack one or more . The diversity of nonphagocytic, low-adhesive, and cytotoxic to IgG-coated gene content is based primarily on diversity in B haplo- targets. types, which typically contain more activating KIRs. The NK cells are generated in bone marrow and develop two hallmark genes for A haplotypes are KIR2DL3 and normally in athymic mice and in humans with DiGeorge KIR3DL1. They are segregated with KIR2DL2 and KIR3DS1 syndrome. Induction of cytotoxicity is possible in NK as alleles in the centromeric (Cen) and telomeric (Tel) cells within minutes, in contrast with the slower conven- motifs, respectively (Fig. 4A). On the basis of this pattern, a simplified typing and scoring method for B haplotype tional induction of the T-cell immune response (9). þ content can be derived (Fig. 4B), viz., KIR2DL3 / Approximately 5% to 15% of human blood lymphocytes þ þ þ KIR2DL2 , Cen-A/A; KIR2DL3 /KIR2DL2 , Cen-A/B; are NK cells (CD56 , CD3/14/19 ), 90% of which are þ þ þ KIR2DL3 /KIR2DL2 , Cen-B/B; KIR3DL1 /KIR3DS1 , CD56dim, with most of them being CD16 ,andtheyare þ þ Tel-A/A; KIR3DL1 /KIR3DS1 , Tel-A/B; and KIR3DL1 / responsible for early innate immunity and antibody- þ dependent cell-mediated cytotoxicity (ADCC) against KIR3DS1 , Tel-B/B. Because KIRs are encoded in chromo- infectionorcancerthroughIFNg, perforin, granzyme, some 19 and ligand HLAs are in 6, these gene FasL, or TRAIL (10). Approximately 10% of human blood families are segregated independently; thus, HLA geno- NK cells are CD56bright, and they participate in late (>16 types cannot be used to predict KIR content, and autolo- hours) inflammatory response by secreting IFNg,TNFa, gous KIR-HLA mismatch is possible. G-CSF, GM-CSF, and IL3. Interactions between DCs and The second level of diversity is gene expression (24). By NK cells are essential for priming adaptive immunity. NK real-time PCR and flow cytometry, more than 10-fold cells may also directly interact with T and B cells through variability in expression has been observed among donors. CD40 ligand and OX40 (11, 12). Nowadays, the easiest method to quantify NK cells expres- sing only one inhibitory KIR gene (i.e., negative for all other MHC inhibitory receptors) is multicolor flow cyto- Regulatory receptors þ Human NK cells are regulated by a sophisticated network metry (24–26). The number of single KIR cells is in direct of surface receptors (Fig. 1), allowing them to distinguish proportion to activity against target cells without the ligand (receptor–ligand mismatched). Although the num- normal from abnormal cells within seconds. Target cell þ lysis occurs when the activating signal dominates the ber of single KIR cells correlates with the presence of self- inhibitory signal (Fig. 2). Early studies revealed similarities ligand (25), the variability among donors renders predic- between murine NK cells and cells responsible for so-called tions based on the ligand alone unacceptable for clinical "hybrid resistance" to parental hematopoietic grafts (13, use. 14). The proposal that the resistance was related to recep- The third level of diversity is allele polymorphism, which tors recognizing the absence of MHC class I arose from has been observed in all inhibitory KIR genes (22, 23). For observations that murine lymphoma cells that were low in example, in the KIR2DL1 family, 25 alleles have been MHC expression were NK-susceptible (15). This led to the observed, and each has different strengths in inhibiting NK "missing-self" hypothesis in the 1980s (16). Break- cells and different durability of surface expression after throughs in human NK receptor biology were seen in the interaction with ligands (27). These differences correlate early 1990s, when the KIR gene family was found to with the intensity of inhibitory signaling through SHP2 and recognize HLA class I (17–19). Identification of other NK b arrestin-2, resulting in differences in immune synapse receptors, such as NCRs, NKG2D, DNAM, 2B4, and CD94/ formation. Mechanistically, the arginine residue at position NKG2A, collectively points to a sophisticated network that 245 in the transmembrane domain is important in inhibi- controls human NK activity. tion. 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 Step 1: KIR Typing for Donor Selection and assays have been developed for rapid, high-throughput Prognostication clinical typing (28). The same assay can be simultaneously Three general steps in NK-based therapy are summarized used for KIR ligand typing. Notably, this assay requires less in Fig. 3. KIR typing is a critical first step because KIR is than 0.1 mg of DNA and can be performed within 1 day. highly polymorphic. KIR typing is not only useful for Using this assay, investigations showed that patients who allogeneic donor selection but also for prognostication in received a donor graft containing the stronger KIR allele had

www.aacrjournals.org Clin Cancer Res; 20(13) July 1, 2014 3391

Downloaded from clincancerres.aacrjournals.org on October 2, 2021. © 2014 American Association for Cancer Research. Leung

IFNα IL1 IL2 IL12 IL15 IL18 IL21

Figure 1. Surface receptors and NK cell their ligands. Cytokine receptors KIR2DL1/2/3 (HLA-C) KIR2DS1/S2 (HLA-C) are shown on top of a human NK cell. Other receptors are broadly KIR3DL1 (HLA-B) KIR2DS4 (HLA-A, -C) classified and color-coded on the KIR3DL2 (HLA-A) KIR2DL4 (HLA-G, HS) basis of their primary function (inhibitory receptors in red, CD94/NKG2A (HLA-E) CD94/NKG2C (HLA-E) Inhibitory Activating activating receptors in green, inhibitory coreceptors in red-black LIR-1 (HLA-A–G) KIR3DL2 (CpG) stripes, and activating coreceptors KLRG-1 (cadherins) CD16 (IgG) in green-black stripes). Their ligands are shown within CEACAM1 (CEACAM5) NKG2D (MIC, ULBP) parentheses. Many other known receptors are not shown, including TIGIT (PVR, PVRL2) NCRs (B7-H6, NKp44L) chemotactic receptors (CCR-2, -5, Coreceptor -7: CXCR-1, -3, -4, -6; CX3CR1; and Chem23R), adhesion Siglec –3, –7, –9 (sialic acid) DNAM-1 (PVR, Nectin-2) receptors (CD2 and b1 and b2 integrins), and activating LAIR-1 (collagen) 2B4 (CD48) coreceptors (CD96, CS1, and TLR). CD300A (PS) NKp80 NTBA (NTBA) (AICL)

© 2014 American Association for Cancer Research CCR Reviews

fewer relapses, better survival, and less transplant-related acute lymphoblastic leukemia (ALL)], total body irradia- mortality (29). These effects were observed regardless of tion, T-cell depletion, and donor type. Statistically, there is primary disease [acute myelogenous leukemia (AML) vs. an interaction effect with HLA-C receptor–ligand mismatch:

Characteristics Figure 2. Dynamic equilibrium. After NK cell Target cell Outcome of target cell cell-to-cell contact, NK cell integrates signals from its surface A receptors in seconds, resulting in – either target-cell attack or no Normal MHC class I response and continual + No response No activating ligands immunosurveillance of other cells. A, healthy cells express normal amount of MHC class I ligands with no activating "stress" ligands. B, B downregulation or absence of MHC ligand for cognate inhibitory No MHC class I No response receptors is insufficient to trigger No activating ligands NK cells. This happens in the physiologic setting with red blood cells and autologous KIR receptor– C ligand mismatch cells, and in pathologic conditions with adult No MHC class I lymphoblastic leukemia. C, Attack target activating ligands sufficient activating ligands must be expressed on target cells to induce NK cell activity. D, if self-MHC D ligands are expressed in normal amount, the reactivity of the NK cell Normal MHC class I is ultimately dependent on the Dynamic equilibrium activating ligands balance of activating and inhibitory signals. Successful NK cell therapy relies on clinical strategies that optimize activation and avoid © 2014 American Association for Cancer Research inhibition by cancer cells. CCR Reviews

3392 Clin Cancer Res; 20(13) July 1, 2014 Clinical Cancer Research

Downloaded from clincancerres.aacrjournals.org on October 2, 2021. © 2014 American Association for Cancer Research. NK Infusions as Cancer Therapy

A

Donor selection, including KIR typing Donor

105 0 0

104 Best

103 O Neg H 2 102 2DL1 2DL22DL33DL1 0 61.2 38.8 B

2 3 4 5 0 10 10 10 10 NK cell purification and processing ex vivo

C in vivo Patient NK cell augmentation QA and QC

Aug NK Tumor Tumor Aug NK NK Tumor Tumor Tumor NK NK Aug Tumor NK Aug Tumor Tumor 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 the donor but absent in the recipient, the donor is mis- KIR allele and receptor–ligand mismatch from donors. The matched. Because B haplotypes contain more genes than A second-best group was those with a stronger KIR allele but haplotypes, mismatch typically signifies a B haplotype in no receptor–ligand mismatch. The worst survival was in the donor. The third model is the receptor–ligand mismatch those with the weaker allele, regardless of mismatch. model proposed by the Memphis group (31). This model Donor KIR typing is not necessary in some KIR mismatch requires donor KIR typing and recipient HLA typing. models (Fig. 4C). The first model was proposed by the A donor is mismatched if an inhibitory receptor is present Perugia group (7); the ligand mismatch model requires in the donor but the ligand is absent in the recipient. typing HLA in both donor and recipient. A donor is mis- Notably, the receptor–ligand mismatch model is the only matched if a ligand is present in the donor but absent in the model applicable to autologous transplantation and anti- recipient. This ligand mismatch model does not require body therapy; studies have shown that the model is useful donor or recipient KIR typing. The second model was for predicting relapse in both therapeutic settings (32–34). proposed by the Nantes group (30); in this model, KIR is Not all potential donors require all three levels of KIR typing typed in both donor and recipient. If a receptor is present in (Fig. 4D).

www.aacrjournals.org Clin Cancer Res; 20(13) July 1, 2014 3393

Downloaded from clincancerres.aacrjournals.org on October 2, 2021. © 2014 American Association for Cancer Research. Leung

A Cen A Tel A 3DL3 2DL3 2DL1 3DP1 2DL4 3DL1 2DS4 3DL2

3DL3 2DS2 2DL2 2DL5B 2DS3/5 2DL1 3DP1 2DL4 3DS1 2DL5A 2DS3/5 2DS1 3DL2

Cen B Tel B B KIR2DL3 KIR3DL1 Centromeric B score Telomeric B score Positive Negative Positive Negative

Positive 12 Positive 12 KIR2DL2 KIR3DS1 Negative 0 Does not Negative 0 Very rare exist Total B score = Centromeric B score + Telomeric B score = (______) *Total B scores range from 0–4 C

Applicable to Applicable to Mismatch Definition of Relationship Test ordered Example autologous antibody model mismatch cell therapy therapy Donor Recipient Donor KIRL Missing KIRL in Ligand vs. Donor HLA a recipient for a C2 C2 No No mismatch recipient recipient KIRL that is HLA KIRL present in donor C1 C2

Donor Recipient Missing KIR in Donor KIR Donor KIR Receptor a recipient for a vs. recipient No No mismatch KIR that is recipient KIR KIR present in donor 2DS/Bx Donor Recipient Missing KIRL in Receptor– Donor KIR Donor KIR a recipient for a C2 ligand vs. recipient KIR that is Yes Yes mismatch recipient HLA KIRL present in donor 2DL2

D C245+ R245– Donor KIR2DL1 allelotyping Not selected

C245– R245+ BW4+ C1+ C2+ Recipient HLA typing Donor KIR genotyping Select donor with highest B score

Missing ≥ 1 ligand

Donor KIR phenotyping Receptor–ligand match Donor KIR genotyping Select donor with highest B score by flow cytometry

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 October 2, 2021. © 2014 American Association for Cancer Research. NK Infusions as Cancer Therapy

Step 2: NK Cell Processing and Quality Another way to increase NK potency is using chimeric Assurance antigen receptor (CAR). Receptor insertion can be mediated using retroviral, lentiviral, mRNA, or Sleeping Beauty trans- NK cell purification poson/transposase systems (49–51). Investigations have The second step for therapy is NK cell processing and shown that NK cells transduced with anti-CD19 CAR or preinfusion quality assessment, including cell count, via- NKG2D are potent against B-lineage and osteosarcoma cells bility, sterility, phenotype, function, and purity (35). Stud- þ in vivo and in vitro (49, 52). CAR cells have been generated ies in the 1980s with autologous LAK cells consisted against GD2 in neuroblastoma and CD33/CD123 in mye- primarily of expanded polyclonal T cells with low NK loid leukemia (51, 53, 54). Clinical trials are ongoing, percentages (36). The easiest way to select highly purified including NCT00995137 and NCT01974479, for B-lineage NK cells is immunomagnetic cell separation (37). After hematologic malignancies. separation, the product typically contains more than 90% For centers without gene modification capabilities, NK NK cells with very few T cells, B cells, and monocytes. Thus, cells could be activated by culturing in the presence of the risks of GVHD, regulatory T cell (Treg) suppression, unmodified CD56 cells and stimulating cytokines. Recent- cytokine competition, Epstein-Barr virus–lymphoprolifera- ly, investigators showed that this method expanded and tive disease, passenger lymphocyte syndrome, cytokine activated NK cells from donors and patients with neuro- storm, and suppression by myeloid suppressor cells or blastoma (55). These cells have great efficacy toward neu- blood DCs are minimized (38–40). The NK phenotype is roblastoma in vitro and in vivo through NCR, DNAM-1, unaltered, and cells retain extensive proliferative capacity perforin, and granzyme B without risk of GVHD. in vivo with potent antitumor response (41). Importantly, One obstacle in NK therapy is insufficient homing to purified NK cells can be infused in small volumes, and þ tumor sites. Recently, investigators used CCR7 cells to studies have shown that purified cells are safe, do not cause transfer CCR7 onto NK cells via trogocytosis, resulting in GVHD, and are detectable in the recipient’s blood for better homing of NK cells to the tumor site in a mouse approximately 2 to 4 weeks after infusion with preferential model and in Transwell migration experiments (56). expansion of KIR-mismatched NK cells (42). Another way to facilitate adhesion of NK cells to tumor cells is using immunocytokines such as antibody against NK cell bioprocessing ex vivo GD2 conjugated to IL2 (57). This chimeric attaches Donor NK cells can be manipulated ex vivo using a to GD2 on neuroblastoma cells on one side and to IL2 combination of strategies to optimize efficacy and spec- receptor on NK cells on the other. ificity (Fig. 5). One challenge has been NK exhaustion, as In addition to mature NK cells, human embryonic stem adoptively transferred cells rapidly lose activating recep- cells (hESC), induced pluripotent stem cells, and NK leu- tor expression and IFNg production capability through kemia cell lines may be used as starting cells (58–60). hESC- downregulation of transcription factors T-bet and eome- þ derived NK cells carry the CD94 CD117low/ phenotype, sodermin (43). Furthermore, a highly acidic tumor envi- which has potent antitumor activity (58). Cytokine-based ronment may render NK cells nonfunctional (44). A culture systems, in particular, enhance the expansion of common strategy to prime and activate cells is using þ NKG2D/NCR NK cells from umbilical cord blood (59). soluble factors such as IL2, -12, -15, -18, and -21 or type Because of the technical difficulty in producing cellular I IFN (45). Studies have shown that IFNa and IL2 syn- products at treatment centers, the National Heart, Lung, and ergistically augment NK cells against solid tumors. Sim- Blood Institute (NHLBI; Bethesda, MD) sponsored the ilarly, IL12 with either IL2 or IL18 can be used to over- Production Assistance for Cellular Therapies program come resistance (31). In cytokine-based culture systems, (PACT; ref. 61). Using this approach, apheresis cells have massive expansion of mature NK cells is possible. In a been sent to remote processing centers and purified, and study of 7 patients with newly diagnosed, untreated activated NK cells were sent to the transplant centers for multiple myeloma, the number of NK cells expanded on infusion into patients (62). average by 1,600-fold after 20 days of culture with con- siderable increase in cytotoxicity (46). Step 3: NK Augmentation In Vivo Alternatively, NK cells could be stimulated using artificial presenting cells with membrane-bound molecules such as Combination with therapeutic antibody or chimeric IL15, IL21, and 41BB ligand (47, 48). Animal studies have shown that these cells are potent against Ewing sarcoma, One mechanism of antibodies in cancer therapy is ADCC. rhabdomyosarcoma, and neuroblastoma and showed pro- NK cells carry high-affinity Fc receptors that mediate this longed survival without GVHD. 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 3395

Downloaded from clincancerres.aacrjournals.org on October 2, 2021. © 2014 American Association for Cancer Research. Leung

NK cells Selection of KIR- Ex vivo NKp mNK cell derived from favorable development and purification ESC/IPS donor mNK expansion and activation

ESC NKp mNK IPS HSC

Augmentation strategies ↑ • Survival (IL21) Figure 5. Clinically feasible • ↑ Cytokine priming approaches to optimize NK cell (IFNα, IL2, IL12, IL18) therapy. Most protocols use mature • ↑ Homing/adhesion NK cells (mNK) as starting cells, (togocytosis, ICs) while a few use stem or progenitor • ↑ Specificity/potency cells such as ESC, induced pluripotent stem cells (IPS), and (mAb, BiKE, CAR) hematopoietic stem cells (HSC) to • ↑ CD16 (ADAM17 inhibitor) generate NK precursors (NKp). NK ↑ • Activating receptor cells can be augmented through (NKG2D, DNAM, NCR) various strategies to obtain large • ↑ Lysis (lenalidomide, numbers of activated NK cells TKI, decitabine, CTLA4) (aNK). After infusion, the NK cells Sensitization • ↓ Inhibition may kill cancer cells directly or strategies (anti-KIR, anti-NKG2a, mediate the development of anti-A2A, anti-PD1 mAb) adaptive immunity via interactions • HDAC inhibitor with immature DCs (iDC), B cells, • ADAM inhibitor T-helper cells (Th), cytotoxic T Tumor Tumor • Proteasome aNK iDC lymphocytes (CTL), and mature inhibitor DCs (mDC). Tumor cells can be • Demethylation DC editing sensitized by various agents to and maturation • RXRγ agonist render them more susceptible to Lysis NK cells. Bold arrows indicate cell • DNA damage IFNγ B cell intrinsic changes. Thin arrows • JAK inhibitor indicate cell extrinsic interactions. Antitumor Ag, antigen. + antibody mDC CD8+ CD4 CTL Th

Tumor-specific Tumor Ag uptake adaptive immunity and presentation

© 2014 American Association for Cancer Research CCR Reviews

attractive to administer NK therapy concurrently with anti- Combination with supplemental medication bodies such as those against CD19, CD20, CD22, CD33, After NK infusion, many regimens include low-dose CD123, HER2, EGFR, and GD2 (63, 64). Typing for recip- IL2 injections. Newer agents are being investigated. Lena- ient and donor FcgRIII 158V/F polymorphism is important lidomide enhances ADCC in rituximab treatment of in optimizing ADCC (65). Stimulating NK cells with an non-Hodgkin lymphoma and B-cell chronic lymphocytic agonistic monoclonal antibody specific for CD137 or with leukemia through enhanced granzyme B and FasL expres- blocking antibodies against KIR/NKG2A may increase cell sion (71). The effects of lenalidomide are partly related þ killing (66–68). Thus, using a second antibody that acti- to CD4 T-cell production of IL2 (72). Imatinib triggers vates donor NK cells may improve the efficacy of antibodies DC-mediated NK activation (73), whereas dasatinib against tumor-associated antigens. enhances NK expansion via eomesodermin (74). Ligation Another method to activate CD16 is a bispecific or tri- of CD86 on NK cells with CTLA4-Ig fusion protein or specific killer cell engager (BiKE or TriKE). A fully humanized blockade of A2A adenosine receptor interaction with CD16-CD33 BiKE has been created that strongly activates CD73 increases tumor killing (75, 76). þ NK cells against CD33 AML blasts (69). Pretreatment with During NK therapy, concurrent suppressive medications ADAM17 inhibitor prevented CD16 shedding and overcame should be avoided, including agents such as azacytidine and inhibition of class I MHC-recognizing inhibitory receptors. sorafenib, which substantially impair PI3K and ERK phos- Incubating cancer cells with BiKE and TriKE increased NK phorylation and hamper NK reactivity (77, 78). Sunitinib cytolytic activity against tumor targets and production of does not inhibit NK cells, and decitabine may augment NK IFNg,TNFa, GM-CSF, IL8, MIP-1a, and RANTES (70). reactivity (78).

3396 Clin Cancer Res; 20(13) July 1, 2014 Clinical Cancer Research

Downloaded from clincancerres.aacrjournals.org on October 2, 2021. © 2014 American Association for Cancer Research. NK Infusions as Cancer Therapy

Combination with tumor cell modulation NK therapy without HCT After CD16, NKG2D is the second most potent activating For allogeneic NK therapy in a nontransplant setting, receptor expressed on NK cells. Unfortunately, NKG2D transient suppression of the host is ligand expression in most cancer cells is low (79). Recently, required to allow time for donor-derived NK expansion various methods have been used to upregulate NKG2D and therapeutic effect. Even in the autologous setting, ligand expression or prevent its shedding from the cell induction of transient leukopenia may promote homeo- surface, including epigenetic modulation with histone dea- static expansion of NK cells and reduce suppressive effects cetylase (HDAC) inhibitors (which may induce glycogen from Tregs and myeloid suppressor cells (32, 33, 92). One synthase kinase-3 activity; ref. 80), proteosome inhibitors, common outpatient regimen includes fludarabine and and demethylating agents. These agents may also work low-dose cyclophosphamide, and this regimen is well together to induce expression of Fas and TRAIL-R2 (DR5; tolerated even in children, with 10 of 10 patients surviving refs. 81, 82). Recently, high-throughput screening of 5,600 free of leukemia (42). The rare occurrence of complica- bioactive compounds revealed spironolactone as a novel tions results in the cost of fresh NK therapy being less RXRg agonist that activates the ATM–CHK2 pathway to than 10% of that for conventional HCT. Infusions of upregulate transcription of all classes of NKG2D ligands purified NK cells were feasible after similar conditioning for cancer prevention and control (83). In another study in 13 elderly patients including septuagenarians (93). using a lentiviral shRNA library targeting more than 10,000 In a study of haploidentical related-donor NK infusion, a human genes, silencing of JAK1 and JAK2 increased high-intensity inpatient regimen of high-dose cyclophos- the susceptibility of a variety of tumor cell types to NK- phamide and fludarabine resulted in a marked increase in mediated lysis (84). endogenous IL15, expansion of donor NK cells, and induc- tion of complete hematologic remission in 5 of 19 patients Clinical Considerations with poor-prognosis AML (94). In a subsequent phase II NK cell therapy is applicable to various forms of cancer study for recurrent ovarian and breast cancer, 20 patients (85, 86), but its current use has been focused primarily in underwent the regimen of fludarabine and cyclophospha- hematologic malignancies (20, 63, 87, 88). NK cells can be mide with or without low-dose total body irradiation (95). used for patients with refractory leukemia and can be given After cell infusion, IL2 was administered subcutaneously for 7 before conventional HCT for induction of remission, after 2 weeks. With a mean NK cell dose of 2.16 10 cells/kg, 9 HCT as consolidation, or in place of HCT. of 13 (69%) patients without irradiation and 6 of 7 with In an early feasibility study, IL2-stimulated NK cells were irradiation had donor DNA detectable after NK cell infu- infused post-HCT into 3 pediatric patients who had persis- sions. In another study of 6 patients with advanced B-cell tent leukemia blasts at the time of transplant (89). No non-Hodgkin lymphoma, 4 patients showed an objective significant toxicity was observed, and complete remission clinical response (96). Unfortunately, Tregs also expanded and complete donor chimerism were observed within 1 in vivo in these two studies; therefore, future strategies month after HCT. In a subsequent prospective phase II should include novel techniques to suppress the expansion study, preemptive immunotherapy with purified NK cells of Tregs. after haploidentical HCT was investigated in 16 patients (90). The patients received 29 NK infusions 3, 40, and Conclusions 100 days after transplantation. The median dose of NK In summary, NK cells are promising in cancer therapy, 7 cells per product was 1.21 10 /kg. With a median fol- considering their speed of action, potency in cancer cell low-up of 5.8 years, 4 of the 16 patients with high-risk killing, applicability to many types of cancer, lack of adverse leukemia remained alive. effects, ease of preparation and administration, availability, In 10 patients with advanced multiple myeloma receiving permissibility of donor selection, affordability, and com- autologous HCT, haploidentical T cell–depleted, KIR plimentary actions with other therapies. The unique prop- ligand–mismatched NK cells were infused (91). No GVHD erties of NK cells open a new arena of novel cell-based or failure of autologous stem cell reconstitution was immunotherapy against cancers that are resistant to con- observed. Encouragingly, 50% of the patients achieved near temporary therapies. complete remission. These results set the stage for future studies of KIR ligand–mismatched NK therapy in the autol- Received October 29, 2013; revised February 6, 2014; accepted February ogous setting. 25, 2014; published online July 1, 2014.

References 1. Kirkwood JM, Butterfield LH, Tarhini AA, Zarour H, Kalinski P, Ferrone 3. Herberman RB, Nunn ME, Holden HT, Lavrin DH. Natural cytotoxic S. Immunotherapy of cancer in 2012. CA Cancer J Clin 2012;62: reactivity of mouse lymphoid cells against syngeneic and allogeneic 309–35. tumors. II. Characterization of effector cells. Int J Cancer 1975;16:230–9. 2. Kiessling R, Klein E, Pross H, Wigzell H. "Natural" killer cells in the 4. Herberman RB, Nunn ME, Lavrin DH. Natural cytotoxic reactivity of mouse. II. Cytotoxic cells with specificity for mouse Moloney leukemia mouse lymphoid cells against syngeneic acid allogeneic tumors. I. cells. Characteristics of the killer cell. Eur J Immunol 1975;5:117–21. Distribution of reactivity and specificity. Int J Cancer 1975;16:216–29.

www.aacrjournals.org Clin Cancer Res; 20(13) July 1, 2014 3397

Downloaded from clincancerres.aacrjournals.org on October 2, 2021. © 2014 American Association for Cancer Research. Leung

5. Kiessling R, Klein E, Wigzell H. "Natural" killer cells in the mouse. I. mismatched haploidentical HSCT for pediatric patients: evaluation of Cytotoxic cells with specificity for mouse Moloney leukemia cells. the functional role of activating KIR and redefinition of inhibitory KIR Specificity and distribution according to genotype. Eur J Immunol specificity. Blood 2009;113:3119–29. 1975;5:112–7. 27. Bari R, Bell T, Leung WH, Vong QP, Chan WK, Das Gupta N, et al. 6. Valiante NM, Parham P. Natural killer cells, HLA class I molecules, and Significant functional heterogeneity among KIR2DL1 alleles and a marrow transplantation. Biol Blood Marrow Transplant 1997;3: pivotal role of arginine 245. Blood 2009;114:5182–90. 229–35. 28. Bari R, Leung M, Turner VE, Embrey C, Rooney B, Holladay M, et al. 7. Ruggeri L, Capanni M, Urbani E, Perruccio K, Shlomchik WD, Tosti A, Molecular determinant-based typing of KIR alleles and KIR ligands. et al. Effectiveness of donor natural killer cell alloreactivity in mis- Clin Immunol 2011;138:274–81. matched hematopoietic transplants. Science 2002;295:2097–100. 29. Bari R, Rujkijyanont P, Sullivan E, Kang G, Turner V, Gan K, et al. Effect 8. MacLennan IC, Connell GE, Gotch FM. Effector activating determi- of donor KIR2DL1 allelic polymorphism on the outcome of pediatric nants on IgG. II. Differentiation of the combining sites for C1q from allogeneic hematopoietic stem-cell transplantation. J Clin Oncol those for cytotoxic K cells and neutrophils by plasmin digestion of 2013;31:3782–90. rabbits IgG. Immunology 1974;26:303–10. 30. Gagne K, Brizard G, Gueglio B, Milpied N, Herry P, Bonneville F, et al. 9. Liu D, Xu L, Yang F, Li D, Gong F, Xu T. Rapid biogenesis and Relevance of KIR gene polymorphisms in bone marrow transplantation sensitization of secretory lysosomes in NK cells mediated by target- outcome. Hum Immunol 2002;63:271–80. cell recognition. Proc Natl Acad Sci U S A 2005;102:123–7. 31. Leung W, Iyengar R, Turner V, Lang P, Bader P, Conn P, et al. 10. De Maria A, Bozzano F, Cantoni C, Moretta L. Revisiting human natural Determinants of antileukemia effects of allogeneic NK cells. J Immunol killer cell subset function revealed cytolytic CD56(dim)CD16þ NK cells 2004;172:644–50. as rapid producers of abundant IFN-gamma on activation. Proc Natl 32. Leung W, Handgretinger R, Iyengar R, Turner V, Holladay MS, Hale GA. Acad Sci U S A 2010;108:728–32. Inhibitory KIR-HLA receptor-ligand mismatch in autologous haema- 11. Li S, Yan Y, Lin Y, Bullens DM, Rutgeerts O, Goebels J, et al. Rapidly topoietic stem cell transplantation for solid tumour and lymphoma. Br J induced, T-cell independent xenoantibody production is mediated by Cancer 2007;97:539–42. marginal zone B cells and requires help from NK cells. Blood 2007; 33. Venstrom JM, Zheng J, Noor N, Danis KE, Yeh AW, Cheung IY, et al. 110:3926–35. KIR and HLA genotypes are associated with disease progression and 12. Croft M. Control of immunity by the TNFR-related molecule OX40 survival following autologous hematopoietic stem cell transplantation (CD134). Annu Rev Immunol 2010;28:57–78. for high-risk neuroblastoma. Clin Cancer Res 2009;15:7330–4. 13. Cudkowicz G, Bennett M. Peculiar immunobiology of bone marrow 34. Delgado DC, Hank JA, Kolesar J, Lorentzen D, Gan J, Seo S, et al. allografts. II. Rejection of parental grafts by resistant F 1 hybrid mice. Genotypes of NK cell KIR receptors, their ligands, and Fcgamma J Exp Med 1971;134:1513–28. receptors in the response of neuroblastoma patients to Hu14.18-IL2 14. Cudkowicz G, Stimpfling JH. Hybrid resistance to parental marrow immunotherapy. Cancer Res 2010;70:9554–61. grafts: association with the K region of H-2. Science 1964;144: 35. Koehl U, Brehm C, Huenecke S, Zimmermann SY, Kloess S, Bremm M, 1339–40. et al. Clinical grade purification and expansion of NK cell products for 15. Karre K, Ljunggren HG, Piontek G, Kiessling R. Selective rejection of H- an optimized manufacturing protocol. Front Oncol 2013;3:118. 2-deficient lymphoma variants suggests alternative immune defence 36. Rosenberg SA, Lotze MT, Muul LM, Leitman S, Chang AE, Ettinghau- strategy. Nature 1986;319:675–8. sen SE, et al. Observations on the systemic administration of autol- 16. Ljunggren HG, Karre K. In search of the 'missing self': MHC molecules ogous lymphokine-activated killer cells and recombinant interleukin-2 and NK cell recognition. Immunol Today 1990;11:237–44. to patients with metastatic cancer. N Engl J Med 1985;313:1485–92. 17. Colonna M, Samaridis J. Cloning of immunoglobulin-superfamily 37. Iyengar R, Handgretinger R, Babarin-Dorner A, Leimig T, Otto M, members associated with HLA-C and HLA-B recognition by human Geiger TL, et al. Purification of human natural killer cells using a natural killer cells. Science 1995;268:405–8. clinical-scale immunomagnetic method. Cytotherapy 2003;5:479–84. 18. D'Andrea A, Chang C, Franz-Bacon K, McClanahan T, Phillips JH, 38. Smyth MJ, Teng MW, Swann J, Kyparissoudis K, Godfrey DI, Haya- Lanier LL. Molecular cloning of NKB1. A natural killer cell receptor for kawa Y. CD4þCD25þ T regulatory cells suppress NK cell-mediated HLA-B allotypes. J Immunol 1995;155:2306–10. immunotherapy of cancer. J Immunol 2006;176:1582–7. 19. Wagtmann N, Biassoni R, Cantoni C, Verdiani S, Malnati MS, Vitale M, 39. Perez-Martinez A, Iyengar R, Gan K, Chotsampancharoen T, Rooney et al. Molecular clones of the p58 NK cell receptor reveal immuno- B, Holladay M, et al. Blood dendritic cells suppress NK cell function and globulin-related molecules with diversity in both the extra- and intra- increase the risk of leukemia relapse after hematopoietic cell trans- cellular domains. Immunity 1995;2:439–49. plantation. Biol Blood Marrow Transplant 2011;17:598–607. 20. Leung W. Use of NK cell activity in cure by transplant. Br J Haematol 40. Shook DR, Leung W. Natural killer cell therapy for cancer: delivering on 2011;155:14–29. a promise. Transfusion 2013;53:245–8. 21. Chan WK, Rujkijyanont P, Neale G, Yang J, Bari R, Das Gupta N, et al. 41. Leung W, Iyengar R, Leimig T, Holladay MS, Houston J, Handgretinger Multiplex and genome-wide analyses reveal distinctive properties of R. Phenotype and function of human natural killer cells purified by KIRþ and CD56þ T cells in human blood. J Immunol 2013;191: using a clinical-scale immunomagnetic method. Cancer Immunol 1625–36. Immunother 2005;54:389–94. 22. Pyo CW, Guethlein LA, Vu Q, Wang R, Abi-Rached L, Norman PJ, et al. 42. Rubnitz JE, Inaba H, Ribeiro RC, Pounds S, Rooney B, Bell T, et al. Different patterns of evolution in the centromeric and telomeric regions NKAML: a pilot study to determine the safety and feasibility of hap- of group A and B haplotypes of the human killer cell Ig-like receptor loidentical natural killer cell transplantation in childhood acute myeloid locus. PLoS ONE 2010;5:e15115. leukemia. J Clin Oncol 2010;28:955–9. 23. Parham P, Moffett A. Variable NK cell receptors and their MHC class I 43. Gill S, Vasey AE, De Souza A, Baker J, Smith AT, Kohrt HE, et al. Rapid ligands in immunity, reproduction and human evolution. Nat Rev development of exhaustion and down-regulation of eomesodermin Immunol 2013;13:133–44. limit the antitumor activity of adoptively transferred murine natural killer 24. Leung W, Iyengar R, Triplett B, Turner V, Behm FG, Holladay MS, et al. cells. Blood 2012;119:5758–68.

Comparison of killer Ig-like receptor genotyping and phenotyping for 44. Helmlinger G, Yuan F, Dellian M, Jain RK. Interstitial pH and pO2 selection of allogeneic blood stem cell donors. J Immunol 2005; gradients in solid tumors in vivo: high-resolution measurements reveal 174:6540–5. a lack of correlation. Nat Med 1997;3:177–82. 25. Shilling HG, Young N, Guethlein LA, Cheng NW, Gardiner CM, Tyan D, 45. Moretta A, Ciccone E, Pantaleo G, Tambussi G, Bottino C, Melioli G, et al. Genetic control of human NK cell repertoire. J Immunol et al. Surface molecules involved in the activation and regulation of T or 2002;169:239–47. natural killer lymphocytes in humans. Immunol Rev 1989;111:145–75. 26. Pende D, Marcenaro S, Falco M, Martini S, Bernardo ME, Montagna D, 46. Alici E, Sutlu T, Bjorkstrand B, Gilljam M, Stellan B, Nahi H, et al. et al. Anti-leukemia activity of alloreactive NK cells in KIR ligand- Autologous antitumor activity by NK cells expanded from myeloma

3398 Clin Cancer Res; 20(13) July 1, 2014 Clinical Cancer Research

Downloaded from clincancerres.aacrjournals.org on October 2, 2021. © 2014 American Association for Cancer Research. NK Infusions as Cancer Therapy

patients using GMP-compliant components. Blood 2008;111: antibody enhances trastuzumab efficacy in xenotransplant models of 3155–62. breast cancer. J Clin Invest 2012;122:1066–75. 47. Fujisaki H, Kakuda H, Shimasaki N, Imai C, Ma J, Lockey T, et al. 67. Vey N, Bourhis JH, Boissel N, Bordessoule D, Prebet T, Charbonnier A, Expansion of highly cytotoxic human natural killer cells for cancer cell et al. A phase 1 trial of the anti-inhibitory KIR mAb IPH2101 for AML in therapy. Cancer Res 2009;69:4010–7. complete remission. Blood 2012;120:4317–23. 48. Denman CJ, Senyukov VV, Somanchi SS, Phatarpekar PV, Kopp LM, 68. Benson DM Jr, Hofmeister CC, Padmanabhan S, Suvannasankha A, Johnson JL, et al. Membrane-bound IL-21 promotes sustained ex vivo Jagannath S, Abonour R, et al. A phase 1 trial of the anti-KIR antibody proliferation of human natural killer cells. PLoS ONE 2012;7:e30264. IPH2101 in patients with relapsed/refractory multiple myeloma. Blood 49. Imai C, Iwamoto S, Campana D. Genetic modification of primary 2012;120:4324–33. natural killer cells overcomes inhibitory signals and induces specific 69. Wiernik A, Foley B, Zhang B, Verneris MR, Warlick E, Gleason MK, et al. killing of leukemic cells. Blood 2005;106:376–83. Targeting natural killer cells to acute myeloid leukemia in vitro with a 50. Shimasaki N, Fujisaki H, Cho D, Masselli M, Lockey T, Eldridge P, et al. CD16 33 bispecific killer cell engager and ADAM17 inhibition. Clin A clinically adaptable method to enhance the cytotoxicity of natural Cancer Res 2013;19:3844–55. killer cells against B-cell malignancies. Cytotherapy 2012;14:830–40. 70. Gleason MK, Verneris MR, Todhunter DA, Zhang B, McCullar V, Zhou 51. Tettamanti S, Magnani CF, Biondi A, Biagi E. Acute myeloid leukemia SX, et al. Bispecific and trispecific killer cell engagers directly activate and novel biological treatments: monoclonal antibodies and cell- human NK cells through CD16 signaling and induce cytotoxicity and based gene-modified immune effectors. Immunol Lett 2013;155:43–6. cytokine production. Mol Cancer Ther 2012;11:2674–84. 52. Chang YH, Connolly J, Shimasaki N, Mimura K, Kono K, Campana D. A 71. Wu L, Adams M, Carter T, Chen R, Muller G, Stirling D, et al. lenali- chimeric receptor with NKG2D specificity enhances natural killer cell domide enhances natural killer cell and monocyte-mediated antibody- activation and killing of tumor cells. Cancer Res 2013;73:1777–86. dependent cellular cytotoxicity of rituximab-treated CD20þ tumor 53. Altvater B, Landmeier S, Pscherer S, Temme J, Schweer K, Kailayangiri cells. Clin Cancer Res 2008;14:4650–7. S, et al. 2B4 (CD244) signaling by recombinant antigen-specific chi- 72. Hsu AK, Quach H, Tai T, Prince HM, Harrison SJ, Trapani JA, et al. The meric receptors costimulates natural killer cell activation to leukemia immunostimulatory effect of lenalidomide on NK-cell function is pro- and neuroblastoma cells. Clin Cancer Res 2009;15:4857–66. foundly inhibited by concurrent dexamethasone therapy. Blood 54. Esser R, Muller T, Stefes D, Kloess S, Seidel D, Gillies SD, et al. NK cells 2011;117:1605–13. engineered to express a GD2-specific antigen receptor display built-in 73. Borg C, Terme M, Taieb J, Menard C, Flament C, Robert C, et al. Novel ADCC-like activity against tumour cells of neuroectodermal origin. J mode of action of c- tyrosine kinase inhibitors leading to NK cell- Cell Mol Med 2012;16:569–81. dependent antitumor effects. J Clin Invest 2004;114:379–88. 55. Rujkijyanont P, Chan WK, Eldridge PW, Lockey T, Holladay M, Rooney 74. Tanaka J, Sugita J, Shiratori S, Shigematsu A, Imamura M. Dasatinib B, et al. Ex vivo activation of CD56(þ) immune cells that eradicate enhances the expansion of CD56þCD3 NK cells from cord blood. neuroblastoma. Cancer Res 2013;73:2608–18. Blood 2012;119:6175–6. 56. Somanchi SS, Somanchi A, Cooper LJ, Lee DA. Engineering lymph 75. Peng Y, Luo G, Zhou J, Wang X, Hu J, Cui Y, et al. CD86 is an activation node homing of ex vivo–expanded human natural killer cells via receptor for NK cell cytotoxicity against tumor cells. PLoS ONE 2013;8: trogocytosis of the chemokine receptor CCR7. Blood 2012;119: e83913. 5164–72. 76. Beavis PA, Divisekera U, Paget C, Chow MT, John LB, Devaud C, et al. 57. Koehn TA, Trimble LL, Alderson KL, Erbe AK, McDowell KA, Grzywacz Blockade of A2A receptors potently suppresses the metastasis of B, et al. Increasing the clinical efficacy of NK and antibody-mediated CD73þ tumors. Proc Natl Acad Sci U S A 2013;110:14711–6. cancer immunotherapy: potential predictors of successful clinical 77. Krusch M, Salih J, Schlicke M, Baessler T, Kampa KM, Mayer F, et al. outcome based on observations in high-risk neuroblastoma. Front The kinase inhibitors sunitinib and sorafenib differentially affect NK cell Pharmacol 2012;3:91. antitumor reactivity in vitro. J Immunol 2009;183:8286–94. 58. Woll PS, Grzywacz B, Tian X, Marcus RK, Knorr DA, Verneris MR, et al. 78. Schmiedel BJ, Arelin V, Gruenebach F, Krusch M, Schmidt SM, Salih Human embryonic stem cells differentiate into a homogeneous pop- HR. Azacytidine impairs NK cell reactivity while decitabine augments ulation of natural killer cells with potent in vivo antitumor activity. Blood NK cell responsiveness toward stimulation. Int J Cancer 2011;128: 2009;113:6094–101. 2911–22. 59. Spanholtz J, Tordoir M, Eissens D, Preijers F, van der Meer A, Joosten I, 79. Diermayr S, Himmelreich H, Durovic B, Mathys-Schneeberger A, et al. High log-scale expansion of functional human natural killer cells Siegler U, Langenkamp U, et al. NKG2D ligand expression in AML from umbilical cord blood CD34-positive cells for adoptive cancer increases in response to HDAC inhibitor valproic acid and contributes immunotherapy. PLoS ONE 2010;5:e9221. to allorecognition by NK-cell lines with single KIR-HLA class I specifi- 60. Tonn T, Schwabe D, Klingemann HG, Becker S, Esser R, Koehl U, et al. cities. Blood 2008;111:1428–36. Treatment of patients with advanced cancer with the natural killer cell 80. Skov S, Pedersen MT, Andresen L, Straten PT, Woetmann A, Odum N. line NK-92. Cytotherapy 2013;15:1563–70. Cancer cells become susceptible to natural killer cell killing after 61. Reed W, Noga SJ, Gee AP, Rooney CM, Wagner JE, McCullough J, exposure to histone deacetylase inhibitors due to glycogen synthase et al. Production Assistance for Cellular Therapies (PACT): four-year kinase-3-dependent expression of MHC class I–related chain A and B. experience from the United States National Heart, Lung, and Blood Cancer Res 2005;65:11136–45. Institute (NHLBI) contract research program in cell and tissue thera- 81. Hallett WH, Ames E, Motarjemi M, Barao I, Shanker A, Tamang DL, pies. Transfusion 2009;49:786–96. et al. Sensitization of tumor cells to NK cell-mediated killing by 62. Koepsell SA, Kadidlo DM, Fautsch S, McCullough J, Klingemann H, proteasome inhibition. J Immunol 2008;180:163–70. Wagner JE, et al. Successful "in-flight" activation of natural killer cells 82. Lundqvist A, Abrams SI, Schrump DS, Alvarez G, Suffredini D, Berg during long-distance shipping. Transfusion 2013;53:398–403. M, et al. Bortezomib and depsipeptide sensitize tumors to tumor 63. Leung W. Immunotherapy in acute leukemia. Semin Hematol 2009;46: necrosis factor-related apoptosis-inducing ligand: a novel method 89–99. to potentiate natural killer cell tumor cytotoxicity. Cancer Res 64. Alderson KL, Sondel PM. Clinical cancer therapy by NK cells via 2006;66:7317–25. antibody-dependent cell-mediated cytotoxicity. J Biomed Biotechnol 83. Leung WH, Vong QP, Lin W, Janke L, Chen T, Leung W. Modulation of 2011;2011:379123. NKG2D ligand expression and metastasis in tumors by spironolactone 65. Cartron G, Dacheux L, Salles G, Solal-Celigny P, Bardos P, Colombat via RXRgamma activation. J Exp Med 2013;210:2675–92. P, et al. Therapeutic activity of humanized anti-CD20 monoclonal 84. Bellucci R, Nguyen HN, Martin A, Heinrichs S, Schinzel AC, Hahn WC, antibody and polymorphism in IgG FcgammaRIIIa gene. et al. Tyrosine kinase pathways modulate tumor susceptibility to Blood 2002;99:754–8. natural killer cells. J Clin Invest 2012;122:2369–83. 66. Kohrt HE, Houot R, Weiskopf K, Goldstein MJ, Scheeren F, Czerwinski 85. Klingemann HG. Cellular therapy of cancer with natural killer cells— D, et al. Stimulation of natural killer cells with a CD137-specific where do we stand? Cytotherapy 2013;15:1185–94.

www.aacrjournals.org Clin Cancer Res; 20(13) July 1, 2014 3399

Downloaded from clincancerres.aacrjournals.org on October 2, 2021. © 2014 American Association for Cancer Research. Leung

86. Koepsell SA, Miller JS, McKenna DH Jr. Natural killer cells: a review of mismatched NK cells for relapsed myeloma in the setting of autolo- manufacturing and clinical utility. Transfusion 2013;53:404–10. gous stem cell transplantation. Br J Haematol 2008;143:641–53. 87. Farnault L, Sanchez C, Baier C, Le Treut T, Costello RT. Hematological 92. Terme M, Ullrich E, Delahaye NF, Chaput N, Zitvogel L. Natural killer malignancies escape from NK cell innate immune surveillance: cell-directed therapies: moving from unexpected results to successful mechanisms and therapeutic implications. Clin Dev Immunol 2012; strategies. Nat Immunol 2008;9:486–94. 2012:421702. 93. Curti A, Ruggeri L, D'Addio A, Bontadini A, Dan E, Motta MR, et al. 88. Farhan S, Lee DA, Champlin RE, Ciurea SO. NK cell therapy: targeting Successful transfer of alloreactive haploidentical KIR ligand-mis- disease relapse after hematopoietic stem cell transplantation. Immu- matched natural killer cells after infusion in elderly high risk acute notherapy 2012;4:305–13. myeloid leukemia patients. Blood 2011;118:3273–9. 89. Koehl U, Sorensen J, Esser R, Zimmermann S, Gruttner HP, Tonn T, 94. Miller JS, Soignier Y, Panoskaltsis-Mortari A, McNearney SA, Yun GH, et al. IL-2 activated NK cell immunotherapy of three children after Fautsch SK, et al. Successful adoptive transfer and in vivo expansion of haploidentical stem cell transplantation. Blood Cells Mol Dis 2004; human haploidentical NK cells in patients with cancer. Blood 2005; 33:261–6. 105:3051–7. 90. Stern M, Passweg JR, Meyer-Monard S, Esser R, Tonn T, Soerensen J, 95. Geller MA, Cooley S, Judson PL, Ghebre R, Carson LF, Argenta PA, et al. et al. Pre-emptive immunotherapy with purified natural killer cells after A phase II study of allogeneic natural killer cell therapy to treat patients haploidentical SCT: a prospective phase II study in two centers. Bone with recurrent ovarian and breast cancer. Cytotherapy 2011;13:98–107. Marrow Transplant 2013;48:433–8. 96. Bachanova V, Burns LJ, McKenna DH, Curtsinger J, Panoskaltsis- 91. Shi J, Tricot G, Szmania S, Rosen N, Garg TK, Malaviarachchi PA, et al. Mortari A, Lindgren BR, et al. Allogeneic natural killer cells for refractory Infusion of haplo-identical killer immunoglobulin-like receptor ligand lymphoma. Cancer Immunol Immunother 2010;59:1739–44.

3400 Clin Cancer Res; 20(13) July 1, 2014 Clinical Cancer Research

Downloaded from clincancerres.aacrjournals.org on October 2, 2021. © 2014 American Association for Cancer Research. Infusions of Allogeneic Natural Killer Cells as Cancer Therapy

Wing Leung

Clin Cancer Res 2014;20:3390-3400.

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

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

Citing articles This article has been cited by 9 HighWire-hosted articles. Access the articles at: http://clincancerres.aacrjournals.org/content/20/13/3390.full#related-urls

E-mail alerts Sign up to receive free email-alerts related to this article or journal.

Reprints and To order reprints of this article or to subscribe to the journal, contact the AACR Publications Department at Subscriptions [email protected].

Permissions To request permission to re-use all or part of this article, use this link http://clincancerres.aacrjournals.org/content/20/13/3390. Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC) Rightslink site.

Downloaded from clincancerres.aacrjournals.org on October 2, 2021. © 2014 American Association for Cancer Research.