infusions of allogeneic natural killer cells as cancer therapy · the proposal that the resistance...

12
Review Infusions of Allogeneic Natural Killer Cells as Cancer Therapy Wing Leung 1,2 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. Ó2014 AACR. Disclosure of Potential Conicts 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, anti- bodies, and cellular therapies have been FDA approved or are in late-phase clinical trials. Infusions of purified natural killer (NK) cells are late- comers in cellular immunotherapy, compared with den- dritic cells (DC), lymphokine-activated killer (LAK) cells, tumor-infiltrating lymphocytes, and conventional cytotoxic T cells. Many investigators consider that the cytolytic activ- ity 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 HLA- haploidentical 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. Author's Afliations: 1 Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children's Research Hospital; and 2 Department of Pediatrics, University of Tennessee, Memphis, Tennessee Corresponding Author: Wing Leung, St. Jude Children's Research Hos- pital, 262 Danny Thomas Place, Memphis, TN 38105-3678. Phone: 901- 595-2617; Fax: 901-521-9005; E-mail: [email protected] doi: 10.1158/1078-0432.CCR-13-1766 Ó2014 American Association for Cancer Research. Clinical Cancer Research Clin Cancer Res; 20(13) July 1, 2014 3390 on May 12, 2021. © 2014 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Upload: others

Post on 22-Dec-2020

0 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Infusions of Allogeneic Natural Killer Cells as Cancer Therapy · The proposal that the resistance was related to recep-tors recognizing the absence of MHC class I arose from

Review

Infusions of Allogeneic Natural Killer Cells as CancerTherapy

Wing Leung1,2

AbstractNatural 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, andnovel augmentation techniques, to improveNK 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

humanNKcells openup anewarenaof 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 InterestW. 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' DisclosuresThe members of the planning committee have no real or apparent conflict of interest to disclose.

Learning ObjectivesUpon 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 SupportThis activity does not receive commercial support.

IntroductionFor many cancers, cure rates remain unacceptably low.

Advances in genetics and immunology have led to biologictherapies with non–cross-resistant mechanisms of actionand no overlapping toxicities. Cancer immunotherapy ispossible with an array of effector mechanisms involvinginnate and adaptive immunity (1). Various cytokines, anti-bodies, and cellular therapies have been FDA approved orare in late-phase clinical trials.

Infusions of purified natural killer (NK) cells are late-comers in cellular immunotherapy, compared with den-dritic cells (DC), lymphokine-activated killer (LAK) cells,tumor-infiltrating lymphocytes, and conventional cytotoxicT cells. Many investigators consider that the cytolytic activ-ity of LAK cells commonly used in the 1980s actuallyrepresents the activity of IL2-activated NK cells, althoughthe products contained polyclonal T cells. Biologically,murine NK cells have been known to kill malignant cellswithout T-cell assistance or prior sensitization since theirinitial description in 1975 (2–5); however, the therapeuticpotential of human NK cells was not revealed until theirbiology of target cell recognition was elucidated in the1990s and the antileukemia effect was observed in HLA-haploidentical hematopoietic cell transplantation (HCT) inthe early 2000s (6, 7). This review covers NK cell biologyrelevant to cancer immunotherapy and outlines avenuesbeing explored in clinical trials.

Author's Affiliations: 1Department of Bone Marrow Transplantation andCellular Therapy, St. Jude Children's Research Hospital; and 2Departmentof Pediatrics, University of Tennessee, Memphis, Tennessee

Corresponding Author: Wing Leung, St. Jude Children's Research Hos-pital, 262 Danny Thomas Place, Memphis, TN 38105-3678. Phone: 901-595-2617; Fax: 901-521-9005; E-mail: [email protected]

doi: 10.1158/1078-0432.CCR-13-1766

�2014 American Association for Cancer Research.

ClinicalCancer

Research

Clin Cancer Res; 20(13) July 1, 20143390

on May 12, 2021. © 2014 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Page 2: Infusions of Allogeneic Natural Killer Cells as Cancer Therapy · The proposal that the resistance was related to recep-tors recognizing the absence of MHC class I arose from

NK Cell BiologyNK cells were first named and characterized by Kiessling

at the Karolinska Institute (Stockholm, Sweden) and byHerberman at the National Cancer Institute (Bethesda,MD; refs. 2–5). The term "natural" referred to their naturalexistence in rodents and "killer" to their spontaneouskilling of lymphoma and leukemia cells in nonimmuneanimals. They were called "N-cells" or "K cells" by someinvestigators (3, 8). These cells were unique, as they lackedmarkers characteristic of B and T lymphocytes and werenonphagocytic, low-adhesive, and cytotoxic to IgG-coatedtargets.NK cells are generated in bone marrow and develop

normally in athymic mice and in humans with DiGeorgesyndrome. Induction of cytotoxicity is possible in NKcells within minutes, in contrast with the slower conven-tional induction of the T-cell immune response (9).Approximately 5% to 15% of human blood lymphocytesare NK cells (CD56þ, CD3/14/19�), 90% of which areCD56dim, with most of them being CD16þ, and they areresponsible for early innate immunity and antibody-dependent cell-mediated cytotoxicity (ADCC) againstinfection or cancer through IFNg , perforin, granzyme,FasL, or TRAIL (10). Approximately 10% of human bloodNK cells are CD56bright, and they participate in late (>16hours) inflammatory response by secreting IFNg , TNFa,G-CSF, GM-CSF, and IL3. Interactions between DCs andNK cells are essential for priming adaptive immunity. NKcells may also directly interact with T and B cells throughCD40 ligand and OX40 (11, 12).

Regulatory receptorsHumanNK cells are regulated by a sophisticatednetwork

of surface receptors (Fig. 1), allowing them to distinguishnormal from abnormal cells within seconds. Target celllysis occurs when the activating signal dominates theinhibitory signal (Fig. 2). Early studies revealed similaritiesbetweenmurineNK cells and cells responsible for so-called"hybrid resistance" to parental hematopoietic grafts (13,14). The proposal that the resistance was related to recep-tors recognizing the absence of MHC class I arose fromobservations that murine lymphoma cells that were low inMHC expression were NK-susceptible (15). This led to the"missing-self" hypothesis in the 1980s (16). Break-throughs in human NK receptor biology were seen in theearly 1990s, when the KIR gene family was found torecognize HLA class I (17–19). Identification of other NKreceptors, such as NCRs, NKG2D, DNAM, 2B4, and CD94/NKG2A, collectively points to a sophisticated network thatcontrols human NK activity.

Step 1: KIR Typing for Donor Selection andPrognosticationThree general steps in NK-based therapy are summarized

in Fig. 3. KIR typing is a critical first step because KIR ishighly polymorphic. KIR typing is not only useful forallogeneic donor selection but also for prognostication in

autologous NK therapy (20). Among known NK receptors,the KIR receptor family is one of the primary determinantsof NK response. Subsets of T cells also expressed KIR (21).Clinical KIR typing includes genotyping for gene contentand A/B haplotype categorization, phenotyping for geneexpression and number of KIRþ cells, and allelotyping forfunctional strength.

The first level of KIR diversity is gene content (22, 23).Only approximately 5% of people have all 15 gene familymembers; others lack one or more genes. The diversity ofgene content is based primarily on diversity in B haplo-types, which typically contain more activating KIRs. Thetwo hallmark genes for A haplotypes are KIR2DL3 andKIR3DL1. They are segregated with KIR2DL2 and KIR3DS1as alleles in the centromeric (Cen) and telomeric (Tel)motifs, respectively (Fig. 4A). On the basis of this pattern, asimplified typing and scoring method for B haplotypecontent 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 chromo-some 19 and ligandHLAs are in chromosome 6, these genefamilies are segregated independently; thus, HLA geno-types cannot be used to predict KIR content, and autolo-gous KIR-HLA mismatch is possible.

The second level of diversity is gene expression (24). Byreal-time PCR and flow cytometry, more than 10-foldvariability in expression has been observed among donors.Nowadays, the easiest method to quantify NK cells expres-sing only one inhibitory KIR gene (i.e., negative for allother MHC inhibitory receptors) is multicolor flow cyto-metry (24–26). The number of single KIRþ cells is in directproportion to activity against target cells without theligand (receptor–ligand mismatched). Although the num-ber of single KIRþ cells correlates with the presence of self-ligand (25), the variability among donors renders predic-tions based on the ligand alone unacceptable for clinicaluse.

The third level of diversity is allele polymorphism, whichhas been observed in all inhibitory KIR genes (22, 23). Forexample, in the KIR2DL1 family, 25 alleles have beenobserved, and each has different strengths in inhibiting NKcells and different durability of surface expression afterinteraction with ligands (27). These differences correlatewith the intensity of inhibitory signaling through SHP2 andb arrestin-2, resulting in differences in immune synapseformation. Mechanistically, the arginine residue at position245 in the transmembrane domain is important in inhibi-tion. Alleles with arginine in position 245 are stronger thanthosewith cysteine in that position (27).On the basis of thismolecular determinant, single-nucleotide polymorphismassays have been developed for rapid, high-throughputclinical typing (28). The same assay can be simultaneouslyused for KIR ligand typing. Notably, this assay requires lessthan 0.1 mg of DNA and can be performed within 1 day.Using this assay, investigations showed that patients whoreceived a donor graft containing the stronger KIR allele had

NK Infusions as Cancer Therapy

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

on May 12, 2021. © 2014 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Page 3: Infusions of Allogeneic Natural Killer Cells as Cancer Therapy · The proposal that the resistance was related to recep-tors recognizing the absence of MHC class I arose from

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

acute lymphoblastic leukemia (ALL)], total body irradia-tion, T-cell depletion, and donor type. Statistically, there isan interaction effectwithHLA-C receptor–ligandmismatch:

© 2014 American Association for Cancer Research

CCR Reviews

2B4 (CD48)

KIR2DL1/2/3 (HLA-C)

NK cell

Inhibitory Activating

Coreceptor

KIR3DL1 (HLA-B)

KIR3DL2 (HLA-A)

CD94/NKG2A (HLA-E)

LIR-1 (HLA-A–G)

KLRG-1 (cadherins)

CEACAM1 (CEACAM5)

TIGIT (PVR, PVRL2)

Siglec –3, –7, –9 (sialic acid)

LAIR-1 (collagen)

CD300A (PS) NKp80

(AICL)

NTBA (NTBA)

NKG2D (MIC, ULBP)

CD16 (IgG)

KIR3DL2 (CpG)

CD94/NKG2C (HLA-E)

IL1 IL2 IL12IL15 IL18 IL21

KIR2DL4 (HLA-G, HS)

KIR2DS4 (HLA-A, -C)

KIR2DS1/S2 (HLA-C)

IFNα

NCRs (B7-H6, NKp44L)

DNAM-1 (PVR, Nectin-2)

Figure 1. Surface receptors andtheir ligands. Cytokine receptorsare shown on top of a human NKcell. Other receptors are broadlyclassified and color-coded on thebasis of their primary function(inhibitory receptors in red,activating receptors in green,inhibitory coreceptors in red-blackstripes, and activating coreceptorsin green-black stripes). Theirligands are shown withinparentheses. Many other knownreceptors are not shown, includingchemotactic receptors (CCR-2, -5,-7: CXCR-1, -3, -4, -6; CX3CR1;and Chem23R), adhesionreceptors (CD2 and b1 and b2integrins), and activatingcoreceptors (CD96,CS1, and TLR).

© 2014 American Association for Cancer Research

CCR Reviews

NK cell

+

A

Target cellCharacteristics

of target cellOutcome

Normal MHC class I

No activating ligands

No MHC class I

No activating ligands

No MHC class I

activating ligands

Normal MHC class I

activating ligands

No response

No response

Attack target

Dynamic equilibrium

B

C

D

Figure 2. Dynamic equilibrium. Aftercell-to-cell contact, NK cellintegrates signals from its surfacereceptors in seconds, resulting ineither target-cell attack or noresponse and continualimmunosurveillance of other cells.A, healthy cells express normalamount of MHC class I ligands withno activating "stress" ligands. B,downregulation or absence ofMHCligand for cognate inhibitoryreceptors is insufficient to triggerNK cells. This happens in thephysiologic setting with red bloodcells and autologous KIR receptor–ligand mismatch cells, and inpathologic conditions with adultlymphoblastic leukemia. C,sufficient activating ligandsmust beexpressed on target cells to induceNK cell activity. D, if self-MHCligands are expressed in normalamount, the reactivity of the NK cellis ultimately dependent on thebalance of activating and inhibitorysignals. Successful NK cell therapyrelies on clinical strategies thatoptimize activation and avoidinhibition by cancer cells.

Leung

Clin Cancer Res; 20(13) July 1, 2014 Clinical Cancer Research3392

on May 12, 2021. © 2014 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Page 4: Infusions of Allogeneic Natural Killer Cells as Cancer Therapy · The proposal that the resistance was related to recep-tors recognizing the absence of MHC class I arose from

Patients with the best survival were those with a strongerKIR allele and receptor–ligand mismatch from donors. Thesecond-best group was those with a stronger KIR allele butno receptor–ligand mismatch. The worst survival was inthose with the weaker allele, regardless of mismatch.Donor KIR typing is not necessary in some KIRmismatch

models (Fig. 4C). The first model was proposed by thePerugia group (7); the ligand mismatch model requirestyping HLA in both donor and recipient. A donor is mis-matched if a ligand is present in the donor but absent in therecipient. This ligand mismatch model does not requiredonor or recipient KIR typing. The second model wasproposed by the Nantes group (30); in this model, KIR istyped in both donor and recipient. If a receptor is present in

the donor but absent in the recipient, the donor is mis-matched. Because B haplotypes contain more genes than Ahaplotypes, mismatch typically signifies a B haplotype inthe donor. The thirdmodel is the receptor–ligandmismatchmodel proposed by the Memphis group (31). This modelrequires donor KIR typing and recipient HLA typing.A donor is mismatched if an inhibitory receptor is presentin the donor but the ligand is absent in the recipient.Notably, the receptor–ligand mismatch model is the onlymodel applicable to autologous transplantation and anti-body therapy; studies have shown that the model is usefulfor predicting relapse in both therapeutic settings (32–34).Not all potential donors require all three levels of KIR typing(Fig. 4D).

© 2014 American Association for Cancer Research

CCR Reviews

Donor selection, including KIR typing

NK cell augmentation in vivoPatient

Donor

QA and QC

Best

NK cell purification and

processing ex vivo

B

A

C

TumorTumor

Tumor Tumor Tumor

Tumor

Tumor Tumor

2DL1

102

0

0

102

103

104

105

103

104

105

38.8

00

2DL2

2DL3

3DL1

Neg

H 2O

NK

NKNKNKNK

NK

NK

Aug

Aug

Aug

Aug

61.2

Figure 3. Three basic steps of allogeneic NK cell therapy. A, the first step includes donor selection based on health history and examination, infectiousbiomarkers, 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 cellpurification and processing to enrich NK cells and to improve NK cell functions. The process needs vigorous steps for quality assurance (QA) andquality control (QC). C, after cell infusion, antitumor activity could be enhanced in vivo using various augmentation agents (Aug).

NK Infusions as Cancer Therapy

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

on May 12, 2021. © 2014 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Page 5: Infusions of Allogeneic Natural Killer Cells as Cancer Therapy · The proposal that the resistance was related to recep-tors recognizing the absence of MHC class I arose from

© 2014 American Association for Cancer Research

CCR Reviews

3DL3 2DL3 2DL1 2DL4 3DL1 2DS4 3DL2

3DL22DL4 3DS1 2DL5A 2DS3/5 2DS1

3DP1

2DL2 2DL5B 2DS3/5 2DL1 3DP13DL3

Centromeric B scoreKIR2DL3

KIR2DL21 2

Does notexist

0Negative

Total B score = Centromeric B score + Telomeric B score = (________)*Total B scores range from 0–4

Negative

Positive

Positive

Telomeric B scoreKIR3DL1

KIR3DS11 2

Very rare0Negative

Negative

Positive

Positive

2DS2

B

ACen A

Cen B

Tel A

Tel B

Mismatch

model

Ligand

mismatch

Receptor

mismatch

Receptor–

ligand

mismatch

Relationship Test ordered

Donor HLA

recipient

HLA

Donor KIRL

vs.

recipient

KIRL

Donor KIR

vs.

recipient

KIR

Donor KIR

vs.

recipient

KIRL

Donor KIR2DL1 allelotyping

Recipient HLA typing

Receptor–ligand mismatch

Select donor with highest B score

Select donor with highest B scoreReceptor–ligand match

Select donor with largest amount of

receptor–ligand mismatched cells

Donor KIR phenotyping

by flow cytometryDonor KIR genotyping

Donor KIR genotyping

Missing ≥ 1 ligand

Not selectedC245+ R245–

BW4+ C1+ C2+

C245– R245+

Donor KIR

recipient

KIR

Donor KIR

recipient

HLA

Definition of

mismatch

Missing KIRL in

a recipient for a

KIRL that is

present in donor

Missing KIR in

a recipient for a

KIR that is

present in donor

Missing KIRL in

a recipient for a

KIR that is

present in donor

Example

Applicable to

antibody

therapy

Applicable to

autologous

cell therapy

No

Donor Recipient

C2

C1

2DS/Bx

2DL2

C2

C2

C2

Donor Recipient

Donor Recipient

No

No No

Yes Yes

C

D

Leung

Clin Cancer Res; 20(13) July 1, 2014 Clinical Cancer Research3394

on May 12, 2021. © 2014 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Page 6: Infusions of Allogeneic Natural Killer Cells as Cancer Therapy · The proposal that the resistance was related to recep-tors recognizing the absence of MHC class I arose from

Step 2: NK Cell Processing and QualityAssuranceNK cell purificationThe second step for therapy is NK cell processing and

preinfusion quality assessment, including cell count, via-bility, sterility, phenotype, function, and purity (35). Stud-ies in the 1980s with autologous LAK cells consistedprimarily of expanded polyclonal T cells with low NKpercentages (36). The easiest way to select highly purifiedNK cells is immunomagnetic cell separation (37). Afterseparation, 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–lymphoprolifera-tive disease, passenger lymphocyte syndrome, cytokinestorm, and suppression by myeloid suppressor cells orblood DCs are minimized (38–40). The NK phenotype isunaltered, and cells retain extensive proliferative capacityin vivo with potent antitumor response (41). Importantly,purified NK cells can be infused in small volumes, andstudies have shown that purified cells are safe, do not causeGVHD, and are detectable in the recipient’s blood forapproximately 2 to 4 weeks after infusion with preferentialexpansion of KIR-mismatched NK cells (42).

NK cell bioprocessing ex vivoDonor NK cells can be manipulated ex vivo using a

combination of strategies to optimize efficacy and spec-ificity (Fig. 5). One challenge has been NK exhaustion, asadoptively transferred cells rapidly lose activating recep-tor expression and IFNg production capability throughdownregulation of transcription factors T-bet and eome-sodermin (43). Furthermore, a highly acidic tumor envi-ronment may render NK cells nonfunctional (44). Acommon strategy to prime and activate cells is usingsoluble factors such as IL2, -12, -15, -18, and -21 or typeI IFN (45). Studies have shown that IFNa and IL2 syn-ergistically augment NK cells against solid tumors. Sim-ilarly, IL12 with either IL2 or IL18 can be used to over-come resistance (31). In cytokine-based culture systems,massive expansion of mature NK cells is possible. In astudy of 7 patients with newly diagnosed, untreatedmultiple myeloma, the number of NK cells expanded onaverage by 1,600-fold after 20 days of culture with con-siderable increase in cytotoxicity (46).Alternatively, NK cells could be stimulated using artificial

presenting cells with membrane-bound molecules such asIL15, IL21, and 41BB ligand (47, 48). Animal studies haveshown that these cells are potent against Ewing sarcoma,rhabdomyosarcoma, and neuroblastoma and showed pro-longed survival without GVHD.

Another way to increase NK potency is using chimericantigen receptor (CAR). Receptor insertion can bemediatedusing retroviral, lentiviral, mRNA, or Sleeping Beauty trans-poson/transposase systems (49–51). Investigations haveshown that NK cells transduced with anti-CD19 CAR orNKG2Dare potent against B-lineage and osteosarcoma cellsin vivo and in vitro (49, 52). CARþ cells have been generatedagainst GD2 in neuroblastoma and CD33/CD123 in mye-loid leukemia (51, 53, 54). Clinical trials are ongoing,including NCT00995137 and NCT01974479, for B-lineagehematologic malignancies.

For centers without gene modification capabilities, NKcells could be activated by culturing in the presence ofunmodifiedCD56� cells and stimulating cytokines. Recent-ly, investigators showed that this method expanded andactivated NK cells from donors and patients with neuro-blastoma (55). These cells have great efficacy toward neu-roblastoma 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 totumor sites. Recently, investigators used CCR7þ cells totransfer CCR7 onto NK cells via trogocytosis, resulting inbetter homing of NK cells to the tumor site in a mousemodel and in Transwell migration experiments (56).Another way to facilitate adhesion of NK cells to tumorcells is using immunocytokines such as antibody againstGD2 conjugated to IL2 (57). This chimeric protein attachesto GD2 on neuroblastoma cells on one side and to IL2receptor on NK cells on the other.

In addition to mature NK cells, human embryonic stemcells (hESC), induced pluripotent stem cells, and NK leu-kemia cell linesmay be used as starting cells (58–60). hESC-derived NK cells carry the CD94þCD117low/� phenotype,which has potent antitumor activity (58). Cytokine-basedculture systems, in particular, enhance the expansion ofNKG2D/NCRþ NK cells from umbilical cord blood (59).

Because of the technical difficulty in producing cellularproducts at treatment centers, theNationalHeart, Lung, andBlood Institute (NHLBI; Bethesda, MD) sponsored theProduction Assistance for Cellular Therapies program(PACT; ref. 61). Using this approach, apheresis cells havebeen sent to remote processing centers and purified, andactivated NK cells were sent to the transplant centers forinfusion into patients (62).

Step 3: NK Augmentation In VivoCombination with therapeutic antibody or chimericproteins

Onemechanismof antibodies in cancer therapy is ADCC.NK cells carry high-affinity Fc receptors that mediate thisfunction. 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 thatare 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 totalB scores. C, all three mismatch models are biologically sound but are fundamentally different in principles and in clinical usages, including therelationship 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.

NK Infusions as Cancer Therapy

www.aacrjournals.org Clin Cancer Res; 20(13) July 1, 2014 3395

on May 12, 2021. © 2014 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Page 7: Infusions of Allogeneic Natural Killer Cells as Cancer Therapy · The proposal that the resistance was related to recep-tors recognizing the absence of MHC class I arose from

attractive to administer NK therapy concurrently with anti-bodies such as those against CD19, CD20, CD22, CD33,CD123, HER2, EGFR, and GD2 (63, 64). Typing for recip-ient and donor FcgRIII 158V/F polymorphism is importantin optimizing ADCC (65). Stimulating NK cells with anagonistic monoclonal antibody specific for CD137 or withblocking antibodies against KIR/NKG2A may increase cellkilling (66–68). Thus, using a second antibody that acti-vates donor NK cells may improve the efficacy of antibodiesagainst tumor-associated antigens.

Another method to activate CD16 is a bispecific or tri-specific killer cell engager (BiKEor TriKE). A fully humanizedCD16-CD33 BiKE has been created that strongly activatesNK cells against CD33þ AML blasts (69). Pretreatment withADAM17 inhibitor prevented CD16 shedding and overcameinhibition of class I MHC-recognizing inhibitory receptors.Incubating cancer cells with BiKE and TriKE increased NKcytolytic activity against tumor targets and production ofIFNg , TNFa, GM-CSF, IL8, MIP-1a, and RANTES (70).

Combination with supplemental medicationAfter NK infusion, many regimens include low-dose

IL2 injections. Newer agents are being investigated. Lena-lidomide enhances ADCC in rituximab treatment ofnon-Hodgkin lymphoma and B-cell chronic lymphocyticleukemia through enhanced granzyme B and FasL expres-sion (71). The effects of lenalidomide are partly relatedto CD4þ T-cell production of IL2 (72). Imatinib triggersDC-mediated NK activation (73), whereas dasatinibenhances NK expansion via eomesodermin (74). Ligationof CD86 on NK cells with CTLA4-Ig fusion protein orblockade of A2A adenosine receptor interaction withCD73 increases tumor killing (75, 76).

During NK therapy, concurrent suppressive medicationsshould be avoided, including agents such as azacytidine andsorafenib, which substantially impair PI3K and ERK phos-phorylation and hamper NK reactivity (77, 78). Sunitinibdoes not inhibit NK cells, and decitabine may augment NKreactivity (78).

© 2014 American Association for Cancer Research

CCR Reviews

NK cells

derived from

ESC/IPS

Tumor

• HDAC inhibitor

• ADAM inhibitor

• Proteasome

inhibitor

• Demethylation

• RXRγ agonist

• DNA damage

• JAK inhibitor

• ↑ 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)

Tumor

IFNγ

CD8+

CTL

CD4+

Th

B cell

Tumor Ag uptake

and presentation

Tumor-specific

adaptive immunity

Antitumor

antibody

DC editing

and maturationLysis

aNK iDC

mDC

mNKNKpHSCESC

IPS

Selection of KIR-

favorable

donor

Ex vivo NKp

development and

mNK expansion

mNK cell

purification

and activation

Sensitization

strategies

Augmentation strategies

Figure 5. Clinically feasibleapproaches to optimize NK celltherapy.Most protocols usematureNK cells (mNK) as starting cells,while a few use stem or progenitorcells such as ESC, inducedpluripotent stem cells (IPS), andhematopoietic stem cells (HSC) togenerate NK precursors (NKp). NKcells can be augmented throughvarious strategies to obtain largenumbers of activated NK cells(aNK). After infusion, the NK cellsmay kill cancer cells directly ormediate the development ofadaptive immunity via interactionswith immature DCs (iDC), B cells,T-helper cells (Th), cytotoxic Tlymphocytes (CTL), and matureDCs (mDC). Tumor cells can besensitized by various agents torender them more susceptible toNK cells. Bold arrows indicate cellintrinsic changes. Thin arrowsindicate cell extrinsic interactions.Ag, antigen.

Leung

Clin Cancer Res; 20(13) July 1, 2014 Clinical Cancer Research3396

on May 12, 2021. © 2014 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Page 8: Infusions of Allogeneic Natural Killer Cells as Cancer Therapy · The proposal that the resistance was related to recep-tors recognizing the absence of MHC class I arose from

Combination with tumor cell modulationAfter CD16, NKG2D is the secondmost potent activating

receptor expressed on NK cells. Unfortunately, NKG2Dligand expression in most cancer cells is low (79). Recently,various methods have been used to upregulate NKG2Dligand expression or prevent its shedding from the cellsurface, including epigenetic modulation with histone dea-cetylase (HDAC) inhibitors (which may induce glycogensynthase kinase-3 activity; ref. 80), proteosome inhibitors,and demethylating agents. These agents may also worktogether to induce expression of Fas and TRAIL-R2 (DR5;refs. 81, 82). Recently, high-throughput screening of 5,600bioactive compounds revealed spironolactone as a novelRXRg agonist that activates the ATM–CHK2 pathway toupregulate transcription of all classes of NKG2D ligandsfor cancer prevention and control (83). In another studyusing a lentiviral shRNA library targeting more than 10,000human genes, silencing of JAK1 and JAK2 increasedthe susceptibility of a variety of tumor cell types to NK-mediated lysis (84).

Clinical ConsiderationsNK cell therapy is applicable to various forms of cancer

(85, 86), but its current use has been focused primarily inhematologic malignancies (20, 63, 87, 88). NK cells can beused for patients with refractory leukemia and can be givenbefore conventional HCT for induction of remission, afterHCT 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 persis-tent leukemia blasts at the time of transplant (89). Nosignificant toxicity was observed, and complete remissionand complete donor chimerism were observed within 1month after HCT. In a subsequent prospective phase IIstudy, preemptive immunotherapy with purified NK cellsafter haploidentical HCT was investigated in 16 patients(90). The patients received 29 NK infusions 3, 40, and100 days after transplantation. The median dose of NKcells per product was 1.21 � 107/kg. With a median fol-low-up of 5.8 years, 4 of the 16 patients with high-riskleukemia remained alive.In 10patientswith advancedmultiplemyeloma receiving

autologous HCT, haploidentical T cell–depleted, KIRligand–mismatched NK cells were infused (91). No GVHDor failure of autologous stem cell reconstitution wasobserved. Encouragingly, 50%of the patients achieved nearcomplete remission. These results set the stage for futurestudies of KIR ligand–mismatched NK therapy in the autol-ogous setting.

NK therapy without HCTFor allogeneic NK therapy in a nontransplant setting,

transient suppression of the host immune system isrequired to allow time for donor-derived NK expansionand therapeutic effect. Even in the autologous setting,induction of transient leukopenia may promote homeo-static expansion of NK cells and reduce suppressive effectsfrom Tregs and myeloid suppressor cells (32, 33, 92). Onecommon outpatient regimen includes fludarabine andlow-dose cyclophosphamide, and this regimen is welltolerated even in children, with 10 of 10 patients survivingfree of leukemia (42). The rare occurrence of complica-tions results in the cost of fresh NK therapy being lessthan 10% of that for conventional HCT. Infusions ofpurified NK cells were feasible after similar conditioningin 13 elderly patients including septuagenarians (93).

In a study of haploidentical related-donor NK infusion, ahigh-intensity inpatient regimen of high-dose cyclophos-phamide and fludarabine resulted in a marked increase inendogenous IL15, expansion of donor NK cells, and induc-tion of complete hematologic remission in 5 of 19 patientswith poor-prognosis AML (94). In a subsequent phase IIstudy for recurrent ovarian and breast cancer, 20 patientsunderwent the regimen of fludarabine and cyclophospha-mide with or without low-dose total body irradiation (95).After cell infusion, IL2was administered subcutaneously for2 weeks. With a mean NK cell dose of 2.16� 107 cells/kg, 9of 13 (69%) patients without irradiation and 6 of 7 withirradiation had donor DNA detectable after NK cell infu-sions. In another study of 6 patients with advanced B-cellnon-Hodgkin lymphoma, 4 patients showed an objectiveclinical response (96). Unfortunately, Tregs also expandedin vivo in these two studies; therefore, future strategiesshould include novel techniques to suppress the expansionof Tregs.

ConclusionsIn summary, NK cells are promising in cancer therapy,

considering their speed of action, potency in cancer cellkilling, applicability tomany types of cancer, lack of adverseeffects, ease of preparation and administration, availability,permissibility of donor selection, affordability, and com-plimentary actions with other therapies. The unique prop-erties of NK cells open a new arena of novel cell-basedimmunotherapy against cancers that are resistant to con-temporary therapies.

Received October 29, 2013; revised February 6, 2014; accepted February25, 2014; published online July 1, 2014.

References1. 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.

2. Kiessling R, Klein E, Pross H, Wigzell H. "Natural" killer cells in themouse. II. Cytotoxic cells with specificity for mouseMoloney leukemiacells. Characteristics of the killer cell. Eur J Immunol 1975;5:117–21.

3. Herberman RB, Nunn ME, Holden HT, Lavrin DH. Natural cytotoxicreactivity of mouse lymphoid cells against syngeneic and allogeneictumors. II. Characterization of effector cells. Int JCancer 1975;16:230–9.

4. Herberman RB, Nunn ME, Lavrin DH. Natural cytotoxic reactivity ofmouse lymphoid cells against syngeneic acid allogeneic tumors. I.Distribution of reactivity and specificity. Int J Cancer 1975;16:216–29.

NK Infusions as Cancer Therapy

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

on May 12, 2021. © 2014 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Page 9: Infusions of Allogeneic Natural Killer Cells as Cancer Therapy · The proposal that the resistance was related to recep-tors recognizing the absence of MHC class I arose from

5. 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 Immunol1975;5:112–7.

6. Valiante NM, Parham P. Natural killer cells, HLA class I molecules, andmarrow transplantation. Biol Blood Marrow Transplant 1997;3:229–35.

7. Ruggeri L, Capanni M, Urbani E, Perruccio K, Shlomchik WD, Tosti A,et al. Effectiveness of donor natural killer cell alloreactivity in mis-matched hematopoietic transplants. Science 2002;295:2097–100.

8. MacLennan IC, Connell GE, Gotch FM. Effector activating determi-nants on IgG. II. Differentiation of the combining sites for C1q fromthose for cytotoxic K cells and neutrophils by plasmin digestion ofrabbits IgG. Immunology 1974;26:303–10.

9. Liu D, Xu L, Yang F, Li D, Gong F, Xu T. Rapid biogenesis andsensitization of secretory lysosomes in NK cells mediated by target-cell recognition. Proc Natl Acad Sci U S A 2005;102:123–7.

10. DeMaria A, Bozzano F, Cantoni C,Moretta L. Revisiting human naturalkiller cell subset function revealed cytolytic CD56(dim)CD16þNK cellsas rapid producers of abundant IFN-gamma on activation. Proc NatlAcad Sci U S A 2010;108:728–32.

11. Li S, Yan Y, Lin Y, Bullens DM, Rutgeerts O, Goebels J, et al. Rapidlyinduced, T-cell independent xenoantibody production is mediated bymarginal zone B cells and requires help from NK cells. Blood 2007;110:3926–35.

12. Croft M. Control of immunity by the TNFR-related molecule OX40(CD134). Annu Rev Immunol 2010;28:57–78.

13. Cudkowicz G, Bennett M. Peculiar immunobiology of bone marrowallografts. II. Rejection of parental grafts by resistant F 1 hybrid mice.J Exp Med 1971;134:1513–28.

14. Cudkowicz G, Stimpfling JH. Hybrid resistance to parental marrowgrafts: association with the K region of H-2. Science 1964;144:1339–40.

15. KarreK, LjunggrenHG,PiontekG,KiesslingR. Selective rejection ofH-2-deficient lymphoma variants suggests alternative immune defencestrategy. Nature 1986;319:675–8.

16. Ljunggren HG, Karre K. In search of the 'missing self': MHCmoleculesand NK cell recognition. Immunol Today 1990;11:237–44.

17. Colonna M, Samaridis J. Cloning of immunoglobulin-superfamilymembers associated with HLA-C and HLA-B recognition by humannatural killer cells. Science 1995;268:405–8.

18. D'Andrea A, Chang C, Franz-Bacon K, McClanahan T, Phillips JH,Lanier LL. Molecular cloning of NKB1. A natural killer cell receptor forHLA-B allotypes. J Immunol 1995;155:2306–10.

19. Wagtmann N, Biassoni R, Cantoni C, Verdiani S, Malnati MS, Vitale M,et al. Molecular clones of the p58 NK cell receptor reveal immuno-globulin-related molecules with diversity in both the extra- and intra-cellular domains. Immunity 1995;2:439–49.

20. Leung W. Use of NK cell activity in cure by transplant. Br J Haematol2011;155:14–29.

21. Chan WK, Rujkijyanont P, Neale G, Yang J, Bari R, Das Gupta N, et al.Multiplex and genome-wide analyses reveal distinctive properties ofKIRþ and CD56þ T cells in human blood. J Immunol 2013;191:1625–36.

22. PyoCW,Guethlein LA, VuQ,WangR, Abi-Rached L, Norman PJ, et al.Different patterns of evolution in the centromeric and telomeric regionsof group A and B haplotypes of the human killer cell Ig-like receptorlocus. PLoS ONE 2010;5:e15115.

23. Parham P, Moffett A. Variable NK cell receptors and their MHC class Iligands in immunity, reproduction and human evolution. Nat RevImmunol 2013;13:133–44.

24. LeungW, Iyengar R, Triplett B, Turner V, Behm FG, Holladay MS, et al.Comparison of killer Ig-like receptor genotyping and phenotyping forselection of allogeneic blood stem cell donors. J Immunol 2005;174:6540–5.

25. Shilling HG, Young N, Guethlein LA, Cheng NW, Gardiner CM, Tyan D,et al. Genetic control of human NK cell repertoire. J Immunol2002;169:239–47.

26. Pende D,Marcenaro S, FalcoM,Martini S, BernardoME,Montagna D,et al. Anti-leukemia activity of alloreactive NK cells in KIR ligand-

mismatched haploidentical HSCT for pediatric patients: evaluation ofthe functional role of activating KIR and redefinition of inhibitory KIRspecificity. Blood 2009;113:3119–29.

27. Bari R, Bell T, Leung WH, Vong QP, Chan WK, Das Gupta N, et al.Significant functional heterogeneity among KIR2DL1 alleles and apivotal role of arginine 245. Blood 2009;114:5182–90.

28. 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.

29. Bari R, Rujkijyanont P, Sullivan E, Kang G, Turner V, Gan K, et al. Effectof donor KIR2DL1 allelic polymorphism on the outcome of pediatricallogeneic hematopoietic stem-cell transplantation. J Clin Oncol2013;31:3782–90.

30. Gagne K, Brizard G, Gueglio B, Milpied N, Herry P, Bonneville F, et al.Relevance of KIRgenepolymorphisms in bonemarrow transplantationoutcome. Hum Immunol 2002;63:271–80.

31. Leung W, Iyengar R, Turner V, Lang P, Bader P, Conn P, et al.Determinants of antileukemia effects of allogeneic NK cells. J Immunol2004;172:644–50.

32. LeungW,Handgretinger R, Iyengar R, Turner V, HolladayMS,HaleGA.Inhibitory KIR-HLA receptor-ligand mismatch in autologous haema-topoietic stemcell transplantation for solid tumour and lymphoma. Br JCancer 2007;97:539–42.

33. Venstrom JM, Zheng J, Noor N, Danis KE, Yeh AW, Cheung IY, et al.KIR and HLA genotypes are associated with disease progression andsurvival following autologous hematopoietic stem cell transplantationfor high-risk neuroblastoma. Clin Cancer Res 2009;15:7330–4.

34. Delgado DC, Hank JA, Kolesar J, Lorentzen D, Gan J, Seo S, et al.Genotypes of NK cell KIR receptors, their ligands, and Fcgammareceptors in the response of neuroblastoma patients to Hu14.18-IL2immunotherapy. Cancer Res 2010;70:9554–61.

35. Koehl U, BrehmC,HueneckeS, ZimmermannSY, Kloess S, BremmM,et al. Clinical grade purification and expansion of NK cell products foran optimized manufacturing protocol. Front Oncol 2013;3:118.

36. Rosenberg SA, Lotze MT, Muul LM, Leitman S, Chang AE, Ettinghau-sen SE, et al. Observations on the systemic administration of autol-ogous lymphokine-activated killer cells and recombinant interleukin-2to patients with metastatic cancer. N Engl J Med 1985;313:1485–92.

37. Iyengar R, Handgretinger R, Babarin-Dorner A, Leimig T, Otto M,Geiger TL, et al. Purification of human natural killer cells using aclinical-scale immunomagnetic method. Cytotherapy 2003;5:479–84.

38. Smyth MJ, Teng MW, Swann J, Kyparissoudis K, Godfrey DI, Haya-kawa Y. CD4þCD25þ T regulatory cells suppress NK cell-mediatedimmunotherapy of cancer. J Immunol 2006;176:1582–7.

39. Perez-Martinez A, Iyengar R, Gan K, Chotsampancharoen T, RooneyB,HolladayM, et al. Blooddendritic cells suppressNKcell function andincrease the risk of leukemia relapse after hematopoietic cell trans-plantation. Biol Blood Marrow Transplant 2011;17:598–607.

40. Shook DR, LeungW. Natural killer cell therapy for cancer: delivering ona promise. Transfusion 2013;53:245–8.

41. LeungW, Iyengar R, Leimig T, HolladayMS, Houston J, HandgretingerR. Phenotype and function of human natural killer cells purified byusing a clinical-scale immunomagnetic method. Cancer ImmunolImmunother 2005;54:389–94.

42. 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 hap-loidentical natural killer cell transplantation in childhood acute myeloidleukemia. J Clin Oncol 2010;28:955–9.

43. Gill S, Vasey AE, De Souza A, Baker J, Smith AT, Kohrt HE, et al. Rapiddevelopment of exhaustion and down-regulation of eomesoderminlimit the antitumor activity of adoptively transferredmurine natural killercells. Blood 2012;119:5758–68.

44. Helmlinger G, Yuan F, Dellian M, Jain RK. Interstitial pH and pO2

gradients in solid tumors in vivo: high-resolution measurements reveala lack of correlation. Nat Med 1997;3:177–82.

45. Moretta A, Ciccone E, Pantaleo G, Tambussi G, Bottino C, Melioli G,et al. Surfacemolecules involved in the activation and regulation of T ornatural killer lymphocytes in humans. Immunol Rev 1989;111:145–75.

46. Alici E, Sutlu T, Bjorkstrand B, Gilljam M, Stellan B, Nahi H, et al.Autologous antitumor activity by NK cells expanded from myeloma

Leung

Clin Cancer Res; 20(13) July 1, 2014 Clinical Cancer Research3398

on May 12, 2021. © 2014 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Page 10: Infusions of Allogeneic Natural Killer Cells as Cancer Therapy · The proposal that the resistance was related to recep-tors recognizing the absence of MHC class I arose from

patients using GMP-compliant components. Blood 2008;111:3155–62.

47. Fujisaki H, Kakuda H, Shimasaki N, Imai C, Ma J, Lockey T, et al.Expansion of highly cytotoxic human natural killer cells for cancer celltherapy. Cancer Res 2009;69:4010–7.

48. Denman CJ, Senyukov VV, Somanchi SS, Phatarpekar PV, Kopp LM,Johnson JL, et al. Membrane-bound IL-21 promotes sustained ex vivoproliferation of human natural killer cells. PLoS ONE 2012;7:e30264.

49. Imai C, Iwamoto S, Campana D. Genetic modification of primarynatural killer cells overcomes inhibitory signals and induces specifickilling of leukemic cells. Blood 2005;106:376–83.

50. Shimasaki N, Fujisaki H, Cho D,Masselli M, Lockey T, Eldridge P, et al.A clinically adaptable method to enhance the cytotoxicity of naturalkiller cells against B-cell malignancies. Cytotherapy 2012;14:830–40.

51. Tettamanti S, Magnani CF, Biondi A, Biagi E. Acute myeloid leukemiaand novel biological treatments: monoclonal antibodies and cell-based gene-modified immune effectors. Immunol Lett 2013;155:43–6.

52. ChangYH,Connolly J, Shimasaki N,MimuraK, KonoK,CampanaD. Achimeric receptor with NKG2D specificity enhances natural killer cellactivation and killing of tumor cells. Cancer Res 2013;73:1777–86.

53. Altvater B, Landmeier S, Pscherer S, TemmeJ, SchweerK,KailayangiriS, et al. 2B4 (CD244) signaling by recombinant antigen-specific chi-meric receptors costimulates natural killer cell activation to leukemiaand neuroblastoma cells. Clin Cancer Res 2009;15:4857–66.

54. Esser R,Muller T, StefesD,Kloess S, Seidel D,Gillies SD, et al. NK cellsengineered to express a GD2-specific antigen receptor display built-inADCC-like activity against tumour cells of neuroectodermal origin. JCell Mol Med 2012;16:569–81.

55. Rujkijyanont P, ChanWK, Eldridge PW, Lockey T, HolladayM, RooneyB, et al. Ex vivo activation of CD56(þ) immune cells that eradicateneuroblastoma. Cancer Res 2013;73:2608–18.

56. Somanchi SS, Somanchi A, Cooper LJ, Lee DA. Engineering lymphnode homing of ex vivo–expanded human natural killer cells viatrogocytosis of the chemokine receptor CCR7. Blood 2012;119:5164–72.

57. Koehn TA, Trimble LL, Alderson KL, Erbe AK, McDowell KA, GrzywaczB, et al. Increasing the clinical efficacy of NK and antibody-mediatedcancer immunotherapy: potential predictors of successful clinicaloutcome based on observations in high-risk neuroblastoma. FrontPharmacol 2012;3:91.

58. Woll PS, Grzywacz B, Tian X, Marcus RK, Knorr DA, Verneris MR, et al.Human embryonic stem cells differentiate into a homogeneous pop-ulation of natural killer cells with potent in vivo antitumor activity. Blood2009;113:6094–101.

59. Spanholtz J, TordoirM, EissensD, Preijers F, vanderMeerA, Joosten I,et al. High log-scale expansion of functional human natural killer cellsfrom umbilical cord blood CD34-positive cells for adoptive cancerimmunotherapy. PLoS ONE 2010;5:e9221.

60. Tonn T, SchwabeD, KlingemannHG, Becker S, Esser R, Koehl U, et al.Treatment of patients with advanced cancer with the natural killer cellline NK-92. Cytotherapy 2013;15:1563–70.

61. Reed W, Noga SJ, Gee AP, Rooney CM, Wagner JE, McCullough J,et al. Production Assistance for Cellular Therapies (PACT): four-yearexperience from the United States National Heart, Lung, and BloodInstitute (NHLBI) contract research program in cell and tissue thera-pies. Transfusion 2009;49:786–96.

62. Koepsell SA, Kadidlo DM, Fautsch S, McCullough J, Klingemann H,Wagner JE, et al. Successful "in-flight" activation of natural killer cellsduring long-distance shipping. Transfusion 2013;53:398–403.

63. LeungW. Immunotherapy in acute leukemia. Semin Hematol 2009;46:89–99.

64. Alderson KL, Sondel PM. Clinical cancer therapy by NK cells viaantibody-dependent cell-mediated cytotoxicity. J Biomed Biotechnol2011;2011:379123.

65. Cartron G, Dacheux L, Salles G, Solal-Celigny P, Bardos P, ColombatP, et al. Therapeutic activity of humanized anti-CD20 monoclonalantibody and polymorphism in IgG Fc receptor FcgammaRIIIa gene.Blood 2002;99:754–8.

66. Kohrt HE, Houot R,Weiskopf K, GoldsteinMJ, Scheeren F, CzerwinskiD, et al. Stimulation of natural killer cells with a CD137-specific

antibody enhances trastuzumab efficacy in xenotransplant models ofbreast cancer. J Clin Invest 2012;122:1066–75.

67. VeyN, 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 incomplete remission. Blood 2012;120:4317–23.

68. Benson DM Jr, Hofmeister CC, Padmanabhan S, Suvannasankha A,Jagannath S, Abonour R, et al. A phase 1 trial of the anti-KIR antibodyIPH2101 in patients with relapsed/refractory multiple myeloma. Blood2012;120:4324–33.

69. Wiernik A, FoleyB, ZhangB, VernerisMR,Warlick E, GleasonMK, et al.Targeting natural killer cells to acute myeloid leukemia in vitro with aCD16 � 33 bispecific killer cell engager and ADAM17 inhibition. ClinCancer Res 2013;19:3844–55.

70. Gleason MK, Verneris MR, Todhunter DA, Zhang B, McCullar V, ZhouSX, et al. Bispecific and trispecific killer cell engagers directly activatehuman NK cells through CD16 signaling and induce cytotoxicity andcytokine production. Mol Cancer Ther 2012;11:2674–84.

71. Wu L, Adams M, Carter T, Chen R, Muller G, Stirling D, et al. lenali-domide enhances natural killer cell andmonocyte-mediated antibody-dependent cellular cytotoxicity of rituximab-treated CD20þ tumorcells. Clin Cancer Res 2008;14:4650–7.

72. Hsu AK, Quach H, Tai T, Prince HM, Harrison SJ, Trapani JA, et al. Theimmunostimulatory effect of lenalidomide on NK-cell function is pro-foundly inhibited by concurrent dexamethasone therapy. Blood2011;117:1605–13.

73. Borg C, TermeM, Taieb J, Menard C, Flament C, Robert C, et al. Novelmode of action of c-kit tyrosine kinase inhibitors leading to NK cell-dependent antitumor effects. J Clin Invest 2004;114:379–88.

74. Tanaka J, Sugita J, Shiratori S, Shigematsu A, Imamura M. Dasatinibenhances the expansion of CD56þCD3� NK cells from cord blood.Blood 2012;119:6175–6.

75. PengY, LuoG, Zhou J,Wang X, Hu J, Cui Y, et al. CD86 is an activationreceptor forNKcell cytotoxicity against tumor cells. PLoSONE2013;8:e83913.

76. Beavis PA, Divisekera U, Paget C, ChowMT, John LB, DevaudC, et al.Blockade of A2A receptors potently suppresses the metastasis ofCD73þ tumors. Proc Natl Acad Sci U S A 2013;110:14711–6.

77. Krusch M, Salih J, Schlicke M, Baessler T, Kampa KM, Mayer F, et al.The kinase inhibitors sunitinib and sorafenib differentially affect NK cellantitumor reactivity in vitro. J Immunol 2009;183:8286–94.

78. Schmiedel BJ, Arelin V, Gruenebach F, Krusch M, Schmidt SM, SalihHR. Azacytidine impairs NK cell reactivity while decitabine augmentsNK cell responsiveness toward stimulation. Int J Cancer 2011;128:2911–22.

79. Diermayr S, Himmelreich H, Durovic B, Mathys-Schneeberger A,Siegler U, Langenkamp U, et al. NKG2D ligand expression in AMLincreases in response to HDAC inhibitor valproic acid and contributesto allorecognition by NK-cell lines with single KIR-HLA class I specifi-cities. Blood 2008;111:1428–36.

80. Skov S, PedersenMT, Andresen L, Straten PT,Woetmann A, OdumN.Cancer cells become susceptible to natural killer cell killing afterexposure to histone deacetylase inhibitors due to glycogen synthasekinase-3-dependent expression of MHC class I–related chain A and B.Cancer Res 2005;65:11136–45.

81. Hallett WH, Ames E, Motarjemi M, Barao I, Shanker A, Tamang DL,et al. Sensitization of tumor cells to NK cell-mediated killing byproteasome inhibition. J Immunol 2008;180:163–70.

82. Lundqvist A, Abrams SI, Schrump DS, Alvarez G, Suffredini D, BergM, et al. Bortezomib and depsipeptide sensitize tumors to tumornecrosis factor-related apoptosis-inducing ligand: a novel methodto potentiate natural killer cell tumor cytotoxicity. Cancer Res2006;66:7317–25.

83. LeungWH, Vong QP, Lin W, Janke L, Chen T, LeungW. Modulation ofNKG2D ligand expression andmetastasis in tumors by spironolactonevia RXRgamma activation. J Exp Med 2013;210:2675–92.

84. Bellucci R, Nguyen HN, Martin A, Heinrichs S, Schinzel AC, HahnWC,et al. Tyrosine kinase pathways modulate tumor susceptibility tonatural killer cells. J Clin Invest 2012;122:2369–83.

85. Klingemann HG. Cellular therapy of cancer with natural killer cells—where do we stand? Cytotherapy 2013;15:1185–94.

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

NK Infusions as Cancer Therapy

on May 12, 2021. © 2014 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Page 11: Infusions of Allogeneic Natural Killer Cells as Cancer Therapy · The proposal that the resistance was related to recep-tors recognizing the absence of MHC class I arose from

86. Koepsell SA, Miller JS, McKenna DH Jr. Natural killer cells: a review ofmanufacturing and clinical utility. Transfusion 2013;53:404–10.

87. Farnault L, Sanchez C, Baier C, Le Treut T, Costello RT. Hematologicalmalignancies 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: targetingdisease relapse after hematopoietic stem cell transplantation. Immu-notherapy 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 afterhaploidentical stem cell transplantation. Blood Cells Mol Dis 2004;33:261–6.

90. SternM,Passweg JR,Meyer-MonardS, Esser R, Tonn T, Soerensen J,et al. Pre-emptive immunotherapy with purified natural killer cells afterhaploidentical SCT: a prospective phase II study in two centers. BoneMarrow Transplant 2013;48:433–8.

91. Shi J, Tricot G, Szmania S, RosenN,Garg TK,Malaviarachchi PA, et al.Infusion of haplo-identical killer immunoglobulin-like receptor ligand

mismatched NK cells for relapsed myeloma in the setting of autolo-gous stem cell transplantation. Br J Haematol 2008;143:641–53.

92. Terme M, Ullrich E, Delahaye NF, Chaput N, Zitvogel L. Natural killercell-directed therapies: moving from unexpected results to successfulstrategies. Nat Immunol 2008;9:486–94.

93. Curti A, Ruggeri L, D'Addio A, Bontadini A, Dan E, Motta MR, et al.Successful transfer of alloreactive haploidentical KIR ligand-mis-matched natural killer cells after infusion in elderly high risk acutemyeloid leukemia patients. Blood 2011;118:3273–9.

94. Miller JS, Soignier Y, Panoskaltsis-Mortari A, McNearney SA, Yun GH,FautschSK, et al. Successful adoptive transfer and in vivoexpansionofhuman haploidentical NK cells in patients with cancer. Blood 2005;105:3051–7.

95. GellerMA,CooleyS, JudsonPL,GhebreR,CarsonLF, ArgentaPA, et al.A phase II study of allogeneic natural killer cell therapy to treat patientswith recurrent ovarian and breast cancer. Cytotherapy 2011;13:98–107.

96. Bachanova V, Burns LJ, McKenna DH, Curtsinger J, Panoskaltsis-Mortari A, LindgrenBR, et al. Allogeneic natural killer cells for refractorylymphoma. Cancer Immunol Immunother 2010;59:1739–44.

Clin Cancer Res; 20(13) July 1, 2014 Clinical Cancer Research3400

Leung

on May 12, 2021. © 2014 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Page 12: Infusions of Allogeneic Natural Killer Cells as Cancer Therapy · The proposal that the resistance was related to recep-tors recognizing the absence of MHC class I arose from

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

  Updated version

  http://clincancerres.aacrjournals.org/content/20/13/3390

Access the most recent version of this article at:

   

   

  Cited articles

  http://clincancerres.aacrjournals.org/content/20/13/3390.full#ref-list-1

This article cites 96 articles, 44 of which you can access for free at:

  Citing articles

  http://clincancerres.aacrjournals.org/content/20/13/3390.full#related-urls

This article has been cited by 9 HighWire-hosted articles. Access the articles at:

   

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

  Subscriptions

Reprints and

  [email protected]

To order reprints of this article or to subscribe to the journal, contact the AACR Publications Department at

  Permissions

  Rightslink site. Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC)

.http://clincancerres.aacrjournals.org/content/20/13/3390To request permission to re-use all or part of this article, use this link

on May 12, 2021. © 2014 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from