+ All Categories
Home > Documents > Cancer Immunotherapy || Immunological Sculpting

Cancer Immunotherapy || Immunological Sculpting

Date post: 19-Dec-2016
Category:
Upload: sarah-s
View: 217 times
Download: 0 times
Share this document with a friend
13
CHAPTER 9 Immunological Sculpting: Natural Killer-Cell Receptors and Ligands Sarah S. Donatelli, Julie Y. Djeu H. Lee Moffitt Cancer Center, Tampa, FL USA I. INTRODUCTION The innate immune system is a crucial component of host defense against both foreign pathogenic invaders and aberrant self cells, such as tissues that have undergone malignant transformation. Natural Killer (NK) cells are integral members of the innate arm of immunity, and cancer patients who are deficient in NK cell number or function exhibit a weakened antitumoral response and ultimately poor prognosis. NK cells are among the first to recognize and kill virally or bacterially infected and cancerous, or “stressed,” cells. NK cells differentiate healthy from stressed cells through a vast array of germline-encoded receptors that confer either activating or inhibitory signals. NK-cell receptors differ from T-cell receptors in that they are recombinase activating gene (RAG) independent; that is, they do not somatically rearrange variable diversity or joining (VDJ) gene segments to form endless combinations of antigen-specific receptors. Rather, NK cells express genes for a finite number of receptors that recognize self antigens, and these ligands provide the basis of the NK cell’s reaction to the target cell. Distinct NK clones that exhibit varied combinations of both inhibitory and activating receptors result from the stochastic, or probabilistically determined, expression of the receptors. The expressed receptors potentiate either stimulatory “kill” or inhibitory “no kill” signals upon contact with ligands provided by the target cell. Major histocompatibility class I (MHC I) molecules constitutively expressed by healthy tissues provide ligands for receptors that transmit inhibitory “no kill” signals to the NK cell. Consequently, a lack of MHC I fails to provide the NK cell with inhibitory signals and thus indirectly potentiates an activation signal. On the other hand, stress-induced proteins normally absent on healthy cells emerge as ligands for receptors that provide the NK cell with an activating “kill” signal. During a given target interaction, the NK-cell’s receptors will associate with a combination of MHC I molecules and stress-induced ligands. The balance of signals that emanates from these interactions dictates the NK-cell’s response to the stressed cell. This chapter introduces the concepts of NK-cell receptor acquisition and signaling, and reviews current knowledge of individual receptors and their ligands. II. NK EDUCATION, LICENSING, AND PRIMING NK cells acquire the capacity to recognize and kill stressed cells through distinct receptor- ligand interactions deemed education and licensing [1]. Before NK cells develop into potent 115 Cancer Immunotherapy. http://dx.doi.org/10.1016/B978-0-12-394296-8.00009-9 Copyright Ó 2013 Elsevier Inc. All rights reserved.
Transcript
Page 1: Cancer Immunotherapy || Immunological Sculpting

CHAPTER 9

Immunological Sculpting:Natural Killer-CellReceptors and Ligands

Sarah S. Donatelli, Julie Y. DjeuH. Lee Moffitt Cancer Center, Tampa, FL USA

115

I. INTRODUCTIONThe innate immune system is a crucial component of host defense against both foreignpathogenic invaders and aberrant self cells, such as tissues that have undergone malignant

transformation. Natural Killer (NK) cells are integral members of the innate arm of immunity,

and cancer patients who are deficient in NK cell number or function exhibit a weakenedantitumoral response and ultimately poor prognosis. NK cells are among the first to recognize

and kill virally or bacterially infected and cancerous, or “stressed,” cells.

NK cells differentiate healthy from stressed cells through a vast array of germline-encoded

receptors that confer either activating or inhibitory signals. NK-cell receptors differ from T-cell

receptors in that they are recombinase activating gene (RAG) independent; that is, they do notsomatically rearrange variable diversity or joining (VDJ) gene segments to form endless

combinations of antigen-specific receptors. Rather, NK cells express genes for a finite number

of receptors that recognize self antigens, and these ligands provide the basis of the NK cell’sreaction to the target cell. Distinct NK clones that exhibit varied combinations of both

inhibitory and activating receptors result from the stochastic, or probabilistically determined,

expression of the receptors.

The expressed receptors potentiate either stimulatory “kill” or inhibitory “no kill” signals upon

contact with ligands provided by the target cell. Major histocompatibility class I (MHC I)molecules constitutively expressed by healthy tissues provide ligands for receptors that transmit

inhibitory “no kill” signals to the NK cell. Consequently, a lack of MHC I fails to provide the NK

cell with inhibitory signals and thus indirectly potentiates an activation signal. On the otherhand, stress-induced proteins normally absent on healthy cells emerge as ligands for receptors

that provide the NK cell with an activating “kill” signal. During a given target interaction, the

NK-cell’s receptors will associate with a combination of MHC I molecules and stress-inducedligands. The balance of signals that emanates from these interactions dictates the NK-cell’s

response to the stressed cell. This chapter introduces the concepts of NK-cell receptor acquisition

and signaling, and reviews current knowledge of individual receptors and their ligands.

II. NK EDUCATION, LICENSING, AND PRIMINGNK cells acquire the capacity to recognize and kill stressed cells through distinct receptor-

ligand interactions deemed education and licensing [1]. Before NK cells develop into potent

Cancer Immunotherapy. http://dx.doi.org/10.1016/B978-0-12-394296-8.00009-9

Copyright � 2013 Elsevier Inc. All rights reserved.

Page 2: Cancer Immunotherapy || Immunological Sculpting

116

SECTION 2Principles of Cancer Immunobiology

effectors that kill damaged cells, they must first become tolerant to healthy tissues through

interaction with and recognition of self MHC I molecules to prevent the occurrence ofautoimmunity [2]. In mice, this education occurs through cis interactions; that is, through the

association of an MHC I molecule with an MHC I-specific receptor expressed on the surface of

the NK cell itself [3]. This interaction requires phosphorylation of the tyrosine in theimmunotyrosine-based inhibitory motif (ITIM) located within the cytoplasmic tail of the

MHC I-binding receptor [4]. NK cells that do not express at least one MHC I-specific receptor

or NK cells that cannot engage at least one MHC I molecule in such cis interactions do notacquire the ability to kill stressed cells and thus remain “unlicensed” [3,4]. Therefore, it is only

after the NK cell is tolerant to self MHC that it can be licensed to kill.

The mechanism by which NK cells are licensed remains incompletely described; however, the

prevailing theory, or the “arming” of NK cells, has been experimentally supported. The basis

for the arming theory is that a positive signal is conferred through the inhibitory ITIM, asablation of the ITIM abrogates NK functionality [1,4]. This postulation seemed counterintu-

itive until recent experiments in mice revealed MHC I-specific receptor variants that exclusively

bind MHC I in cis (on the surface of the NK cell itself) are utilized for licensing. Association ofthese variants with cis MHC I results in sequestration of the inhibitory receptors, allowing for

a physical separation between the activating and inhibitory receptors. The activating receptors

then have the freedom to contact their ligands and transmit the resulting positive signalsrequired for the mobilization of lytic granules. Therefore, such spatial segregation positively

regulates NK-cell cytolysis, suggesting that physical distance from inhibitory molecules may be

required for effective function of activating receptors [3]. This requirement may apply to bothMHC I-specific and non-MHC I-specific activating receptors, as their signaling pathways are

shared and conserved, with the tyrosine phosphatases recruited by ITIM signaling

dephosphorylating kinases crucial early in the lytic cascade. These experiments resolved theseemingly counterintuitive fact that positive regulation can ultimately be conferred by

inhibitory ITIM signaling.

NK-cell licensing can occur in both the bone marrow during development, and in peripheral,

mature NK cells. Importantly, the capacity to kill can be revoked in the absence of MHC I,

suggesting that NK development is a constant and malleable process [5]. The licensing signalvaries with respect to the diversity of the host MHC haplotype, the number of MHC I specific

receptors per NK cell interacting with host MHC, and the affinity of said receptors for their

cognate MHC molecule [1]. Importantly, licensing is a steady-state process involved in NKcell maintenance, and under inflammatory conditions, unlicensed NK cells can kill stressed

cells, as cytokine stimulation overrides MHC I-dependent licensing in both mice [1] and

humans [6].

During steady-state maintenance as well as inflammatory siege, the killing capacity of NK

cells is enhanced by cytokines in the immune milieu. Cytokines particularly important to

NK-cell development and function are interleukin (IL) IL-2, IL-15, IL-12, IL-18, IL-21, andtype I interferons (IFN). Most of these cytokines are produced by other members of the

immune system, such as T cells, activated macrophages, and dendritic cells. IL-2 and IL-15,

produced by T cells and dendritic cells respectively, promote NK survival, proliferation,cytokine production, and cytotoxicity [7e12], likely by increase of activating surface killing

receptors [13]. Similarly, IL-12 and IL-21, sourced from activated macrophages and T cells

respectively, promote proliferation [14,15], cytokine production [16], and cytotoxicity[17,18], while macrophage and dendritic cell-derived IL-18 promotes survival [19]. IL-6

produced by T cells and macrophages increases IFNg and IL-17 production and cytotoxicity[20,21]. Virally induced type I interferons a/b also induce potent cytokine production and

cytotoxicity [22,23]. Therefore, interplay of NK cells with other members of the immune

system, as well as with their putative targets, is essential for a healthy NK-mediated hostdefense.

Page 3: Cancer Immunotherapy || Immunological Sculpting

CHAPTER 9Immunological Sculpting: Natural Killer-Cell Receptors and Ligands

117

III. HOW RECEPTOR: LIGAND INTERACTIONS TRIGGER CELL LYSISA. Cell-to-cell interactions

NK-cell receptors interact with a combination of stimulating and repressive ligands when they

investigate unfamiliar cells, and the balance of signals resulting from these interactions

determines the fate of the target. If the activation signals overcome the inhibitory signals, theNK cell will lyse the target cell. Inhibitory signals emanate primarily from MHC I ligands,

although there are also non-MHC-related inhibitory ligands [24]. All healthy cells (with the

exception of erythrocytes and platelets) express MHC I and thus, will only engage the MHCI-specific inhibitory receptor present on all functionally competent NK cells, signaling the NK

cell to spare the cell. Stressed cells, on the other hand, often downregulate MHC I, thus

preventing transduction of the inhibitory signal, allowing for triggering of NK-cell activation.The process of NK-mediated rejection of MHC I-deficient targets is known as “missing self.” In

addition to MHC I dysregulation, stressed cells can also display non-MHC I-related activatingligands, such as stress proteins induced by viral infections or malignant transformation, that

will trigger NK-cell-mediated cytolysis through their activating receptors. The specific receptors

and their ligands are discussed in detail in the following sections. There is evidence thata stronger signal (determined by either the strength or quantity of the receptor-ligand

interactions) is required for cytotoxicity of the target, whereas a weaker signal may only elicit

inflammatory cytokines such as IFNg that will recruit members of the adaptive immuneresponse for a closer investigation of the putative invader. As mentioned earlier, in mice, MHC

I-specific receptors can associate with MHC I molecules in cis, that is, MHC I molecules

expressed on the NK cell’s own plasma membrane. These cis interactions enhanceNK-mediated cytolytic activity of target cells by reducing the necessity for trans binding, or

NK-expressed MHC I-specific inhibitory receptor binding to target cell MHC I, and lowering

the threshold for activation through triggering receptors [25,26]. This caveat suggests that theNK-cell-intrinsic MHC repertoire can contribute to determination of the signal strength

required to kill the target cell, and NK cells diverse in MHC alleles may be more innately

“active” than NK cells that do not exhibit MHC diversity.

B. Signaling

When an NK cell encounters a potential target, intracellular signals reverberate via the surface

receptors. The major stimulatory or inhibitory signaling pathways are conserved among

respective receptors (Figure 9.1). Activating receptors have short cytoplasmic tails that lack anycapacity for signaling. Therefore, activating receptors generally associate with

immunotyrosine-based activation motif (ITAM)-containing adaptor molecules that can be

phosphorylated to activate downstream signals that end in lytic granule and/or cytokinesecretion. ITAM-containing adaptor proteins associate with the short cytoplasmic tails by

binding to charged transmembrane amino acids.

The most frequently used adaptor molecules are dynax activating protein 12kD (DAP12),

CD3z, FcRg, and DAP10. DAP12 is associated with NK activating receptors through charged

residues in the transmembrane region of the receptor’s cytoplasmic tail, and upon receptor-ligand interaction, Src family kinases such as lymphocyte-specific protein tyrosine kinase (Lck)

and Fyn phosphorylate the tyrosine in the adaptor’s ITAM. Spleen tyrosine kinase (Syk) [27] or

zeta-chain-associated protein kinase 70 (Zap70)[28] is recruited by binding to thephosphotyrosine in the ITAM and activating phosphoinositide-3 kinase (PI3K) that leads to

sequential Rac1, PAK1, and MAPK/ERK activation to mobilize lytic granules that kill the target

cell [29,30]. Recently, protein kinase C-Ө (PKC-Ө) has emerged as a requirement for IFNg

release, but not cytolysis, during sustained ITAM signaling [31]. Other adaptors, such as CD3z

and FcRg, function in the same manner as DAP12, as they contain the same ITAM (Figure 9.1).

DAP10, on the other hand, does not express an ITAM but contains a PI3K-binding domain.The Fyn/Lck phosphorylation of this tyrosine-containing domain recruits PI3K which results

Page 4: Cancer Immunotherapy || Immunological Sculpting

FIGURE 9.1Conserved signaling pathways of NK receptors.Activating receptor ligation leads to association of

ITAM-containing adaptor proteins and subsequent entry

into the PI3K signaling pathway, ultimately leading to IFNgand lytic granule secretion. Inhibitory receptor ligation

leads to activating/inhibitory receptor colocalization, ITIM

phosphorylation and subsequent recruitment of SHP-1 and

SHP-2 to the SH2 domain, which dephosphorylates critical

signaling proteins at the activating receptor/adaptor

protein complex. Activating receptors are depicted in

green, while inhibitory receptors are depicted as red.

Signaling proteins are blue. Green arrows depict

sequential activation by phosphorylation of the previous

signaling protein.

SECTION 2Principles of Cancer Immunobiology

118

in mobilization of granules via the same pathway as DAP12 (Figure 9.1)[32]. In addition,

antibody-dependent cell cytotoxicity by NK cells can occur via this pathway through FcgRIII(CD16), which couples to CD3z, or FcεRg. Overall, a multitude of ligands can trigger NK cells

and the initial signaling molecules activated may vary depending on the receptor, but they

merge downstream into a conserved common cascade ending in ERK activation andsubsequent lytic granule mobilization.

In contrast, inhibitory receptors have long cytoplasmic tails that contain one or moreimmunotyrosine inhibitory motifs (ITIM). Upon receptor-ligand interaction, these ITIMs are

phosphorylated by Lck and Fyn (Figure 9.1). Src homology-2 domain containing phosphatase

1 and 2 (SHP-1 and SHP-2) then bind to the phosphotyrosine in the ITIM through their SH2domains [33,34]. SHP-1 and SHP-2 then function to disrupt the lytic cascade by specific

dephosphorylation of Syk and Zap70, crucial early signaling proteins necessary for NK

activation tyrosines [35].

IV. RECEPTORS FOR MHC I AND MHC I-RELATED MOLECULESA. Killer immunoglobulin-like receptors (KIR)

MHC Imolecules provide the most robust and abundant signals to NK cells. It is throughMHC

molecules that NK cells can sample the health of the environment. These interactions are

crucial to NK function, providing both inhibitory and activating signals. The largest family ofmolecules expressed in human NK cells that recognize MHC I on target cells are the killer

immunoglobulin-like receptors (KIR) and can be inherited as two haplotypes, A and B

(Figure 9.2). All KIR contain either two (KIR2D) or three (KIR3D) extracellular immuno-globulin (Ig)-like domains and may exhibit either short (KIR2/3DS) or ITIM-containing long

(KIR2/3DL) cytoplasmic tails that play activating or inhibitory roles in NK cell target signaling,

respectively (Figure 9.3). For simplicity, in this review, all stimulatory KIR gene products arereferred to as “aKIR,” and all inhibitory KIR gene products are referred to as “iKIR.”

As stated earlier, aKIR provide stimulatory signals to NK cells when they contact their targetligands, causing lytic granule mobilization that induces target cell lysis or cytokine secretion to

Page 5: Cancer Immunotherapy || Immunological Sculpting

FIGURE 9.2KIR haplotypes. Schematic depiction of the KIR genotypesof haplotypes A and B. The A haplotype is fixed in gene

content, while the B haplotype varies extensively in gene

content. The boxes on and below the line are KIRs all

variably expressed in a given B haplotype. Framework

genes present on all haplotypes are outlined in bold black

line, activating KIR are depicted in green and inhibitory KIR

in red. The numbers below the series of boxes in haploype

B represent the various KIR combinations that can be

expressed in a given B haplotype.

FIGURE 9.3NK-cell surface receptors. Receptors contributingto NK activation are green and grouped with a solid

green line, while receptors contributing to NK

inhibition are red and grouped with a solid red line.

Activating co-receptors are grouped with a dashed,

color-coded line. ITIMs are depicted as yellow

slashes in the red cytoplasmic tails. Abbreviated

receptor names are displayed proximal to the

receptor depiction.

CHAPTER 9Immunological Sculpting: Natural Killer-Cell Receptors and Ligands

119

amplify the immune response. They contain no intrinsic signaling capacity in their shortcytoplasmic tails, but they do contain a charged lysine or arginine that facilitates association

with DAP12. Six aKIR that contain two extracellular domains exhibiting different MHC I allelic

preferences have been identified. Like their homologous inhibitory relatives, KIR2DS1,KIR2DS2, and KIR2DS3 bind human leukocyte antigen (HLA)-C, MHC I, alleles. This caveat

implies that when an NK cell sees a target expressing HLA-C, it can receive both inhibitory and

activating signals from the same ligand and adds to the complexity of the ensuing reaction.Furthermore, it suggests that the surface ratio of activating-to-inhibitory KIR may be a deter-

mining factor in the fate of the target cell. KIR2DS4 is exceptional in that it is the only KIR

expressed in Haplotype A individuals. Furthermore, proteins other than MHC molecules havebeen identified as ligands. In addition to HLA-C1, -C2, and -A11 [36], KIR2DS4 is triggered

by unidentified melanoma proteins [37], and unknown proteins expressed by chronic

lymphocytic leukemia (CLL) cells [38], suggesting that this aKIR may play a distinct role inantitumoral defense. Ligands for KIR2DS5 and KIR2DS3 remain unidentified.

Page 6: Cancer Immunotherapy || Immunological Sculpting

SECTION 2Principles of Cancer Immunobiology

120

In contrast to aKIR, iKIR provide negative intracellular signals by recruiting SHP-1 and SHP-2to the ITIMs in their long cytoplasmic tails upon ligand binding. Eight iKIR that exhibit

different MHC allelic preferences have been identified. KIR2DL1, KIR2DL2, and KIR2DL3

bind HLA-C alleles [39], KIR3DL1 binds HLA-B alleles [40,41], and KIR3DL2 binds HLA-A3and A11 [42]. The ligand for KIR2DL5 remains unidentified. KIR2DL4 is distinct in that it is

expressed in all humans, and it has been experimentally shown to exhibit both activating andinhibitory signals. Furthermore, KIR2DL4 recognizes soluble HLA-G, which is expressed in the

placenta, where classical HLAmolecules are absent, and functions during pregnancy to prevent

spontaneous abortion of the fetus.

It has been documented that KIR expression is controlled epigenetically by DNA methylation

of cytosines in CpG dinucleotides rather than by genetic sequence differences in promoters

[43]. These DNA methylation patterns are maintained over numerous cell divisions, andcontribute to the diversity of gene expression in individual NK clones and their progeny [44].

KIRs are expressed stochastically leading to a repertoire of NK clones that express different

combinations of activating and inhibitory KIRs. Genotyping and epidemiological studies haveyielded two predominant patterns of KIR expression (Figure 9.2). Haplotype A individuals

exhibit a relatively fixed gene content consisting of five iKIRs along with one aKIR, KIR2DS4,

and KIR2DL4, which can exhibit both inhibitory and activating functions. Conversely,Haplotype B individuals express variable gene content consisting of multiple aKIR and iKIR

combinations [45]. The frequencies of these haplotypes vary significantly with ethnicity;

furthermore, correlations between KIR haplotypes and susceptibility to certain cancers andinfections are beginning to emerge.

B. Natural cytotoxicity receptors

Apart from MHC-dependent interactions, NK cells rely on interactions with non-MHC-related

proteins to distinguish stressed from healthy cells. The presence of non-MHC-related ligandswas first noted when NK cells readily lysedMHC I-deficient tumor cells. We now know that this

feature is a function due both to a lack of MHC I-mediated inhibition (“missing self”) as well

as engagement of activating receptors that recognize non-MHC I molecules. The receptorsdiscovered to play a large role in this MHC-independent target lysis were termed the natural

cytotoxicity receptors (NCR) (Figure 9.3). Deletion of even one NCR reduces the ability of NK

cells to kill tumor targets in vivo [46]; this weakness is important because tumor-producedtransforming growth factor b-1 (TGFb) downregulates NCR family members to evade NK

cytolysis [47]. Additionally, NCRs can potentiate interactions with antigen-presenting cells,

such as dendritic cells, that can aid in cellular maturation for recruitment of the adaptive armof immunity [48].

The NCR consist of NKp30, NKp44, NKp46, and NKp80 [49e51] and are named for their

respective molecular weights. NKp30, NKp46, and Nkp80 are expressed on activated andresting NK cells while NKp44 is upregulated by IL-2. NKp46 ligands include hemagglutinin

and hemagglutinin-neuraminidase of influenza and parainfluenza [52], as well as heparin

sulfate proteoglycans, which also bind NKp30 [53]. Other ligands for NKp30 are nuclear factorHLA-B-associated transcript 3, a factor released from tumor cells [54], B7-H6 [55], CMV pp65,

and BAT3 [56].

Of the NCRs, only NKp44 associates with and signals through DAP12 (Figure 9.1)[49]. NKp30

and NK46 couple to CD3z and FcεRg adaptor molecules, which form heterotrimeric adaptor

complexes [50,51]. NKp80 is an activating receptor that binds to the genetically linkedactivation-induced C-type lectin (AICL) and stimulates NK cytotoxicity against malignant

myeloid cells [57]. NKp80 does not require an adaptor protein; rather, it signals through

a hemi-ITAM in its cytoplasmic tail through recruitment of Syk [58] and likely progressionthrough the PI3K activation pathway.

Page 7: Cancer Immunotherapy || Immunological Sculpting

CHAPTER 9Immunological Sculpting: Natural Killer-Cell Receptors and Ligands

121

C. CD94/NKG2 and NKG2D

The NKG2 family is similar to KIR in that it contains both inhibitory and activating receptors;

however, either MHC I or non-MHC I molecules can function as ligands. Furthermore, somefamily members, NKG2A, NKG2C, and NKG2E, heterodimerize with CD94, while NKG2D

associates with itself to form a homodimer (Figure 9.3).

NKG2D is distantly related to the other NKG2 family members and does not dimerize withCD94, but rather forms homodimers that associate with DAP10 to yield an activation signal. In

humans, NKG2D complexes bind stress-inducible MHC class I chain-related gene A and B

(MICA/ MICB), whose steady-state distribution is restricted to intestinal epithelia, but isupregulated in many epithelial tumors [59,60]. Additional NKG2D ligands are UL16-binding

proteins (ULBP) 1/2/3/4 which are induced by cytomegalovirus infection [61e63] and in

mice, NKG2D recognizes the retinoic acid-inducible early gene-1 (Rae-1)-like proteins [62]and H-60, a minor MHC derived antigen. NKG2D mediates direct tumor target killing as well

as induction of antitumoral T-cell responses [64], and is therefore a crucial component of the

NK defense. However, tumors have demonstrated evasion of NKG2D-mediated killing bysecretion of TGFb, which downregulates NKG2D on NK cells [65], and NKG2D ligands on

tumor cells [66]. Additionally, tumors shed soluble MICA/B, which functions as a decoy

receptor [67].

The other three NKG2 family members, NKG2A, NKG2C, and NKG2E, form heterodimeric

complexes with CD94, another lectin-like protein. These complexes recognize the nonclassicalMHCmolecule, HLA-E. HLA-E is deemed nonclassical in part because it presents only peptides

derived from classical MHC I molecules. This feature allows NK cells to indirectly explore the

MHC I environment of the putative target [68]. NKG2A contains an ITIM; thus upon bindingto HLA-E it elicits an inhibitory signal. Interestingly, NKG2C/E-CD94 also binds HLA-E, yet it

does not contain an ITIM. Rather, it associates with DAP12, transmitting an activation signal.

D. CD160 (BY55)

CD160 is an activating receptor expressed in the CD56dimCD16þ cytotoxic NK subset that

constitutes the major NK fraction in circulating human NK cells (Figure 9.3)[69,70].Engagement of CD160 recruits PI3K to activate the lytic cascade [71]. Both

glycosylphosphatidylinositol (GPI)-anchored and transmembrane isoforms are found on NK

cells. The major GPI-anchored form exhibits homology to KIR2DL4; however, its signalingconstituents have not yet been elucidated. On the other hand, the transmembrane isoform,

which is transiently expressed on cytokine-activated NK cells, contains a transmembrane

region that activates Erk1/2 [72]. CD160 is a promiscuous receptor that binds a variety ofligands including MHC class 1a and 1b molecules HLA-C [69], soluble HLA-G [73], HLA-A2,

HLA-B7, and HLA-E [69], as well as the herpesvirus entry mediator protein [74].

E. SLAM family receptors

In vivo, NK cells not only contact tissue cells, they also contact other NK cells. To prevent

unmitigated self killing, NK express signaling lymphocyte activation molecule (SLAM)-relatedreceptors. SLAM-family receptors expressed on human NK cells are NTB-A and CRACC, as well

as 2B4 (CD244) (Figure 9.3). NTB-A and CRACC serve as self-ligands during homotypic

interactions between NK cells, while the 2B4 ligand is CD48, a glycophosphatidylinositol(GPI)-linked member of the CD2 family of receptors expressed on hematopoietic cells [75].

Engagement of 2B4, NTB-A and CRACC onMHC I-negative targets (i.e., stressed cells) activates

NK-cell cytotoxicity, while 2B4, NTB-A, or CRACC-expressing, MHC I-positive cells(i.e., neighboring NK cells) are spared [76].

Signaling of SLAM-family members is distinct from that of other NK-cell receptors as they donot recruit traditional ITAM-containing adaptor proteins such as DAP10 or DAP12. Rather, the

Page 8: Cancer Immunotherapy || Immunological Sculpting

SECTION 2Principles of Cancer Immunobiology

122

cytoplasmic domains contain tyrosine-based motifs that undergo phosphorylation to recruitSH2-domain containing SLAM-associated protein (SAP) or Ewing’s sarcoma-activated

transcripts-2 (EAT-2). In mice, the SLAM-family receptors can exhibit either activating or

inhibitory functions, depending on the adaptor that they recruit; however, in humans,SLAM-family receptors have only been shown to be stimulatory [77].

F. Additional NK receptors

In addition to directly lysing target cells, NK also work in concert with B lymphocytes to

mediate antibody-dependent cellular cytotoxicity (ADCC) [78]. NK express CD16 which binds

IgG-opsonized cells (Figure 9.3). The activating CD16 then associates with ITAM-containingCD3z and FCRg to bind Syk and proceed through the NK activation pathway to ultimately lyse

the target cell [79].

NK cells also express an array of receptors that promote the formation of NK-celletarget cellconjugates and therefore act as coreceptors for the lytic granule inducing receptors. Some of

these receptors have signaling capacity that promotes activation and cytotoxicity in addition to

enhancing adhesion. Others primarily function to form andmaintain the contacts between theNK cell and the target cell. Typically, these receptors recognize adhesion molecules present on

the target cells. The primary adhesion receptor that is required for stable conjugate formation is

leukocyte function associated antigen 1 (LFA-1) (Figure 9.3)[80], an integrin that binds tointercellular adhesion molecule 1 (ICAM-1). The binding of LFA-1 to ICAM-1 results in actin

polymerization for enhanced adhesion to the target cell [81]. LFA-1 has been demonstrated as

a co-activator during NCR-mediated stromal cell killing and NKG2D-mediated killing ofdendritic cells [82]. Another important activating receptor is DNAX accessory molecule-1

(DNAM-1), which also potentiates adhesion and cytotoxicity (Figure 9.3) [31,83] and mayhave a role in NK-cell migration [84]. The ligands for DNAM-1 in humans are CD155 (polio

virus receptor (PVR) or Nectin-5), and CD112 (Nectin-2), both of which are upregulated on

tumor cells [85], suggesting a role for the receptor in tumor cell lysis [86]. DNAM-1 signaling isdependent upon Fyn [83], and downstream signaling is dependent upon association with

LFA-1 [87]. Two additional receptors that recognize Nectins are class I-restricted T-cell-

associated molecule (CRTAM) which binds Necl-2 and enhances NK activation, and CD96(Tactile), which recognizes Necl-5 and primarily promotes adhesion, but not activation

(Figure 9.3). Another molecule that promotes NK-celletarget cell interactions is CD100 (or

Semaphorin 4D (SEMA4D)) (Figure 9.3), which potentiates enhanced cytotoxicity and cyto-kine production upon CD72 ligation by enhancing the adhesion between NK cells and their

targets [88]. CD72 is primarily expressed on B cells [89], implicating this interaction in

NK-mediated killing of stressed B cells.

In addition to receptors that bind adhesion molecules on target cells and thus promote

NK-target cell cytotoxicity, NK cells also exhibit receptors that recognize adhesion molecules,

yet are inhibitory. KLRG1 is an ITIM-containing inhibitory receptor that recognizes the clas-sical cadherins, E-, N-, and R-cadherin (Figure 9.3)[90]. Reduced levels of E-cadherin in

malignant epithelial tumors correlate with tumor metastasis, suggesting a potential role for NK

detection of malignant epithelia [91]. LAIR-1 recognizes a common collagen motif andcontains a single extracellular Ig-like domain and two cytoplasmic tyrosine-based inhibitory

motifs (ITIMs) (Figure 9.3)[92]. Cross-linking of LAIR-1 on NK cells delivers a potent inhib-

itory signal that is capable of inhibiting target cell lysis mediated by resting and activated NKcells [92,93] and B cells [94].

Other receptors can help to differentiate subsets of NK cells. While some have knownsignaling functions, others have only been identified as developmental markers. A major

marker for NK subsets is CD56 (Figure 9.3); CD56lo cells constitute the majority of circu-

lating NK cells and exhibit high cytotoxicity capacity to target cells. Conversely, CD56hi cellsconstitute approximately 10% of circulating NK cells and produce IFNg but have low

Page 9: Cancer Immunotherapy || Immunological Sculpting

CHAPTER 9Immunological Sculpting: Natural Killer-Cell Receptors and Ligands

123

cytolytic capacity. In addition, CD27 is expressed on the CD56hi subset of NK and bindsCD70 (Figure 9.3)[95], and this interaction promotes NK-mediated cytotoxicity and primes

CD8þ T-cell responses [96]. Moreover, NK-cell differentiation is accompanied by expression

of a post-translation modification of P-selectin glycoprotein ligand-1 (PSGL-1), deemed thePEN5 epitope, that allows for binding to the integrin L-selectin, suggesting that PEN5/PSGL-1

potentially serves as a NK-homing/trafficking receptor [97]. Of note, NK cells also expressnon-KIR MHC I-specific inhibitory receptors. For example, Lilr1b (ILT2/CD85j /LIR-1),

which is a potent MHC I-binding inhibitory receptor, possesses four ITIMs in its cytoplasmic

tail (Figure 9.3)[98].

Aberrantly expressed glycoproteins and glycolipids have recently been characterized as tumor

markers and can be engaged by sialic acid-binding immunoglobulin (Ig)-like lectins (Siglecs)

which are broadly expressed on hematopoietic cells. Of the receptors, Siglec 3 (CD33) isexpressed on activated NK cells and contains two ITIMs that negatively regulate NK-cell

activation (Figure 9.3)[99,100]. Siglec-7 (p70/AIRM) and Siglec-9 are CD33-related Siglecs

that are also expressed on NK cells (Figure 9.3) [101] and have been implicated in clinicalpathogenesis. Decreased Siglec-7 expression is an early marker of NK dysfunction and high

viral load in HIV infections [102]. Furthermore, Siglec-7 expressed on NK preferentially binds

internally branched a2,6-linked disialic gangliosides such as disialosyl globopentaosylcer-amide (DSGb5), a ganglioside upregulated on renal cell carcinoma that contributes to

metastases [103]. Siglec-9’s ligand is MUC-16 [104], which is overexpressed in cancer [105] and

functions to inhibit NK-cell-mediated antitumor responses [106] by acting as an anti-adhesivemolecule and preventing the formation of the immunological synapse between target cells

and NK cells [107].

V. CONCLUSIONNK cells are a crucial part of the first line immune response, particularly in the context of

cancer. In contrast to T cells, which are negatively selected and clonally deleted in the thymusif they exhibit self-reactivity, NK cells are genetically primed to tolerate self MHC and yet

recognize and kill malignant tissue through detection of aberrantly expressed self-proteins.Through an array of receptors, NK cells can differentiate stressed cancerous cells from healthy

tissue, and the balance of inhibitory and activating signals that result from the interactions

determines the fate of the putative malignancy (Figure 9.3). Unfortunately, tumors can evadeNK-cell-mediated destruction by secretion of immunosuppressive molecules and/or shedding

soluble variants of activating receptor ligands. Therefore, in the tumor microenvironment,

NK-cell function is compromised. Identifying mechanisms that maintain the expression ofstimulatory receptors may provide therapeutic benefit by shifting the intratumoral NK cell

towards an aggressive and active phenotype that is ready to detect and destroy invading

malignancies.

References[1] Elliott JM, Yokoyama WM. Unifying concepts of MHC-dependent natural killer cell education. Trends

Immunol 2011 Aug;32(8):364e72.

[2] Anfossi N, Andre P, Guia S, Falk CS, Roetynck S, Stewart CA, et al. Human NK cell education by inhibitory

receptors for MHC class I. Immunity 2006 Aug;25(2):331e42.

[3] ChalifourA,ScarpellinoL,Back J,BrodinP,DevevreE,GrosF, et al. ARole for cis Interactionbetween the Inhibitory

Ly49A receptor and MHC class I for natural killer cell education. Immunity 2009 Mar 20;30(3):337e47.

[4] Kim S, Poursine-Laurent J, Truscott SM, Lybarger L, Song YJ, Yang L, et al. Licensing of natural killer cells by

host major histocompatibility complex class I molecules. Nature 2005 Aug 4;436(7051):709e13.

[5] Joncker NT, Shifrin N, Delebecque F, Raulet DH. Mature natural killer cells reset their responsiveness when

exposed to an altered MHC environment. J Exp Med 2010 Sep 27;207(10):2065e72.

[6] Juelke K, Killig M, Thiel A, Dong J, Romagnani C. Education of hyporesponsive NK cells by cytokines. Eur JImmunol 2009 Sep;39(9):2548e55.

Page 10: Cancer Immunotherapy || Immunological Sculpting

SECTION 2Principles of Cancer Immunobiology

124

[7] Fehniger TA, Bluman EM, Porter MM, Mrozek E, Cooper MA, VanDeusen JB, et al. Potential mechanisms ofhuman natural killer cell expansion in vivo during low-dose IL-2 therapy. J Clin Invest 2000 Jul;106(1):117e24.

[8] Warren HS, Kinnear BF, Kastelein RL, Lanier LL. Analysis of the costimulatory role of IL-2 and IL-15 in

initiating proliferation of resting (CD56dim) human NK cells. J Immunol 1996 May 1;156(9):3254e9.

[9] Lotzova E, Savary CA, Herberman RB. Induction of NK cell activity against fresh human leukemia in culture

with interleukin 2. J Immunol 1987 Apr 15;138(8):2718e27.

[10] Brilot F, Strowig T, Roberts SM, Arrey F, Munz C. NK cell survival mediated through the regulatory synapse

with human DCs requires IL-15Ralpha. J Clin Invest 2007 Nov;117(11):3316e29.

[11] Carson WE, Giri JG, Lindemann MJ, Linett ML, Ahdieh M, Paxton R, et al. Interleukin (IL) 15 is a novel

cytokine that activates human natural killer cells via components of the IL-2 receptor. J Exp Med 1994 Oct

1;180(4):1395e403.

[12] Ferlazzo G, Pack M, Thomas D, Paludan C, Schmid D, Strowig T, et al. Distinct roles of IL-12 and IL-15 in

human natural killer cell activation by dendritic cells from secondary lymphoid organs. Proc Natl Acad Sci U

S A 2004 Nov 23;101(47):16606e11.

[13] Ferlazzo G, Thomas D, Lin SL, Goodman K, Morandi B, Muller WA, et al. The abundant NK cells in human

secondary lymphoid tissues require activation to express killer cell Ig-like receptors and become cytolytic.

J Immunol 2004 Feb 1;172(3):1455e62.

[14] Loza MJ, Perussia B. The IL-12 signature: NK cell terminal CD56þhigh stage and effector functions.

J Immunol 2004 Jan 1;172(1):88e96.

[15] Wendt K, Wilk E, Buyny S, Schmidt RE, Jacobs R. Interleukin-21 differentially affects human natural killer cell

subsets. Immunology 2007 Dec;122(4):486e95.

[16] Girart MV, Fuertes MB, Domaica CI, Rossi LE, Zwirner NW. Engagement of TLR3, TLR7, and NKG2D regulateIFN-gamma secretion but not NKG2D-mediated cytotoxicity by human NK cells stimulated with suboptimal

doses of IL-12. J Immunol 2007 Sep 15;179(6):3472e9.

[17] Wu CY, Gadina M, Wang K, O’Shea J, Seder RA. Cytokine regulation of IL-12 receptor beta2 expression:

differential effects on human T and NK cells. Eur J Immunol 2000 May;30(5):1364e74.

[18] Skak K, Frederiksen KS, Lundsgaard D. Interleukin-21 activates human natural killer cells and modulates theirsurface receptor expression. Immunology 2008 Apr;123(4):575e83.

[19] Hodge DL, Subleski JJ, Reynolds DA, Buschman MD, Schill WB, Burkett MW, et al. The proinflammatory

cytokine interleukin-18 alters multiple signaling pathways to inhibit natural killer cell death. J InterferonCytokine Res 2006 Oct;26(10):706e18.

[20] Malejczyk J, Malejczyk M, Urbanski A, Kock A, Jablonska S, Orth G, et al. Constitutive release of IL6 byhuman papillomavirus type 16 (HPV16)-harboring keratinocytes: a mechanism augmenting the NK-cell-

mediated lysis of HPV-bearing neoplastic cells. Cell Immunol 1991 Aug;136(1):155e64.

[21] Passos ST, Silver JS, O’Hara AC, Sehy D, Stumhofer JS, Hunter CA. IL-6 promotes NK cell production of IL-17during toxoplasmosis. J Immunol 2010 Feb 15;184(4):1776e83.

[22] Nguyen KB, Salazar-Mather TP, Dalod MY, Van Deusen JB, Wei XQ, Liew FY, et al. Coordinated and distinct

roles for IFN-alpha beta, IL-12, and IL-15 regulation of NK cell responses to viral infection. J Immunol 2002Oct 15;169(8):4279e87.

[23] Sato K, Hida S, Takayanagi H, Yokochi T, Kayagaki N, Takeda K, et al. Antiviral response by natural killer cellsthrough TRAIL gene induction by IFN-alpha/beta. Eur J Immunol 2001 Nov;31(11):3138e46.

[24] Lanier LL. DAP10- and DAP12-associated receptors in innate immunity. Immunol Rev 2009 Jan;227(1):150e60.

[25] Doucey MA, Scarpellino L, Zimmer J, Guillaume P, Luescher IF, Bron C, et al. Cis association of Ly49A with

MHC class I restricts natural killer cell inhibition. Nat Immunol 2004 Mar;5(3):328e36.

[26] Scarpellino L, Oeschger F, Guillaume P, Coudert JD, Levy F, Leclercq G, et al. Interactions of Ly49 familyreceptors with MHC class I ligands in trans and cis. J Immunol 2007 Feb 1;178(3):1277e84.

[27] Jiang K, Zhong B, Gilvary DL, Corliss BC, Vivier E, Hong-Geller E, et al. Syk regulation of phosphoinositide3-kinase-dependent NK cell function. J Immunol 2002 Apr 1;168(7):3155e64.

[28] Lanier LL, Corliss BC, Wu J, Leong C, Phillips JH. Immunoreceptor DAP12 bearing a tyrosine-based acti-

vation motif is involved in activating NK cells. Nature 1998 Feb 12;391(6668):703e7.

[29] Jiang K, Zhong B, Gilvary DL, Corliss BC, Hong-Geller E, Wei S, et al. Pivotal role of phosphoinositide-3

kinase in regulation of cytotoxicity in natural killer cells. Nat Immunol 2000 Nov;1(5):419e25.

[30] Djeu JY, Jiang K, Wei S. A view to a kill: signals triggering cytotoxicity. Clin Cancer Res 2002 Mar;8(3):636e40.

[31] Tassi I, CellaM, Presti R, Colucci A, Gilfillan S, LittmanDR, et al. NK cell-activating receptors require PKC-theta forsustained signaling, transcriptional activation, and IFN-gamma secretion. Blood 2008 Nov 15;112(10):4109e16.

[32] Sutherland CL, Chalupny NJ, Schooley K, VandenBos T, Kubin M, Cosman D. UL16-binding proteins, novel

MHC class I-related proteins, bind to NKG2D and activate multiple signaling pathways in primary NK cells.J Immunol 2002 Jan 15;168(2):671e9.

Page 11: Cancer Immunotherapy || Immunological Sculpting

CHAPTER 9Immunological Sculpting: Natural Killer-Cell Receptors and Ligands

125

[33] Binstadt BA, Brumbaugh KM, Dick CJ, Scharenberg AM, Williams BL, Colonna M, et al. Sequentialinvolvement of Lck and SHP-1 with MHC-recognizing receptors on NK cells inhibits FcR-initiated tyrosine

kinase activation. Immunity 1996 Dec;5(6):629e38.

[34] Burshtyn DN, Scharenberg AM, Wagtmann N, Rajagopalan S, Berrada K, Yi T, et al. Recruitment of tyrosinephosphatase HCP by the killer cell inhibitor receptor. Immunity 1996 Jan;4(1):77e85.

[35] McVicar DW, Burshtyn DN. Intracellular signaling by the killer immunoglobulin-like receptors and Ly49. Sci

STKE 2001 Mar 27;2001(75): re1.

[36] Graef T, Moesta AK, Norman PJ, Abi-Rached L, Vago L, Older Aguilar AM, et al. KIR2DS4 is a product of gene

conversion with KIR3DL2 that introduced specificity for HLA-A*11 while diminishing avidity for HLA-C.J Exp Med 2009 Oct 26;206(11):2557e72.

[37] Katz G, Gazit R, Arnon TI, Gonen-Gross T, Tarcic G, Markel G, et al. MHC class I-independent recognition of

NK-activating receptor KIR2DS4. J Immunol 2004 Aug 1;173(3):1819e25.

[38] Giebel S, Nowak I, Wojnar J, Krawczyk-Kulis M, Holowiecki J, Kyrcz-Krzemien S, et al. Association of

KIR2DS4 and its variant KIR1D with leukemia. Leukemia 2008 Nov;22(11):2129e30, discussion 30e1.

[39] Winter CC, Long EO. A single amino acid in the p58 killer cell inhibitory receptor controls the ability of natural

killer cells to discriminate between the two groups of HLA-C allotypes. J Immunol 1997 May 1;158(9):4026e8.

[40] Cella M, Longo A, Ferrara GB, Strominger JL, Colonna M. NK3-specific natural killer cells are selectively

inhibited by Bw4-positive HLA alleles with isoleucine 80. J Exp Med 1994 Oct 1;180(4):1235e42.

[41] Carr WH, Pando MJ, Parham P. KIR3DL1 polymorphisms that affect NK cell inhibition by HLA-Bw4 ligand.J Immunol 2005 Oct 15;175(8):5222e9.

[42] Dohring C, Scheidegger D, Samaridis J, Cella M, Colonna M. A human killer inhibitory receptor specific for

HLA-A1,2. J Immunol 1996 May 1;156(9):3098e101.

[43] Santourlidis S, Trompeter HI, Weinhold S, Eisermann B, Meyer KL, Wernet P, et al. Crucial role of DNA

methylation in determination of clonally distributed killer cell Ig-like receptor expression patterns in NK cells.J Immunol 2002 Oct 15;169(8):4253e61.

[44] Moretta L, Moretta A. Killer immunoglobulin-like receptors. Curr Opin Immunol 2004 Oct;16(5):626e33.

[45] Kulkarni S, Martin MP, Carrington M. The Yin and Yang of HLA and KIR in human disease. Semin Immunol

2008 Dec;20(6):343e52.

[46] Sivori S, Pende D, Bottino C, Marcenaro E, Pessino A, Biassoni R, et al. NKp46 is the major triggering receptorinvolved in the natural cytotoxicity of fresh or cultured human NK cells. Correlation between surface density

of NKp46 and natural cytotoxicity against autologous, allogeneic or xenogeneic target cells. Eur J Immunol

1999 May;29(5):1656e66.

[47] Ghio M, Contini P, Negrini S, Boero S, Musso A, Poggi A. Soluble HLA-I-mediated secretion of TGF-beta1 by

human NK cells and consequent down-regulation of anti-tumor cytolytic activity. Eur J Immunol 2009

Dec;39(12):3459e68.

[48] Moretta A. Natural killer cells and dendritic cells: rendezvous in abused tissues. Nat Rev Immunol 2002

Dec;2(12):957e64.

[49] Cantoni C, Bottino C, Vitale M, Pessino A, Augugliaro R, Malaspina A, et al. NKp44, a triggering receptor

involved in tumor cell lysis by activated human natural killer cells, is a novel member of the immunoglobulin

superfamily. J Exp Med 1999 Mar 1;189(5):787e96.

[50] Pessino A, Sivori S, Bottino C, Malaspina A, Morelli L, Moretta L, et al. Molecular cloning of NKp46: a novel

member of the immunoglobulin superfamily involved in triggering of natural cytotoxicity. J Exp Med 1998

Sep 7;188(5):953e60.

[51] Pende D, Parolini S, Pessino A, Sivori S, Augugliaro R, Morelli L, et al. Identification and molecular

characterization of NKp30, a novel triggering receptor involved in natural cytotoxicity mediated by human

natural killer cells. J Exp Med 1999 Nov 15;190(10):1505e16.

[52] Mandelboim O, Lieberman N, Lev M, Paul L, Arnon TI, Bushkin Y, et al. Recognition of haemagglutinins on

virus-infected cells by NKp46 activates lysis by human NK cells. Nature 2001 Feb 22;409(6823):1055e60.

[53] Bloushtain N, Qimron U, Bar-Ilan A, Hershkovitz O, Gazit R, Fima E, et al. Membrane-associated heparan

sulfate proteoglycans are involved in the recognition of cellular targets by NKp30 and NKp46. J Immunol

2004 Aug 15;173(4):2392e401.

[54] Pogge von Strandmann E, Simhadri VR, von Tresckow B, Sasse S, Reiners KS, Hansen HP, et al. Human

leukocyte antigen-B-associated transcript 3 is released from tumor cells and engages the NKp30 receptor on

natural killer cells. Immunity 2007 Dec;27(6):965e74.

[55] Brandt CS, Baratin M, Yi EC, Kennedy J, Gao Z, Fox B, et al. The B7 family member B7-H6 is a tumor cell ligand

for the activating natural killer cell receptor NKp30 in humans. J Exp Med 2009 Jul 6;206(7):1495e503.

[56] Arnon TI, Achdout H, Levi O, Markel G, Saleh N, Katz G, et al. Inhibition of the NKp30 activating receptor by

pp65 of human cytomegalovirus. Nat Immunol 2005 May;6(5):515e23.

Page 12: Cancer Immunotherapy || Immunological Sculpting

SECTION 2Principles of Cancer Immunobiology

126

[57] Tomlinson MG, Lin J, Weiss A. Lymphocytes with a complex: adapter proteins in antigen receptor signaling.Immunol Today 2000 Nov;21(11):584e91.

[58] Dennehy KM, Klimosch SN, Steinle A. Cutting edge: NKp80 uses an atypical hemi-ITAM to trigger NK

cytotoxicity. J Immunol 2011 Jan 15;186(2):657e61.

[59] Groh V, Bahram S, Bauer S, Herman A, Beauchamp M, Spies T. Cell stress-regulated human major histo-

compatibility complex class I gene expressed in gastrointestinal epithelium. Proc Natl Acad Sci U S A 1996

Oct 29;93(22):12445e50.

[60] Groh V, Rhinehart R, Secrist H, Bauer S, Grabstein KH, Spies T. Broad tumor-associated expression and

recognition by tumor-derived gamma delta T cells of MICA and MICB. Proc Natl Acad Sci U S A 1999 Jun8;96(12):6879e84.

[61] Bauer S, Groh V, Wu J, Steinle A, Phillips JH, Lanier LL, et al. Activation of NK cells and T cells by NKG2D,

a receptor for stress-inducible MICA. Science 1999 Jul 30;285(5428):727e9.

[62] Steinle A, Li P, Morris DL, Groh V, Lanier LL, Strong RK, et al. Interactions of human NKG2D with its ligands

MICA,MICB, and homologs of themouse RAE-1 protein family. Immunogenetics 2001May-Jun;53(4):279e87.

[63] Cosman D, Mullberg J, Sutherland CL, Chin W, Armitage R, Fanslow W, et al. ULBPs, novel MHC class

I-related molecules, bind to CMV glycoprotein UL16 and stimulate NK cytotoxicity through the NKG2D

receptor. Immunity 2001 Feb;14(2):123e33.

[64] Westwood JA, Kelly JM, Tanner JE, Kershaw MH, Smyth MJ, Hayakawa Y. Cutting edge: novel priming of

tumor-specific immunity by NKG2D-triggered NK cell-mediated tumor rejection and Th1-independent

CD4þ T cell pathway. J Immunol 2004 Jan 15;172(2):757e61.

[65] Castriconi R, Cantoni C, Della Chiesa M, Vitale M, Marcenaro E, Conte R, et al. Transforming growth factor

beta 1 inhibits expression of NKp30 and NKG2D receptors: consequences for the NK-mediated killing of

dendritic cells. Proc Natl Acad Sci U S A 2003 Apr 1;100(7):4120e5.

[66] Eisele G, Wischhusen J, Mittelbronn M, Meyermann R, Waldhauer I, Steinle A, et al. TGF-beta and metal-

loproteinases differentially suppress NKG2D ligand surface expression on malignant glioma cells. Brain 2006Sep;129(Pt 9):2416e25.

[67] Groh V, Wu J, Yee C, Spies T. Tumour-derived soluble MIC ligands impair expression of NKG2D and T-cell

activation. Nature 2002 Oct 17;419(6908):734e8.

[68] Borrego F, Ulbrecht M, Weiss EH, Coligan JE, Brooks AG. Recognition of human histocompatibility leukocyte

antigen (HLA)-E complexed with HLA class I signal sequence-derived peptides by CD94/NKG2 confers

protection from natural killer cell-mediated lysis. J Exp Med 1998 Mar 2;187(5):813e8.

[69] Barakonyi A, Rabot M, Marie-Cardine A, Aguerre-Girr M, Polgar B, Schiavon V, et al. Cutting edge: engage-

ment of CD160 by its HLA-C physiological ligand triggers a unique cytokine profile secretion in the cytotoxicperipheral blood NK cell subset. J Immunol 2004 Nov 1;173(9):5349e54.

[70] Le Bouteiller P, Tabiasco J, Polgar B, Kozma N, Giustiniani J, Siewiera J, et al. CD160: a unique activating NK

cell receptor. Immunol Lett 2011 Aug 30;138(2):93e6.

[71] Rabot M, El Costa H, Polgar B, Marie-Cardine A, Aguerre-Girr M, Barakonyi A, et al. CD160-activating NK cell

effector functionsdependon thephosphatidylinositol 3-kinase recruitment. Int Immunol2007Apr;19(4):401e9.

[72] Giustiniani J, Bensussan A, Marie-Cardine A. Identification and characterization of a transmembrane isoform

of CD160 (CD160-TM), a unique activating receptor selectively expressed upon human NK cell activation.

J Immunol 2009 Jan 1;182(1):63e71.

[73] Fons P, Chabot S, Cartwright JE, Lenfant F, L’Faqihi F, Giustiniani J, et al. Soluble HLA-G1 inhibits angio-

genesis through an apoptotic pathway and by direct binding to CD160 receptor expressed by endothelial

cells. Blood 2006 Oct 15;108(8):2608e15.

[74] Cai G, Freeman GJ. The CD160, BTLA, LIGHT/HVEM pathway: a bidirectional switch regulating T-cell

activation. Immunol Rev 2009 May;229(1):244e58.

[75] Latchman Y, Reiser H. Enhanced murine CD4þ T cell responses induced by the CD2 ligand CD48. Eur

J Immunol 1998 Dec;28(12):4325e31.

[76] Stark S, Watzl C. 2B4 (CD244), NTB-A and CRACC (CS1) stimulate cytotoxicity but no proliferation in

human NK cells. Int Immunol 2006 Feb;18(2):241e7.

[77] Veillette A, Dong Z, Latour S. Consequence of the SLAM-SAP signaling pathway in innate-like andconventional lymphocytes. Immunity 2007 Nov;27(5):698e710.

[78] Leibson PJ. Signal transduction during natural killer cell activation: inside the mind of a killer. Immunity

1997 Jun;6(6):655e61.

[79] Lanier LL, Yu G, Phillips JH. Analysis of Fc gamma RIII (CD16) membrane expression and association with

CD3 zeta and Fc epsilon RI-gamma by site-directed mutation. J Immunol 1991 Mar 1;146(5):1571e6.

[80] Fuchs A, Colonna M. The role of NK cell recognition of nectin and nectin-like proteins in tumor

immunosurveillance. Semin Cancer Biol 2006 Oct;16(5):359e66.

Page 13: Cancer Immunotherapy || Immunological Sculpting

CHAPTER 9Immunological Sculpting: Natural Killer-Cell Receptors and Ligands

127

[81] Mace EM, Zhang J, Siminovitch KA, Takei F. Elucidation of the integrin LFA-1-mediated signaling pathway ofactin polarization in natural killer cells. Blood 2010 Aug 26;116(8):1272e9.

[82] Poggi A, Zocchi MR. Antigen presenting cells and stromal cells trigger human natural killer lymphocytes to

autoreactivity: evidence for the involvement of natural cytotoxicity receptors (NCR) and NKG2D. Clin DevImmunol 2006 Jun-Dec;13(2-4):325e36.

[83] Shibuya A, Campbell D, Hannum C, Yssel H, Franz-Bacon K, McClanahan T, et al. DNAM-1, a novel

adhesion molecule involved in the cytolytic function of T lymphocytes. Immunity 1996 Jun;4(6):573e81.

[84] Reymond N, Imbert AM, Devilard E, Fabre S, Chabannon C, Xerri L, et al. DNAM-1 and PVR regulate

monocyte migration through endothelial junctions. J Exp Med 2004 May 17;199(10):1331e41.

[85] Soriani A, Zingoni A, Cerboni C, Iannitto ML, Ricciardi MR, Di Gialleonardo V, et al. ATM-ATR-dependent up-

regulationofDNAM-1andNKG2D ligandsonmultiplemyelomacells by therapeutic agents results in enhanced

NK-cell susceptibility and is associated with a senescent phenotype. Blood 2009 Apr 9;113(15):3503e11.

[86] Gilfillan S, Chan CJ, CellaM,HaynesNM, Rapaport AS, Boles KS, et al. DNAM-1promotes activation of cytotoxic

lymphocytes by nonprofessional antigen-presenting cells and tumors. J ExpMed2008Dec 22;205(13):2965e73.

[87] Shibuya K, Lanier LL, Phillips JH, Ochs HD, Shimizu K, Nakayama E, et al. Physical and functional associ-

ation of LFA-1 with DNAM-1 adhesion molecule. Immunity 1999 Nov;11(5):615e23.

[88] Mizrahi S, Markel G, Porgador A, Bushkin Y, Mandelboim O. CD100 on NK cells enhance IFNgamma

secretion and killing of target cells expressing CD72. PLoS One 2007;2(9):e818.

[89] Kumanogoh A, Watanabe C, Lee I, Wang X, Shi W, Araki H, et al. Identification of CD72 as a lymphocytereceptor for the class IV semaphorin CD100: a novel mechanism for regulating B cell signaling. Immunity

2000 Nov;13(5):621e31.

[90] Ito M, Maruyama T, Saito N, Koganei S, Yamamoto K, Matsumoto N. Killer cell lectin-like receptor G1 bindsthree members of the classical cadherin family to inhibit NK cell cytotoxicity. J Exp Med 2006 Feb

20;203(2):289e95.

[91] Colonna M. Cytolytic responses: cadherins put out the fire. J Exp Med 2006 Feb 20;203(2):261e4.

[92] Meyaard L. The inhibitory collagen receptor LAIR-1 (CD305). J Leukoc Biol 2008 Apr;83(4):799e803.

[93] Meyaard L, Adema GJ, Chang C, Woollatt E, Sutherland GR, Lanier LL, et al. LAIR-1, a novel inhibitory

receptor expressed on human mononuclear leukocytes. Immunity 1997 Aug;7(2):283e90.

[94] Merlo A, Tenca C, Fais F, Battini L, Ciccone E, Grossi CE, et al. Inhibitory receptors CD85j, LAIR-1, andCD152 down-regulate immunoglobulin and cytokine production by human B lymphocytes. Clin Diagn Lab

Immunol 2005 Jun;12(6):705e12.

[95] Vossen MT, Matmati M, Hertoghs KM, Baars PA, Gent MR, Leclercq G, et al. CD27 defines phenotypically and

functionally different human NK cell subsets. J Immunol 2008 Mar 15;180(6):3739e45.

[96] Kelly JM, Darcy PK, Markby JL, Godfrey DI, Takeda K, Yagita H, et al. Induction of tumor-specific T cellmemory by NK cell-mediated tumor rejection. Nat Immunol 2002 Jan;3(1):83e90.

[97] Andre P, Spertini O, Guia S, Rihet P, Dignat-George F, Brailly H, et al. Modification of P-selectin glycoprotein

ligand-1 with a natural killer cell-restricted sulfated lactosamine creates an alternate ligand for L-selectin. ProcNatl Acad Sci U S A 2000 Mar 28;97(7):3400e5.

[98] Cosman D, Fanger N, Borges L, Kubin M, Chin W, Peterson L, et al. A novel immunoglobulin superfamilyreceptor for cellular and viral MHC class I molecules. Immunity 1997 Aug;7(2):273e82.

[99] Vivier E,DaeronM. Immunoreceptor tyrosine-based inhibitionmotifs. Immunol Today 1997 Jun;18(6):286e91.

[100] Ulyanova T, Blasioli J, Woodford-Thomas TA, Thomas ML. The sialoadhesin CD33 is a myeloid-specific

inhibitory receptor. Eur J Immunol 1999 Nov;29(11):3440e9.

[101] Ikehara Y, Ikehara SK, Paulson JC. Negative regulation of T cell receptor signaling by Siglec-7 (p70/AIRM) andSiglec-9. J Biol Chem 2004 Oct 8;279(41):43117e25.

[102] Brunetta E, Fogli M, Varchetta S, Bozzo L, Hudspeth KL, Marcenaro E, et al. The decreased expression ofSiglec-7 represents an early marker of dysfunctional natural killer-cell subsets associated with high levels of

HIV-1 viremia. Blood 2009 Oct 29;114(18):3822e30.

[103] Kawasaki Y, Ito A,WithersDA, Taima T, KakoiN, Saito S, et al. GangliosideDSGb5, preferred ligand for Siglec-7,inhibits NK cell cytotoxicity against renal cell carcinoma cells. Glycobiology 2010 Nov;20(11):1373e9.

[104] Belisle JA, Horibata S, Jennifer GA, Petrie S, Kapur A, Andre S, et al. Identification of Siglec-9 as the receptor

for MUC16 on human NK cells, B cells, and monocytes. Mol Cancer 2010;9:118.

[105] Niloff JM, Knapp RC, Schaetzl E, Reynolds C, Bast Jr RC. CA125 antigen levels in obstetric and gynecologic

patients. Obstet Gynecol 1984 Nov;64(5):703e7.

[106] Patankar MS, Jing Y, Morrison JC, Belisle JA, Lattanzio FA, Deng Y, et al. Potent suppression of natural killer

cell response mediated by the ovarian tumor marker CA125. Gynecol Oncol 2005 Dec;99(3):704e13.

[107] Gubbels JA, Felder M, Horibata S, Belisle JA, Kapur A, Holden H, et al. MUC16 provides immune protection

by inhibiting synapse formation between NK and ovarian tumor cells. Mol Cancer 2010;9:11.


Recommended