Open access peer-reviewed chapter

The Role of Activating and Inhibitory NK Cell Receptors in Antitumor Immune Response

Written By

Gordana Konjević, Ana Vuletić, Katarina Mirjačić Martinović and Radan Džodić

Submitted: 08 December 2016 Reviewed: 12 May 2017 Published: 13 December 2017

DOI: 10.5772/intechopen.69729

From the Edited Volume

Natural Killer Cells

Edited by Mourad Aribi

Chapter metrics overview

2,408 Chapter Downloads

View Full Metrics

Abstract

Natural killer (NK) cells express many newly identified activating and inhibitory receptors that upon engagement by cognate ligands on target tumor cells regulate NK cell antitumor activity. Recently, several paired NK cell receptor families that include receptors with similar binding specificities but opposite function have been defined. The expression of most important activating receptors, natural killer group 2D (NKG2D), natural cytotoxic receptors (NCR), DNAX accessory molecule-1 (DNAM1) and activating killer cell immunoglobulin-like receptors (KAR) is often decreased, while the expression of most prominent inhibitory NK cell receptors, killer cell inhibitory immunoglobulin-like receptors (KIR) and CD94/NKG2A, may occasionally be increased in malignancies. These data indicate that impaired NK cell antitumor response results from NK cell receptor alterations induced by suppressive factors in the tumor microenvironment, including cytokines, growth factors, enzymes and metabolites, as well as by chronic NK cell receptor engagement by the tumor. The established alterations in NK cell receptor expression in cancer patients represent potential disease biomarkers and may aid in choosing therapies that upregulate activating or block inhibitory receptor function. Accumulating knowledge of NK cell biology has been helpful in creating novel therapeutic approaches that by release from tumor-influenced immunosuppression potentiate NK cell activity in cancer patients.

Keywords

  • NK cells
  • inhibitory and activating receptors
  • immunosuppression
  • cancer
  • immunotherapy

1. Introduction

Natural killer (NK) cells are equipped with multiple activating and inhibitory cell surface receptors and play a key role in controlling tumor growth and metastasis. NK cells have originally been described to belong to the innate arm of the immune system and are able to discriminate between normal and transformed cells on the basis of major histocompatibility complex (MHC) class I molecule expression in the organism. As normal cells express major histocompatibility complex (MHC) class I molecules that engage NK cell inhibitory receptors, they are protected from NK cell‐mediated lysis. Therefore, according to the “missing‐self” hypothesis, activation of NK cells occurs in contact with malignantly transformed cells that have lost MHC class I molecules and that have additionally acquired stress‐induced ligands for activating NK cell receptors. Maintenance of NK cell antitumor function relies on the balance between these activating and inhibitory signals mediated by NK cell receptors [1].

Considering that NK cells are defined as CD3CD56+CD16+/− cells according to the density of expression of these receptors, these cells are divided into two subsets. The larger cytotoxic subset with high density of CD16, low density of CD56 (CD56dimCD16bright), and abundant perforin and granzyme granules, and the other, smaller regulatory subset with low density or absence of CD16, high density of CD56 (CD56 brightCD16dim/−) and the ability to produce abundant cytokines including interferon‐gamma (IFN‐γ), tumor necrosis factor (TNF), interleukin (IL)‐10, IL‐13, and granulocyte‐macrophage colony‐stimulating factor (GM‐CSF) [2]. CD56 brightCD16dimsubset has a greater migratory potential and is recruited into tumor tissue, although CD56 dimCD16 brightNK cells have also been detected in certain tumors [3]. Recent data have shown in mucosa‐associated lymph tissue (MALT), the presence of a novel family of tissue‐resident innate lymphoid cells (ILC), with NK‐like characteristics. This novel family is classified into three groups (ILC1, ILC2, and ILC3) and contrary to classical cytotoxic NK cells, considered to belong to ILC1 group, the involvement of other ILC subsets in cancer progression or resistance is still controversial [4].

Several new families of receptors have been recently identified on NK cells and growing knowledge indicates that some families are composed of paired receptors that inspite of binding similar ligands have opposite, activating, or inhibitory function [5]. The most important activating NK cell receptors are NKp46, NKp30, and NKp44 that belong to natural cytotoxic receptors (NCR) family, natural killer group 2D (NKG2D) that belongs to NKG2 calcium‐dependent lectin‐like (NKG2 C‐lectin) family, DNAX accessory molecule 1 (DNAM1) that belongs to the family of nectin‐binding adhesion molecules and activating killer cell immunoglobulin‐like receptors (KAR). On the contrary, the most important family of inhibitory NK cell receptors belongs to killer cell immunoglobulin‐like (KIR) receptor family, while NKG2 C‐lectin family also includes inhibitory CD94/NKG2A NK cell receptors (Table 1). Upon binding ligands, these activating and inhibitory NK cell receptors cooperate and determine NK cell cytotoxicity against transformed cells [6]. However, tumor‐derived immunosuppressive factors play a major role in evading NK cell responses to tumors by interfering with NK cell activation pathways or the complex receptor array that regulate NK cell activation and antitumor activity [3, 7].

The objective of this chapter is to present current data based on the knowledge of many newly identified activating and inhibitory NK cell receptors whose balance regulates NK cell antitumor activity. Numerous alterations in NK cell receptor expression and signaling pathways induced by tumor‐derived immunosuppressive factors that are responsible for poor NK cell cytotoxic function in cancer patients will be defined. Understanding of the alterations in NK cell receptors and function can contribute in the assessment of biomarkers of the state of malignant disease and may aid in the selection of immunotherapy that supports an effective NK cell antitumor response.

Receptor familesLigandsSignaling molecules
NCRActivatingNKp46HSPG, heparin, vimentinFcRγ and CD3ζ, ZAP70/Syk
NKp44NKp44LDAP12, ZAP70/Syk
NKp30a, bB7-H6, BAG-6CD3ζ, ZAP70/Syk
InhibitoryNKp44PCNAITIM, SHP1, 2
NKp30c??
Paired receptors
NKG2ActivatingNKG2DMICA/B, ULBP1-6DAP10, Grb2, Vav, SOS
CD94/NKG2CHLA-EDAP12, ZAP70/Syk
InhibitoryCD94/NKG2AHLA-EITIM, SHP1, 2
CD94/NKG2E
CD94/NKG2F
KIRInhibitory2DL1/L2/L3HLA-CITIM, SHP1, 2
3DL1/L2/L3HLA- A, -B
Activating2DS1/S2/S4HLA-C/?DAP12, ZAP70/Syk
3DS1?
Adhesion moleculesActivatingDNAM-1PVR, nectin-2ITSM, Vav, PLCγ
InhibitoryTIGITPVR, nectin-2ITIM, SHP1, 2
TACTILE
FcγRIIIActivatingCD16Fc IgGFcRγ and CD3ζ, ZAP70/Syk
NKPR1ActivatingCD161??
InhibitoryLLT1ITIM, SHP1, 2

Table 1.

NK cell activating and inhibitory receptors.

Advertisement

2. NK cell receptors

2.1. Natural cytotoxic receptors

The NCR represent a group of human NK cell activating receptors that belong to the immunoglobulin superfamily and include NKp46, NKp30, and NKp44. NKp46 and NKp30 are constitutively expressed on all activated and resting NK cells, whereas NKp44 expression on NK cells requires activation by IL‐2. This makes NKp46 and NKp30 the only NK‐specific markers known today, although recent evidence suggests that a very small subset of T cells expresses NKp46 [8].

Tumor‐associated ligands for most NCR have only been recently described. B7‐H6 was the first identified cellular ligand for an NCR expressed on the surface of cancer cells that binds to the activating receptors NKp30 [9]. Recently, a novel isoform of the mixed lineage leukemia‐5 (MLL5) nuclear protein was proposed as a cancer cell‐expressed ligand for NKp44 [10]. Ligands for NKp46 have been described to have viral structural motives that belong to hemagglutinin‐like molecules or to be heparan sulfate proteoglycans (HSPG) [11]. The cytoplasmatic domains of NCR do not possess any signaling motives except for NKp44 that contains an immunoreceptor tyrosine‐based inhibition motif (ITIM) that has been reported functional only upon binding to certain ligands. Therefore, NCR associate with CD3 ζ‐chain, FcεRIγ and DAP12 adaptor proteins that undergo phosphorylation of their immunoreceptor tyrosine‐based activation motives (ITAM), recruit sarcoma homology 2 (SH2) domains of spleen tyrosine kinase (Syc)/zeta‐chain‐associated protein kinase 70 (ZAP70) and ultimately lead to extracellular signal‐regulated kinase (ERK) activation and cytotoxic granule mobilization [8].

Conversely, it has recently been shown that the constitutively inactive cytoplasmatic ITIM domain of NKp44 receptor becomes functionally active only upon binding proliferating cell nuclear antigen (PCNA) leading to the inhibition of NK cell function [12] that might be involved in tumor immune evasion. Similarly, as NKp30 is comprised of three different isoforms, NKp30a, b, and c it has been shown that binding of NKp30c induces an immunosuppressive signal by producing IL‐10 that is associated with reduced NK cell effector functions. Therefore, the final outcome of NKp44 and NKp30 activation depends on the presence of ligands on target cells, as well as receptor isoforms expressed on the surface of NK cells, respectively [13].

NCR, aside from direct antitumor cytotoxicity can also mediate the production of proinflammatory cytokines by NK cells that have an immunoregulatory role and engage in orchestrating antitumor immune response. In this sense, it has been shown that cross‐linking of NKp46 and NKp44 resulted in NK cell production of IFNγ and TNF that are responsible for dendritic cell (DC) maturation and regulation of adaptive response to tumors. However, binding of inhibitory NKp30c induces production of IL‐10 that reduces NK cell function [14].

Downregulation of NCR expression can be induced by soluble factors such as transforming growth factor beta (TGF‐β), IL‐10, L‐kynurenine, a product of tryptophan degradation by tumor‐derived indolamin‐2,3‐dioxygenase (IDO). Decreased expression of some NCR has been reported in several malignancies, namely low NKp46 in melanoma, pancreatic, gastric, cervical cancer, and acute myeloid leukemia (AML) [1517], NKp44 in numerous solid and hematological malignancies, and NKp30 in breast, hepatocellular cancer (HCC), chronic lymphocytic leukemia (CLL), and AML [15, 16, 1820]. Moreover, ILC3 cells expressing NKp44 activating receptor in human nonsmall‐cell lung cancer (NSCLC) tissue were shown to have tumor protective role [4]. However, overexpression of PCNA in certain tumors results in NKp44 receptor‐mediated inhibition of NK cell activity [8].

2.2. Families of paired receptors in NK cells

2.2.1. NK group 2 calcium‐dependent lectin‐like receptor family

This family of calcium‐dependent lectin‐like receptors includes CD94‐NKG2‐A/C/E/F/H heterodimer and NKG2D homodimer receptors present on NK cells. This family is composed of paired NK cells receptors with similar binding specificities to nonclassical MHC class I molecules on the surface of potential target cells but either activating or inhibitory functions [21].

2.2.1.1. Natural killer group 2D

It is an activating receptor expressed on virtually all NK cells. NKG2D recognizes a number of MHC‐class‐I‐related molecules, major histocompatibility complex class I chain‐related molecule (MIC) A/B and UL16‐binding protein (ULBP) expressed on cells in stressful conditions, such as transformation [22]. Owing to their ability to trigger activation of the immune system, it has now been shown that their expression is very tightly controlled at transcriptional level [23]. NKG2D is a pivotal activating receptor that upon binding stress‐induced ligands and phosphorylation of intracellular adaptor protein DNAX‐activation protein 10 (DAP10) recruits various signaling pathways and induces tumor cytotoxicity [24].

Decreased expression of NKG2D in cancer patients is mediated by TGFβ and IL‐10 produced by tumor cells and immunosuppressive immune cells, tumor‐associated macrophages (TAM), myeloid‐derived suppressor cells (MDSC), regulatory T cells (Treg) in the tumor microenvironment [25]. Also, persistent stimulation of NKG2D receptor by its tumor cell‐associated ligands, as well as, soluble ligands induced by matrix metalloproteinases (MMP) proteolytic cleavage from tumor cells may lead to functional exhaustion of NK cells [26].

Downregulation of NKG2D expression together with degradation of its intracellular signal transducing adapter, DAP10 [27], by its ligand has also been shown in experimental settings of conjugate formation between NK cells, not only of healthy individuals but also of metastatic melanoma patients, with K562, an erythromyeloid cell line, or FemX, a melanoma‐derived tumor cell line [2830]. This effect is more pronounced for K562 tumor cells as they highly express MICA/B ligands specific for NKG2D receptors [31], while for FemX tumor cell line so far no data exist regarding the expression of MICA/B ligands, although it can be assumed that, as most tumor cells, this cell line expresses some level of these stress‐induced ligands. The importance of NKG2D receptor in NK cell cytotoxic function underlies a significant positive correlation of its expression with NK cytotoxicity in healthy individuals, as well as in melanoma patients, in spite of different levels of expression [3234].

These data indicate that activating NKG2D receptor has a role in NK cell tumor immunosurveillance and in immune‐mediated rejection of tumor cells and that NKG2D downregulation as a consequence of tumor immunoediting favors tumor progression [13].

2.2.1.2. Heterodimeric NK group 2 calcium‐dependent lectin‐like A/B/C/E/H receptors

These heterodimer receptors consist of CD94 subunit that is associated with the member of the NKG2 family (A/B/C/E/H). Prototypic member of this family CD94‐NKG2A has an ITIM that consequently leads to the inhibition of NK cell activity by inducing dephosphorylation of surrounding tyrosine kinases and adaptor proteins. By contrast, receptors CD94‐NKG2‐C, CD94‐NKG2‐E, and CD94‐NKG2‐H, associate with DAP12 and function as activating receptors that participate in NK cell antitumor response. These receptors bind to nonclassical MHC class I, human leukocyte antigen (HLA)‐E molecules on the surface of malignantly transformed target cells [21].

Interestingly, the ligand for this receptor family, HLA‐E molecule assembles at the endoplasmic reticulum with peptides derived from the leader peptides of HLA‐A, B, C, and G molecules and as in malignant transformation, the expression of classical MHC class I molecules is downregulated, the expression of HLA‐E is consequently upregulated [35]. The importance of the inhibitory activity of NKG2A receptor expression is demonstrated in breast cancer and colorectal cancer (CRC) as its increased expression is associated with poor disease prognosis [18, 36]. Conversely, decreased expression of NKG2C activating receptor resulting in NK cell dysfunction has so far been reported for AML [37].

2.2.2. Killer cell immunoglobulin‐like receptor family

This is a well‐known paired NK cell receptor family with either inhibitory or activating functions that interact specifically with MHC class I molecules and their tissue protective role is based on the higher binding affinity and signal transduction by inhibitory KIR receptors, compared to activating KAR receptors, for MHC class I ligands. According to KIR expression on NK cells, they are divided into KIR haplotype A and B, with A being more frequent and including inhibitory receptors compared to B that includes both types of receptors with predominance of activating receptors [38].

The most prominent inhibitory killer cell immunoglobulin‐like receptors (KIR) are KIR2DL1 (CD158a), KIR2DL2/3 (CD158b), and KIR3DL1‐2 and inhibit NK cell activity through an ITIM by recruiting SH2 domain protein tyrosine phosphatases (SHP)‐1 and ‐2 and adaptor proteins, including DAP‐10. Moreover, blocking actin cytoskeleton‐dependent raft recruitment of different receptors may be a general mechanism by which inhibitory receptors control NK cell activation [39]. Thus, activating KAR, such as KIR2DS1, KIR2DS4,and KIR2DL4, associate with DAP12 adaptor protein that deliver activation signals through an ITAM that after phosphorylation by sarcoma (Src) family kinases recruit Syk/ZAP‐70 tyrosine kinases to mediate downstream activation signaling [40]. Clinical studies have correlated KIR gene content with cancer [41]. It has recently been reported that higher expression of inhibitory CD158a and CD158b receptors, especially in patients that express their specific HLA‐C ligands, is associated not only with susceptibility to tumors but also with disease progression, shown by increase in NK cells with these receptors in advanced stages of melanoma [42, 43].

Inhibitory KIR receptor upregulation on NK cells in malignant tumors has been reported to be associated with immunosuppressive, as well as immunostimulatory cytokines [4448]. Higher expression of certain inhibitory KIR receptors has been shown in pancreatic, gastric, and CRC without association with disease progression [15]. Other studies in patients with metastatic melanoma, as well as in NSCLC and breast cancer, show an increase in the expression of CD158a and CD158b inhibitory KIR receptors on NK cells [33, 4951] that in melanoma negatively correlates with NK cell cytotoxicity [33]. In this sense, as inhibitory KIR play prominent roles in regulating NK cell activation therapeutic strategies designed to diminish KIR function that may be able to potentiate NK cell activity in treating patients with malignancies.

2.2.3. Family of nectin‐binding adhesion molecules

It is an important family of adhesion molecules that includes CD226 (DNAM1), CD96 (T cell‐activated increased late expression (TACTILE)), and T‐cell immunoglobulin and ITIM domain (TIGIT). These receptors bind nectin proteins, CD112 (nectin‐2), and CD155 (poliovirus receptor (PVR)) and have been recently identified as crucial regulators of NK cell function. This is another family of paired NK cell receptors with similar binding specificities on the surface of target cells but with either activating (CD226) or inhibitory (CD96 and TIGIT) functions [5].

2.2.3.1. Stimulatory nectin‐binding adhesion molecule

Costimulatory adhesion receptor DNAM1, a member of immunoglobulin‐superfamily has recently been shown to have a role in the recognition of tumor cells and NK cell‐mediated responses to tumors. It binds to PVR and nectin‐2, and recruits the tyrosine kinase fibroblast endothelial kinase (Fyn) and serine threonine protein kinase C (PKC). Moreover, interaction of DNAM1 with target cell ligands induces actin polymerization and activation of other surface receptors, which permit stable interaction of NK cells and target cells. Furthermore, the involvement of this receptor in the NK cell‐mediated responses to tumors is beginning to be elucidated and results to date suggest that DNAM1 has a role in the recognition of tumor cells, as well as migration of NK cells [35, 52].

The importance of DNAM1 has been reported in breast cancer, CRC, AML, and melanoma [17, 18, 37, 53] and in melanoma, it has been shown that tumor‐associated fibroblasts (TAF) by modulating the surface expression of DNAM‐1 based on cell‐to‐cell interactions could inhibit NK cell function [54]. Also, DNAM1 ligand CD155 upregulation on multiple myeloma (MM) cells has been reported and resulted in increased sensitivity to NK cell‐mediated lysis [55].

2.2.3.2. Inhibitory nectin‐binding adhesion molecules

TIGIT and TACTILE (CD96) receptors that unlike activating DNAM1 receptor contain an ITIM motive subsequently inhibit NK cell antitumor activity and counteract DNAM‐mediated activation. These receptors are important in settings in which the tumor is mainly nonimmunogenic, as it does not express stress ligands or costimulatory molecules, which is a common situation for many epithelial cell malignancies. For this reason, these inhibitory molecules may be promising therapeutic targets for the treatment of malignancies [5].

2.3. CD16 (Fc γ receptor IIIA)

This is one of the most important NK cell cytotoxic receptors that contains two extracellular Ig‐like domains and is involved in both direct [56] and, as an Fc gamma receptor type IIIA, is involved in antibody‐dependent cell‐mediated cytotoxicity (ADCC) [57], as well as cytokine production, proliferation, and postactivational NK cell apoptotic death [56].

CD16, as well as NKp46, associates with two cytoplasmatic domains composed of FcεRIγ or T‐cell receptor (TCR) ζ chains that comprise ITAM, which upon ligand‐binding become phosphorylated and induce signal transduction by activation of nonreceptor tyrosine kinases Syk and ZAP‐70 [57].

The expression CD16 as a prominent NK cell cytotoxic receptor has been found to be decreased on NK cells in breast cancer and MM patients [18, 58] not only due to postactivational receptor internalization but also following target cell induced activation of MMP, namely ADAM 17, as shown during in vitro NK cell cultivation with tumor cells [59, 60]. Moreover, as CD16 defines the two functionally different NK cell subsets, its decreased expression influences the ratio of these subsets, leading to the loss CD16bright cytotoxic subset, a finding that has been detected in numerous malignancies such as breast cancer, MM, and melanoma [6, 33].

2.4. Natural killer cell surface protein P1A

This is a member of the C‐type lectin family that is primarily designated as an activating receptor that plays a role in NK cell‐mediated cytotoxicity and it has been previously shown that natural killer cell surface protein P1A (NKR‐P1A)/CD161 participates in triggering NK cell cytotoxicity against numerous human tumor cell lines [61], while more recently, it has been suggested that its activating mechanism may require interaction with costimulatory receptors [62]. On identification of its lectin‐like transcript 1 (LLT1) ligand, its inhibitory potential has recently been described, but its function still remains controversial. In support of the inhibitory function of NKR‐P1A, it has been shown that its cytoplasmic tail contains tyrosine residue in an atypical motive (AxYxxL) that may function as a weak ITIM [63, 64]. There are some reports of decreased expression and cytokine‐mediated upregulation of NKR‐P1A/CD161 in metastatic melanoma and MM [29, 33, 58].

Advertisement

3. Tumor‐induced immunosuppression

It has now been well documented that tumor‐induced immunosuppression affects NK cell receptor repertoire and leads to progressive local and systemic inhibition of NK cell function. In tumors, a complex composition of immunosuppressive molecules TGF‐β, IL‐10, IDO, prostaglandin E2 (PGE2), vascular endothelial growth factor (VEGF), nitric oxide synthase (NOS), and reactive oxygen species (ROS) are produced by regulatory immune cells such as Treg, MDSC, TAM, and by tumor cells themselves (Figure. 1). These factors generate a chronic inflammatory immunosuppressive milieu that leads to the suppression of the antitumor effector NK cell function that supports tumor progression [3, 7, 13, 65]. In this sense, considering that the expression of activating NK cell receptors is decreased in most malignancies, and as some studies report that the expression of inhibitory KIR and CD94/NKG2A receptors on NK cells in different tumors remains unchanged, this suggests that activating receptors are main targets of tumor‐mediated suppression [13].

Figure 1.

Mechanisms of NK cell receptor dysregulation in tumors. The tumor induces alterations in NK cell activating receptors by producing suppressive tumor‐derived mediators including immunosuppressive cytokines (transforming growth factor beta - TGF‐β), enzymes (indolamin‐2,3‐dioxygenase - IDO), factors (L‐kynurenine, prostaglandin E2 - PGE2) that together with the presence of hypoxia can suppress NK cell antitumor activity. Chronic engagement of NK cell activating receptors with either tumor cell surface‐expressed or shed NK cell ligands (NKG2D ligand - NKG2D-L) leads to progressive inhibition of NK cell antitumor response. Also, tumor‐expressed proliferating cell nuclear antigen (PCNA) associated with HLA‐I molecule by binding NKp44 activating receptor induces unconventional inhibitory signals in NK cells. Moreover, interferon gamma (IFN‐γ) released by NK cells induces increased expression of HLA‐I molecules on tumor cells, as well as tumor‐produced IDO, that lead to inhibition of NK cell function.

Furthermore, NK cell dysfunction due to decreased activating NK cell receptor expression may be mediated by chronic tumor cell ligand‐NK cell receptor engagement that leads to an exhausted NK cell phenotype characterized by upregulated programmed death 1 (PD‐1) checkpoint immunoreceptor expression [66]. Conversely, the appearance of soluble NK cell ligands due to proteolytic cleavage from tumor cells also leads to NK cell dysfunction due to chronic NK cell receptor stimulation in the absence of target tumor cells [26].

In this sense, it has now been established that during the development of solid tumors, NK cells are frequently rendered functionally impaired as a consequence of cancer immunoediting that induces immune tolerance to tumors owing to impairment of NK cell receptor repertoire and signaling, as well as immunoselection of nonimmunogenic tumor cells.

Advertisement

4. NK cell receptors as therapeutic targets in cancer immunotherapy

As NK cell antitumor activity is regulated by numerous activating and inhibitory NK cell receptors, alterations in NK cell receptor expression and signaling underlie diminished cytotoxic NK cell function. Based on this and on predictive in vitro findings [6, 4648, 6770], cytokines, including IFNα, IL‐2, IL‐12, IL‐15, and IL‐18 have been used systemically or for ex vivo–activation and expansion of NK cells and have led to improved NK cells antitumor activity by increasing the expression of NK cell activating receptors and by inducing cytotoxic effector molecules [7175]. Moreover, this cytokine‐based therapy enhances NK cell proliferation and regulatory function, and it has been shown that it induces NK cells exhibiting cytokine induced memory‐like properties [76] that represent a newly‐defined NK cell subset with improved NK cell activity and longevity.

Considering the effect of tumor‐derived immunosuppressive molecules on the decrease in the expression of activating NK cell receptors, early stage clinical trials have been introduced that use monoclonal antibodies, alone or in combination, to neutralize TGF‐β, IDO, or PD‐1 checkpoint inhibitor in different malignancies that led to improved antitumor NK cell function [77].

Since inhibitory KIR play prominent roles in regulating NK cell activation, therapeutic strategies in cancer to diminish KIR function have been developed. In autologous settings, anti‐inhibitory KIR monoclonal antibody therapy has been introduced to support autologous NK cell administration by rendering them with higher antitumor activity, as reported in AML and MM. On the other hand, allogeneic hematopoietic stem cell transplants (HSCT) based on partially KIR/HLA mismatched alloreactive NK cell transfer that relieve donor NK cells from inhibition by recipient’s MHC class I molecules, show beneficial graft‐versus‐tumor (GvT) effect in both pediatric and adult high‐risk leukemia [78].

Moreover, it has been recognized that classical and novel pharmacological agents, such as proteasome inhibitors or histonedeacetylase inhibitors and certain chemotherapeutics [72, 74, 75], upregulate cognate ligands for activating receptors on tumor cells and provide better NK cell antitumor response.

Also, a new effective approach, in clinical trials, designed to enhance NK cell—tumor cell interaction includes genetically modified NK cells expressing cytokine transgenes, chimeric antigen receptors (CARs), or overexpressing activating receptors that recognize NK cell ligands on tumor cells are showing promising results.

As it has been shown that therapeutic antibodies that are already in use for cancer treatment also trigger NK cell‐mediated ADCC activity [79, 80] have been modified to increase binding of their Fc fragment to CD16 on NK cells. These therapeutic antibodies by binding CD16 receptor induce a potent activating signal, which overcomes inhibitory signals and results in both cytotoxicity and cytokine responses [81] that enhance NK cell activation against tumor cells.

Advertisement

5. Conclusion

The knowledge of NK cells has grown to include many newly identified activating and inhibitory NK cell receptors and defined alterations in receptor expression and signaling pathways that are responsible for poor NK cell cytotoxic function in cancer patients, may be considered as biomarkers of the state of disease. These findings regarding NK cell receptors may aid in the selection of classical and newly developed therapies that favorably modulate NK cell receptor expression and function, and release them from tumor‐derived immunosuppression in order to achieve an effective NK cell antitumor response.

Advertisement

Acknowledgments

This work was supported by the grants of the Ministry of Education, Science and Technological Development of the Republic of Serbia, numbers III41031 and 175056.

References

  1. 1. Caligiuri MA. Human natural killer cells. Blood. 2008;112:461-469
  2. 2. Cooper MA, Fehniger TA, Caligiuri MA. The biology of human natural killer‐cell subsets. Trends in Immunology. 2001;22:633-640
  3. 3. Vitalle M, Cantoni C, Pietra G, Mingari MC, Moretta L. Effect of tumor cells and tumor microenvironment on NK‐cell functionEuropean Journal of Immunology. 2014;44:1582-92
  4. 4. Montaldo E, Vacca P, Vitale C, Moretta F, Locatelli F, Mingari MC, Moretta L. Human innate lymphoid cells. Immunology Letters. 2016;179:2-8
  5. 5. Martinet L, Smyth MJ. Balancing natural killer cell activation through paired receptors. Nature Reviews Immunology. 2015;15:243-54
  6. 6. Konjević G, Vuletić A, Mirjačić Martinović K. Natural killer cell receptors: Alterations and therapeutic targeting in malignancies. Journal of Immunology Research. 2016;64:25-35
  7. 7. Pahl J, Cerwenka A. Tricking the balance: NK cells in anti‐cancer immunity. Immunobiology. 2017;222:11-20
  8. 8. Kruse PH, Matta J, Ugolini S, Vivier E. Natural cytotoxicity receptors and their ligands. Immunology and Cell Biology. 2014;92:221-229
  9. 9. 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. Journal of Experimental Medicine. 2009;206:1495-1503
  10. 10. Baychelier F, Sennepin A, Ermonval M, Dorgham K, Debré P, Vieillard V. Identification of a cellular ligand for the natural cytotoxicity receptor NKp44. Blood. 2013;122:2935-2942
  11. 11. Arnon TI, Achdout H, Lieberman N, Gazit R, Gonen‐Gross T, Katz G, et al. The mechanisms controlling the recognition of tumor‐ and virus‐infected cells by NKp46. Blood. 2004;103:664-672
  12. 12. Rosental B, Brusilovsky M, Hadad U, Oz D, Appel MY, Afergan F, et al. Proliferating cell nuclear antigen is a novel inhibitory ligand for the natural cytotoxicity receptor NKp44. The Journal of Immunology. 2011;187:5693-5702
  13. 13. Stojanovic A, Correia MP, Cerwenka A. Shaping of NK cell responses by the tumor microenvironment. Cancer Microenvironment. 2013;6:135-46
  14. 14. Hudspeth K, Silva‐Santos B, Mavilio D. Natural cytotoxicity receptors: Broader expression patterns and functions in innate and adaptive immune cells. Frontiers in Immunology. 2013;4:69
  15. 15. Peng YP, Zhu Y, Zhang JJ, Xu ZK, Qian ZY, Dai CC, et al. Comprehensive analysis of the percentage of surface receptors and cytotoxic granules positive natural killer cells in patients with pancreatic cancer, gastric cancer, and colorectal cancer. Journal of Translational Medicine. 2013;11:262
  16. 16. Garcia‐Iglesias T, Del Toro‐Arreola A, Albarran‐Somoza B, Del Toro‐Arreola S, Sanchez‐Hernandez PE, Ramirez‐Duenas MG, et al. Low NKp30, NKp46 and NKG2D expression and reduced cytotoxic activity on NK cells in cervical cancer and precursor lesions. BMC Cancer. 2009;9:186
  17. 17. Mirjačić Martinović KM, Babović NLJ, Džodić RR, Jurišić VB, Tanić NT, Konjević GM. Decreased expression of NKG2D, NKp46, DNAM‐1 receptors, and intracellular perforin and STAT‐1 effector molecules in NK cells and their dim and bright subsets in metastatic melanoma patients. Melanoma Research. 2014;24:295-304
  18. 18. Mamessier E, Sylvain A, Bertucci F, Castellano R, Finetti P, Houvenaeghel G, et al. Human breast tumor cells induce self‐tolerance mechanisms to avoid NKG2D‐mediated and DNAM‐mediated NK cell recognition. Cancer Research. 2011;71:6621-6632
  19. 19. Fiegler N, Textor S, Arnold A, Rolle A, Oehme I, Breuhahn K, et al. Downregulation of the activating NKp30 ligand B7–H6 by HDAC inhibitors impairs tumor cell recognition by NK cells. Blood. 2013;122:684-93
  20. 20. Reiners KS, Topolar D, Henke A, Simhadri VR, Kessler J, Sauer M, et al. Soluble ligands for NK cell receptors promote evasion of chronic lymphocytic leukemia cells from NK cell anti‐tumor activity. Blood. 2013;121:3658-65
  21. 21. Braud VM, Allan DS, O’Callaghan CA, Söderström K, D’Andrea A, Ogg GS, et al. HLA‐E binds to natural killer cell receptorsCD94/NKG2A, Band C. Nature. 1998;391:795-799
  22. 22. Lopez‐Larrea C, Suarez‐Alvarez B, Lopez‐Soto A, Lopez‐Vazquez A, Gonzalez S. The NKG2D receptor: Sensing stressed cells. Trends in Molecular Medicine. 2008;14:179-89
  23. 23. Zafirova B, Wensveen FM, Gulin M, Polić B. Regulation of immune cell function and differentiation by the NKG2D receptor. Cellular and Molecular Life Sciences. 2011;68:3519-3529
  24. 24. Wu J, Song Y, Bakker AB, Bauer S, Spies T, Lanier LL, et al. An activating immunoreceptor complex formed by NKG2D and DAP10. Science. 1999;285:730-732
  25. 25. Schiavoni G, Gabriele L, Mattei F. The tumor microenvironment: A pitch for multiple players. Frontiers in Oncology. 2013;3:90
  26. 26. Chitadze G, Bhat J, Lettau M, Janssen O, Kabelitz D. Generation of soluble NKG2D ligands: Proteolytic cleavage, exosome secretion and functional implications. Scandinavian Journal of Immunology. 2013;78:120-129
  27. 27. Gonzalez‐Gugel E, Saxena M, Bhardwaj N. Modulation of innate immunity in the tumor microenvironment. Cancer Immunology, Immunotherapy. 2016;65:1261-1268
  28. 28. Konjević G, Mirjacić Martinović K, Vuletić A, Jurišić V, Spužić I. Distribution of several activating and inhibitory receptors on CD3‐CD16+NKcells and their correlation with NK cell function in healthy individuals. The Journal of Membrane Biology. 2009;230:113-123
  29. 29. Mirjačić Martinović K, Konjević G, Babović N, Inić M. The stage dependent changes in NK cell activity and the expression of activating and inhibitory NK cell receptors in melanoma patients. Journal of Surgical Research. 2011;171:637-649
  30. 30. Konjević G, Vuletić A, Mirjacić Martinović K. Changes in NK cell effector function, subset distribution and receptor repertoire following in vitro NK cell and K562 tumor cell contact in melanoma patients. In: EACR Proceedings Book 5-8 June 2014; Munich. Germany: European Journal of Cancer; 50(Suppl. 5):p 220
  31. 31. Ito N, DeMarco RA, Mailliard RB, Han J, Rabinowich H, Kalinski P, et al. Cytolytic cells induce HMGB1 release from melanoma cell lines. Journal of Leukocyte Biology. 2007; 81:75-83
  32. 32. Konjević G, Mirjačić Martinović K, Vuletić A, Jović V, Jurišić V, Babović N, et al. Low expression of CD161 and NKG2D activating NK receptor is associated with impaired NK cell cytotoxicity in metastatic melanoma patients. Clinical & Experimental Metastasis. 2007;24:1-11
  33. 33. Konjević G, Mirjacić Martinović K, Jurisić V, Babović N, Spuzić I. Biomarkers of suppressed natural killer (NK) cell function in metastatic melanoma: Decreased NKG2D and increased CD158a receptors on CD3‐CD16+ NK cells. Biomarkers. 2009;14:258-270
  34. 34. Mirjačić Martinović K, Srdić‐Rajić T, Babović N, Džodić R, Jurišić V, Konjević G. Decreased expression of pSTAT, IRF‐1 and DAP10 signalling molecules in peripheral blood lymphocytes of patients with metastatic melanoma. Journal of Clinical Pathology. 2016;69:300-306
  35. 35. Pegram HJ, Andrews DM, Smyth MJ, Darcy PK, Kershaw MH. Activating and inhibitory receptors of natural killer cells. Immunology and Cell Biology. 2011;89:216-224
  36. 36. Bossard C, Bezieau S, Matysiak‐Budnik T, Volteau C, Laboisse CL, Jotereau F, et al. HLA‐E/b2 microglobulin overexpression in colorectal cancer is associated with recruitment of inhibitory immune cells and tumor progression. International Journal of Cancer. 2012;131:855-863
  37. 37. Sanchez‐Correa B, Morgado S, Gayoso I, Bergua JM, Casado JG, Arcos MJ, et al. Human NK cells in acute myeloid leukaemia patients: Analysis of NK cell‐activating receptors and their ligands. Cancer Immunology, Immunotherapy. 2011;60:1195-1205
  38. 38. Uhrberg M, Valiante NM, Shum BP, Shilling HG, Lienert‐Weidenbach K, Corliss B, et al. Human diversity in killer cell inhibitory receptor genes. Immunity. 1997;7:753-763
  39. 39. Guia S, Jaeger BN, Piatek S, Mailfert S, Trombik T, Fenis A, et al. Confinement of activating receptors at the plasma membrane controls natural killer cell tolerance. Science Signaling. 2011;4(167):ra21
  40. 40. Purdy AK, Campbell KS. Natural killer cells and cancer: Regulation by the killer cell Ig‐like receptors (KIR). Cancer Biology & Therapy. 2009;8:2211-2220
  41. 41. Boyton RJ, Altmann DM. Natural killer cells, killer immunoglobulin‐like receptors and human leucocyte antigen class I in disease. Clinical & Experimental Immunology. 2007;149:1-8
  42. 42. Campillo JA, Martınez‐Escribano JA, Moya‐Quiles MR, Marın LA, Muro M, Guerra N, et al. Natural killer receptors on CD8 T cells and natural killer cells from different HLA‐C phenotypes in melanoma patients. Clinical Cancer Research. 2006;12:4822-4831
  43. 43. Naumova E, Mihaylova A, Ivanova M, Mihailova S. Impact of KIR/HLA ligand combinations on immune responses in malignant melanoma. Cancer Immunology, Immunotherapy. 2007;56:95-100
  44. 44. Zwirner NW, Domaica CI. Cytokine regulation of natural killer cell effector functions. BioFactors. 2010;36:274-288
  45. 45. Konjević G, Mirjačić Martinović K, Vuletić A, Radenković S. Novel aspects of in vitro IL‐2 or IFN‐α enhanced NK cytotoxicity of healthy individuals based on NKG2D and CD161 NK cell receptor induction. Biomedicine & Pharmacotherapy. 2010;64:663-671
  46. 46. Mirjačić Martinović K, Babović N, Džodić R, Jurišić V, Matković S, Konjević G. Favorable in vitro effects of combined IL‐12 and IL‐18 treatment on NK cell cytotoxicity and CD25 receptor expression in metastatic melanoma patients. Journal of Translational Medicine. 2015;13:120
  47. 47. Vuletić AM, Jovanć IP, Jurišić VB,Milovanoviić ZM, Nikolić SS, Tanić NT, et al. In‐vitro activation of natural killer cells from regional lymph nodes of melanoma patients with interleukin‐2 and interleukin‐15. Melanoma Research. 2015;25:22-34
  48. 48. Mirjačić Martinović KM, Babović NL, Džodić RR, Jurišić VB, Ninković AZ, Konjević GM. Beneficial in‐vitro effects of interleukin‐2, interleukin‐12, and their combination on functional and receptor characteristics of natural killer cells in metastatic melanoma patients with normal serum lactate dehydrogenase levels. Melanoma Research. 2016;26:551-564
  49. 49. Varker KA, Terrell CE, Welt M, Suleiman S, Thornton L, Andersen BL, et al. Impaired natural killer cell lysis in breast cancer patients with high levels of psychological stress is associated with altered expression of killer immunoglobulin like receptors. Journal of Surgical Research. 2007;139:36-44
  50. 50. Al Omar SY, Marshall E, Middleton D, Christmas SE. Increased killer immunoglobulin‐like receptor expression and functional defects in natural killer cells in lung cancer. Immunology. 2011;133:94-104
  51. 51. Vuletić A, Jurišić V, Jovanić I, Milovanović Z, Nikolić S, Konjević G. Distribution of several activating and inhibitory receptors on CD3(‐)CD56(+) NK cells in regional lymph nodes of melanoma patients. Journal of Surgical Research. 2013;183:860-868
  52. 52. Lakshmikanth T, Burke S, Ali TH, Kimpfler S, Ursini F, Ruggeri L, et al. NCRs and DNAM‐1 mediate NK cell recognition and lysis of human and mouse melanoma cell lines in vitro and in vivo. Journal of Clinical Investigation. 2009;119:1251-1263
  53. 53. Zhang Z, Su T, He L, Wang H, Ji G, Liu X, et al. Identification and functional analysis of ligands for natural killer cell activating receptors in colon carcinoma. Tohoku Journal of Experimental Medicine. 2012;226:59-68
  54. 54. Balsamo M, Scordamaglia F, Pietra G, Manzini C, Cantoni C, Boitano M, et al. Melanoma‐associated fibroblasts modulate NK cell phenotype and antitumor cytotoxicity. Proceedings of the National Academy of Sciences of the United States of America. 2009;106:20847-20852
  55. 55. Morisaki T, Onishi H, Katano M. Cancer immunotherapy using NKG2D and DNAM‐1systems. Anticancer Research. 2012;32:2241-2247
  56. 56. Warren HS, Kinnear BF. Quantitative analysis of the effect of CD16 ligation on human NK cell proliferation. Journal of Immunology. 1999;162:735-42
  57. 57. Lanier LL. Natural killer cell receptor signaling. Current Opinion in Immunology. 2003;15:308-314
  58. 58. Konjević G, Vuletić A, Mirjačić Martinović K, Colović N, Čolović M, Jurišić V. Decreased CD161 activating and increased CD158a inhibitory receptor expression on NK cells underlies impaired NK cell cytotoxicity in patients with multiple myeloma. Journal of Clinical Pathology. 2016;pii:jclinpath‐2016‐203614. DOI: 10.1136/jclinpath‐2016‐203614
  59. 59. Jewett A, Tseng HC. Tumor induced inactivation of natural killer cell cytotoxic function; implication in growth, expansion and differentiation of cancer stem cells. Journal of Cancer.2011;2:443-457
  60. 60. Romee R, Foley B, Lenvik T, Wang Y, Zhang B, Ankarlo D, et al. NK cell CD16 surface expression and function is regulated by a disintegrin and metalloprotease‐17 (ADAM17). Blood. 2013;121:3599-3608
  61. 61. Azzoni L, Zatsepina O, Abebe B, Bennett IM, Kanakaraj P, Perussia B. Differential transcriptional regulation of CD161 and a novel gene, 197/15a, by IL‐2, IL‐15, and IL‐12 in NK and T cells. Journal of Immunology. 1998;161:3493-3500
  62. 62. Lanier LL, Chang C, Phillips JH. Human NKR‐P1A. A disulfide‐ linked homodimer of the C‐type lectin superfamily expressed by a subset of NK and T lymphocytes. Journal of Immunology. 1994;153:2417-2428
  63. 63. Ryan JC, Seaman WE. Divergent functions of lectin‐like receptors on NK cells. Immunological Reviews. 1997;155:79-89
  64. 64. Rosen DB, Bettadapura J, Alsharifi M, Mathew PA, Warren HS, Lanier LL. Cutting edge: Lectin‐like transcript‐1 is a ligand for the inhibitory human NKR‐P1A receptor. Journal of Immunology. 2005;175:7796-7799
  65. 65. Vuletić A, Jovanić I, Jurišić V, Milovanović Z, Nikolić S, Spurnić I, et al. Decreased Interferon γ Production in CD3+ and CD3‐ CD56+ Lymphocyte Subsets in Metastatic Regional Lymph Nodes of Melanoma Patients. Pathology & Oncology Research. 2015;21:1109-1114
  66. 66. Della Chiesa M, Pesce S, Muccio L, Carlomagno S, Sivori S, Moretta A, Marcenaro E. Features of memory‐like and PD‐1(+) human NK cell subsets. Frontiers in Immunology. 2016;7:351. DOI: 10.3389/fimmu.2016.00351
  67. 67. Konjević G, Mirjačić Martinović K, Vuletić A, Babović N. In‐vitro IL‐2 or IFN‐a‐induced NKG2D and CD161 NK cell receptor expression indicates novel aspects of NK cell activation in metastatic melanoma patients. Melanoma Research. 2010;20:459-467
  68. 68. Konjevic G, Mirjacic‐Martinovic K, Vuletic A, Babovic N. In vitro increased natural killer cell activity of metastatic melanoma patients with interferon‐α alone as opposed to its combination with 13‐cis retinoic acid is associated with modulation of NKG2D and CD161 activating receptor expression. Journal of BUON. 2012;17:761-769
  69. 69. Konjević G, Jurišić V, Jović V, Vuletić A, Mirjačić Martinović K, Radenković S, et al. Investigation of NK cell function and their modulation in different malignancies. Journal of Immunology Research. 2012;52:139-156
  70. 70. Mirjačić Martinović KM, Vuletić AM, Lj Babović N, Džodić RR, Konjević GM, Jurišić VB. Attenuated in vitro effects of IFN‐α, IL‐2 and IL‐12 on functional and receptor characteristics of peripheral blood lymphocytes in metastatic melanoma patients. Cytokine. 2017;96:30-40
  71. 71. Konjević G, Spužić I. Stage dependence of NK cell activity and its modulation by interleukin 2 in patients with breast cancer. Neoplasma. 1993;40:81-85
  72. 72. Konjević G, Jović V, Radulović S, Jelić S, Džodić R, Spužić I. Therapeutic implications of the kinetics of immunomodulation during single or combined treatment of melanoma patients with dacarbazine and interferon‐alpha. Neoplasma. 2001;48:175-181
  73. 73. Konjević G, Jović V, Radomirović V, Spuzić I: [Correlation between functional capability and phenotypic characteristics of peripheral blood lymphocytes in patients with malignant melanoma]. Glas Srp Akad Nauka Med. 2002;73:121-36
  74. 74. Konjević G, Jović V, Jurisić V, Radulović S, Jelić S, Spuzić I. IL‐2‐mediated augmentation of NK‐cell activity and activation antigen expression on NK‐ and T‐cell subsets in patients with metastatic melanoma treated with interferon‐alpha and DTIC. Clinical & Experimental Metastasis. 2003;20:647-655
  75. 75. Konjević G, Jović V, Vuletić A, Radulović S, Jelić S, Spužić I. CD69 on CD56+ NK cells and response to chemoimmunotherapy in metastatic melanoma. European Journal of Clinical Investigation. 2007;37:887-896
  76. 76. Ni J, Miller M, Stojanovic A, Garbi N, Cerwenka A. Sustained effector function of IL‐12/15/18‐preactivated NK cells against established tumors. Journal of Experimental Medicine. 2012;209:2351-2365
  77. 77. Adams JL, Smothers J, Srinivasan R, Hoos A. Big opportunities for small molecules in immuno‐oncology. Nature Reviews Drug Discovery. 2015;14:603-622
  78. 78. Pietra G, Vitale C, Pende D, Bertaina A, Moretta F, Falco M, et al. Human natural killer cells: News in the therapy of solid tumors and high‐risk leukemias. Cancer Immunology, Immunotherapy. 2016;65:465-476
  79. 79. Seidel UJ, Schlegel P, Lang P. Natural killer cell mediated antibody‐dependent cellular cytotoxicity in tumor immunotherapy with therapeutic antibodies. Frontiers in Immunology. 2013;4:76
  80. 80. Campbell KS, Hasegawa J. Natural killer cell biology: An update and future directions. Journal of Allergy and Clinical Immunology. 2013;132:536-544
  81. 81. Cheng M, Chen Y, Xiao W, Sun R, Tian Z. NK cell‐based immunotherapy for malignant diseases. Cellular & Molecular Immunology. 2013;10:230-252

Written By

Gordana Konjević, Ana Vuletić, Katarina Mirjačić Martinović and Radan Džodić

Submitted: 08 December 2016 Reviewed: 12 May 2017 Published: 13 December 2017