Next Article in Journal
Transcriptome Analysis in Haematococcus pluvialis: Astaxanthin Induction by High Light with Acetate and Fe2+
Next Article in Special Issue
Characterization and Oral Delivery of Proinsulin-Transferrin Fusion Protein Expressed Using ExpressTec
Previous Article in Journal
Modulation of Protein Quality Control and Proteasome to Autophagy Switch in Immortalized Myoblasts from Duchenne Muscular Dystrophy Patients
Previous Article in Special Issue
Transient Expression and Purification of Horseradish Peroxidase C in Nicotiana benthamiana
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Fusion Proteins of NKG2D/NKG2DL in Cancer Immunotherapy

Department of Biological Sciences, Clemson University, 190 Collings Street, Clemson, SC 29634, USA
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2018, 19(1), 177; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms19010177
Submission received: 30 November 2017 / Revised: 22 December 2017 / Accepted: 3 January 2018 / Published: 7 January 2018
(This article belongs to the Special Issue Recombinant Proteins)

Abstract

:
NKG2D (natural killer group 2, member D) is an important activating receptor in natural killer (NK) cells and some T cells. NKG2D ligands (NKG2DLs) are specifically expressed on most tumor cells. The engagement of these ligands on tumor cells to NKG2D on NK cells will induce cell-mediated cytotoxicity and have target cells destroyed. This gives NKG2D/NKG2DLs great potential in cancer therapeutic application. The creation of NKG2D/NKG2DL-based multi-functional fusion proteins is becoming one of the most promising strategies in immunotherapy for cancer. Antibodies, cytokines, and death receptors have been fused with NKG2D or its ligands to produce many powerful fusion proteins, including NKG2D-based chimeric antigen receptors (CARs). In this article, we review the recent developments of the fusion proteins with NKG2D/NKG2DL ligands in cancer immunotherapy.

1. Introduction

Natural killer (NK) cells activating receptor NKG2D (natural killer group 2, member D) and NKG2D ligands (NKG2DLs) play critical roles in cancer immune surveillance, which is one of the most significant findings in the fight against cancer in recent decades [1,2,3,4]. NKG2D, encoded by the gene KLRK1, was first found on the surface of NK cells as an immunosurveillance receptor. It was also found in CD8+ (cluster of differentiation) cytotoxic T cells, some NKT cells, some γδ T cells, and a small subset of CD4+ cytotoxic T cells [4,5,6,7]. NKG2D is a type II transmembrane protein. In humans, two NKG2D proteins recruit four hematopoietic cell signal transducer (DAP10) adaptor proteins to form a hexameric receptor complex. NKG2D associates with DAP10 adaptor proteins by ionic interaction in their transmembrane segments. The two NKG2D ectodomains in the hexameric receptor complex serve as ligand binders. When binding to ligands, this receptor complex triggers the activation of NK cells through the PI3K (phosphatidylinositol-3 kinase) and Grb2-Vav1 (growth factor receptor-bound protein 2, vav guanine nucleotide exchange factor 1) signaling pathways to promote Ca2+ influx, actin cytoskeleton reorganization, and microtubule polarization. As a result, the contents in the granules of NK cells are released to induce the apoptosis of target cells.
Although NKG2D is a quite conserved receptor, NKG2DLs are very diverse. They are structurally similar to major histocompatibility complex (MHC) class I proteins (MIC family), which are highly polymorphic [8]. MICA [4] and MICB [2] are the most extensively studied NKG2DLs in humans. Other NKG2DLs include UL16 binding proteins (ULBPs) [9] in human and mouse, murine UL16-binding protein-like transcript 1 (MULT1) [10], retinoic acid early transcript 1 (Rae1) [11,12] and histocompatibility antigen 60 (H60) [13] in mouse. NKG2DLs are rarely expressed in healthy cells. They are induced when the cell is under the stress of virus infection or malignant transformation, and therefore are called “induced-self” ligands [14]. Cells expressing these ligands will be determinedly detected and eliminated by NK cells. The diversity and the inductivity of NKG2DLs provide NK cells with an effective mechanism in immunosurveillance. However, advanced tumor cells tend to down-regulate or shed off NKG2DLs to escape immune elimination [15].
The combination of the specificity of NKG2DLs on stressed cells, the ability of the NKG2D/NKG2DLs pathway to active NK cells, and the powerful cytotoxicity of NK cells to target cells provides a great application potential of NKG2D/NKG2DLs in cancer immunotherapy. A great number of studies applying NKG2D/NKG2DLs in cancer immunotherapy have been conducted. Firstly, direct up-regulation of NKG2DLs in cancer cells is a straightforward strategy. NKG2DLs have been ectopically expressed in many tumor cell lines, which suppressed the establishment of tumors in vivo [11,16]. Many drugs, such as all-trans retinoic acid (ATRA) [17,18], trichostatin A [17], vitamin D3 [17], and some histone deacetylase inhibitors [19,20] were reported to up-regulate NKG2DLs and could be potential treatments for cancers. Some viral proteins (e.g., E1A in adenovirus), when expressed in tumor cells could up-regulate NKG2DLs and reduce the tumorigenicity of the tumor cells [21,22]. Some chemotherapeutic agents or radiotherapies, as a side-effect, could also induce up-regulation of NKG2DLs by causing DNA damage [23]. Secondly, the expression of NKG2D in NK cells can also be up-regulated as a possible treatment for cancer. The gamma-chain containing cytokines, such as interleukin (IL)-2 [24], IL-7 [25], IL-12 [26], and IL-15 [27], were reported to increase NKG2D expression in human and mouse NK and CD8+ T cells. An IL-15 superagonist mutant (N72D) alone or associated with a dimeric IL-15 receptor α/Fc fusion protein was found to significantly up-regulate NKG2D expression in NK cells and CD8+ T cells [28,29]. Other cytokines, like IL-21 [30], interferon (IFN)-γ [31], and transforming growth factor (TGF)-β [32] were reported to decrease NKG2D expression. Lastly, the association between the polymorphism of NKG2DLs and its genetic predisposition to different cancer types [33] can also be investigated in cancer prediction and therapy.

2. Strategies of Fusion Proteins

The generation of multi-functional fusion proteins consisting of NKG2D/NKG2DLs has become a very active research area in developing effective cancer immunotherapies using various strategies (Figure 1). NKG2D/NKG2DLs fusion proteins involving monoclonal antibodies, cytokines, death receptors, and chimeric antigen receptors are all discussed in this review (Table 1).

2.1. NKG2DL + Antibody

There have been more than a dozen monoclonal antibodies (mAbs) approved by the US Food and Drug Administration (FDA) to treat certain cancers [34]. They can be just naked mAbs that label cancer cells to the immune system, boost immune response against the cancer cells (e.g., antibody-dependent cell-mediated cytotoxicity (ADCC)), or block the antigens that stimulate cancer cell growth. The antibodies can also be conjugated with chemotherapy drugs or radioactive particles to enhance the delivery accuracy to cancer cells. Two different antibodies can also be combined together to bridge cancer cells and immune cells, facilitating immune responses. Fusing a NKG2DL to a tumor-targeting antibody will create a bispecific fusion protein to bridge NK cells and tumor cells, which will redirect and trigger NK cells against the tumor cells.
NKG2DLs usually used in this strategy include H60 [35,36] and Rae1β [35,37] in mouse, and MICA [38,39,40,41] and ULBP2 [38,42,43,44] in human. Since this kind of fusion proteins is always secreted, only the extracellular domain of the NKG2DLs are used. Many antibodies were used against various tumor antigens, such as TNT-3 [35,36] against universal nuclear antigen [45], BB4 [42] against CD138 (syndecan-1), 7D8 [38] against CD20, rG7S [39] against CD24, and antibodies against human epidermal growth factor receptor 2 (HER2) [37], prostate carcinoma membrane antigen (PSMA) [43] and carcinoembryonic antigen (CEA) [44]. As angiogenesis plays an important role in the progression of solid cancer, antibodies against vascular endothelial growth factor receptor (VEGFR) [39,46] were also used in this strategy.
The NKG2DL gene could be fused with the antibody heavy chain gene and co-transfected with the light chain gene to produce a full antibody fused with NKG2DL [35]. The NKG2DL gene, heavy chain gene, and light chain gene could all be linked together by some linker sequences to become a single fusion gene. Or only a fragment of immunoglobulin—such as the single-chain variable fragment (scFv) or crystallizable fragment (Fc)—was used to fuse with the NKG2DL [38,39,42]. The Fc portion in the fusion protein can bind to the Fc receptors (CD16) on NK cells, transmitting activating signals into NK cells. After activating, NK cells can lyse the target cells and secrete cytokines to induce adaptive responses [47]. Germain et al. [40] created a fusion protein with the extracellular domain of MICA and Fc region, and then chemically conjugated it with the Fab fragments of three monoclonal antibodies against tumor-associated antigens: anti-CEA against carcinoembryonic antigen [48], trastuzumab against HER2 [49], and rituximab against CD20 [50].
The NKG2DL-antibody fusion proteins are all soluble. E. coli, yeast, and mammalian cells (e.g., 293T) are used to produce them. They are then purified, concentrated, and used to treat the antigen-positive tumor cells. These fusion proteins showed great binding specificity to the target cells due to the antibody activity. Meanwhile, their NKG2DL portion triggered the cytotoxicity of NK cells against the tumor cells. The Fc portion of the antibody also induced ADCC [38]. Anti-VEGFR-NKG2DL fusion protein demonstrated superior anti-tumor efficacy in a mouse model bearing human breast tumor [39], and induced antiangiogenic activities in vitro [46].

2.2. NKG2DL + Cytokine

Some activating cytokines have essential roles in maintaining cytotoxic lymphocytes’ function and increasing their sensitivity and capability to lyse tumor cells [51], and consequently are used in cancer therapy. For example, IL-2 has been used in the treatment of several malignancies for 30 years [52]. IL-12 is even more efficient than IL-2 in activation of NK cells [53], and also has a potent anti-tumor activity through augmentation of the cytotoxic activity of NK cells, activation of CD8+ T cells, and the inhibition of angiogenesis [54,55]. In order to enhance the functions of these cytokines, they are fused with NKG2DLs which can guide the cytokines to the NKG2D-expressing NK cells or CD8+ T cells, subsequently activating them through both the cytokine/receptor pathway and the NKG2D/NKG2DL pathway.
Tietje et al. [56] in 2014 fused mouse IL-12 and the mouse MULT1E (extracellular domain of MULT1) to create a bi-functional protein. MULT1E binds to NKG2D receptor to activate NK cells, and simultaneously IL-12 engages the IL-12 receptors on NK cells to enhance the cytotoxicity of the killer cells against the tumor cells. The gene of this fusion protein was transfected into tumor cells, and a tumor cell line that stably secreted the fusion protein was generated. When co-culturing this tumor cell line with NK cells, IFN-γ was over-produced and NK cells were highly activated. When intravenously injecting the tumor cell line expressing this fusion protein into a mouse model, the tumor genesis and growth were suppressed compared to injecting the wild type tumor cells. After demonstrating the validity of this strategy in mouse, Tietje et al. [57] created a human version of fusion protein by fusing human IL-12 with the extracellular domain of human MICA. This fusion protein was more effective than either IL-12 or MICA alone in priming NK cells, and was able to increase the proliferation of human peripheral blood mononuclear cells (PBMCs) and their production of IFN-γ.
IL-2 was also applied in creating fusion proteins recently. In order to improve the safety of using IL-2, a mutant form of IL-2 was used which had a poor affinity to the IL-2 receptor. It was fused with a special NKG2DL, orthopoxvirus major histocompatibility complex class I-like protein (OMCP) from cowpox virus [58]. The ligand OMCP had a greater affinity with NKG2D than the other NKG2DLs, and could bind to both human and murine NKG2D [59]. It helped the mutant form of IL-2 to selectively activate the IL-2 signaling pathway only on NKG2D-bearing cells (e.g., NK cells). This fusion protein significantly reduced the broad toxicity and vascular complications associated with the wild type IL-2, and provided a superior NK cell-mediated cancer immunotherapy.

2.3. NKG2DL + Fas

One hallmark of cancer is evading apoptosis (programmed cell death). Cancer cells can resist apoptosis by secreting antagonistic decoy receptors [60], expressing anti-apoptotic molecules (such as BCL2 family members [61]), down-regulating and mutating pro-apoptotic genes (such as BAX, APAF1, or Fas [62,63,64]). Fas (CD95) is a death receptor that transduces apoptotic signals into the cells by forming death-inducing signaling complex (DISC) [65]. Its ligand (FasL) is produced by activated T cells and NK cells [60]. Fas/FasL interaction plays a critical role in the immune progression of cancer [66]. Applying Fas to NKG2DLs fusion protein can graft its apoptosis-inducing function on the fusion protein to make it more powerful.
Kotturi et al. [67] designed and evaluated a fusion protein—MULT1E-FasTI—which fused the extracellular domain of MULT1 and the transmembrane and intracellular domains of Fas. This fusion gene was transfected into a mouse tumor cell line. The expressed protein MULT1E-FasTI was anchored on the cell surface. When treated with recombinant protein NKG2D-Fc, apoptosis of the tumor cells was induced. NK cells were also activated in the co-culture with these tumor cells. Tumor cells expressing MULT1E-FasTI proliferated significantly more slowly compared to tumor cells without the fusion protein in mouse. Kotturi et al. [68] packaged this fusion gene in an adenoviral delivery vector and directly injected the viral particles into the tumors in mice. This treatment induced the apoptosis of the tumor cells in vivo and significantly delayed the tumor growth.

2.4. NKG2D + Antibody

Antibodies can also be fused with NKG2D. These fusion proteins use the NKG2D portion to target tumor cells and use the Fc portion of the antibody to induce ADCC against tumor cells. Salih’s group [69] generated a fusion protein of this kind, consisting of the human NKG2D extracellular domain and the human IgG1 Fc portion. They modified the Fc with S239D/I332E to greatly increase its affinity with the Fc receptors on NK cells. The fusion protein highly enhanced the ADCC, degranulation, and IFN-γ production of NK cells in response to breast cancer cells, despite the fact that the NKG2D portion may block some of the NKG2DLs on the cancer cells and decrease their exposure to NK cells. This fusion protein represents a more attractive potential in the immunotherapy to HER2/neu-low or -negative breast cancer compared to the anti-breast cancer antibody trastuzumab, because it did not target the HER2 antigens but the more widely expressed NKG2DLs. Salih’s group [70] also used similar NKG2D-Fc fusion proteins with modifications in the Fc part to mediate ADCC of NK cells against leukemia. Wu et al. [71] generated two versions (mouse and human) of Dap10-Fc-NKG2D by fusing Dap10 signal peptide, IgG1 Fc (Hinge-CH2-CH3), and NKG2D extracellular domain. They used iron oxide nanoparticles (IONPs) [72] as vehicles to deliver their fusion proteins to multiple NKG2DL-positive tumor cells in vitro.
Sentman’s group [73] used another strategy to create a fusion protein anti-CD3 scFv-NKG2D, in which anti-CD3 single-chain variable fragment was used to target CD3 on T cells. This fusion protein engaged T cells to tumor cells, stimulating T cells to produce IFN-γ and induce cytotoxicity against NKG2DL-positive tumor cells in vitro. A significantly improved anti-tumor activity and survival promotion was observed in vivo.

2.5. NKG2D + Cytokine

Many cytokines, like IL-2, IL-15 and IL-21 were chosen to fuse with NKG2D because of their ability to promote immune cell functions and induce anti-tumor responses. The fusion protein NKG2D-Fc-IL-2 was first designed to use NKG2D to deliver IL-2 to the tumors, in order to achieve a local high-dose in the microenvironment and avoid the severe side effects of systemic administration of IL-2 [74]. Its delivery of IL-2 to the tumor location in vivo was significant, and it enhanced antigen-specific T cell immune response and controlled tumor growth.
IL-15 plays an important role in the development, activation, and persistence of NK cells and CD8+ T cells. Gong’s group [75] generated the fusion protein dsNKG2D-IL-15 consisting of two identical extracellular domains of human NKG2D and human IL-15. This fusion protein targeted the NKG2DL-positive tumor cells through its NKG2D domains and presented IL-15 to NK or CD8+ T cells to promote their activation and cytotoxicity to tumor cells. They found that it significantly retarded the growth of transplanted colon cancers [75] and melanoma [76] in mouse tumor models. Its efficiency in suppressing cancer growth was better than IL-15 cytokine therapy alone. They also delivered their fusion gene into tumor cells by loading the DNA with chitosan nanoparticles [77]. Tumor cells uptaking the nanoparticles with the fusion gene stimulated NK cells and CD8+ T cells in vitro. Intramuscular injection of the nanoparticles with the fusion gene also suppressed tumor growth and prolonged the survival of tumor-bearing mice.
IL-21 is mainly produced by activated T cells and modulates the functions of T, B, and NK cells. IL-21/IL-21R signal pathway regulates both innate and adaptive immune systems, and is significant in inflammatory responses, the development of autoimmune diseases and inflammatory disorders, anti-tumor and antiviral response [78,79]. Tan et al. [80] fused two copies of NKG2D extracellular domain cDNA with IL-21 gene in a vector, and used chitosan nanoparticles to deliver this fusion gene vector into tumor cells. Tumor cells treated with the nanoparticles secreted the fusion protein, which activated NK and CD8+ cells to lyse tumor cell in co-culture. Intramuscularly injecting these NKG2D-IL-21 nanoparticles increased IL-21 levels in serum and promoted the activation of many lymphocytes (including NK cells). After intravenous injection, these NKG2D-IL-21 nanoparticles labeled with fluorescein were detected accumulating and remaining in the tumor tissues of mouse. Tumor growth was sharply retarded, and the life span of tumor-bearing mice was prolonged after injection.

2.6. NKG2D CARs

Chimeric antigen receptors (CARs) are engineered receptors that graft an extracellular antigen-recognition domain onto an intracellular signaling domain in immune effector cells (usually T cells) [81]. The extracellular antigen-recognition domain usually is a single-chain variable fragment derived from monoclonal antibodies. The intracellular signaling domain in the first-generation CARs is typically the CD3ζ signaling chain of T cell receptors (TCRs) [82]. The T cells expressing CARs become antigen-specific cells that are able to recognize specific antigen-expressing cells and subsequently stimulate T cell proliferation, cytokines release, and cytotoxicity to kill the target cells. The second-generation CARs added an additional co-stimulatory signaling domain (e.g., CD28 or 4-1BB) to the first-generation CARs, which significantly enhanced the persistence and proliferation of CAR T cells [83]. The second-generation CD19-specific CAR T cells have demonstrated clinical efficacy in the treatment of B-cell acute lymphoblastic leukemia [84]. The third-generation CARs added two additional co-stimulatory signaling domains to the first-generation CARs, which were even better than the second generation in the anti-tumor efficacy [85]. CARs can basically graft any antigen-recognition capability to T cells, but so far the clinical trials of CARs therapy have been only in patients with blood cancers.
NKG2D as a receptor targeting tumor cells can replace the scFv portion in the typical CARs. In 2005, Sentman’s group [86] first reported this NKG2D CAR, which fused murine CD3ζ chain cytoplasmic region and the full-length murine NKG2D. They used retroviruses to transduct this fusion gene into mouse primary T cells. These T cells responded to tumor cells expressing NKG2DLs in co-culture, produced a large amount of T-helper 1 (Th1) cytokines and proinflammatory chemokines, and induced the lysis of tumor cells bearing NKG2DLs. T cells with this NKG2D CAR inhibited the tumor growth in vivo and showed resistance to new tumor cell challenge. Sentman’s group also created a human version of this fusion gene and applied it on primary human T cells [87]. Similar anti-tumor responses were also observed. The same strategy was also used effectively against not only multiple myeloma, ovarian carcinoma, and lymphoma in vitro and in vivo [88,89,90,91], but also tumor vasculature expressing NKG2DLs [92] and heterogeneous tumors consisting of NKG2DL-expressing and ligand-deficient tumor cells [93]. The group went further and investigated the molecular mechanisms of their NKG2D CARs in cellular cytotoxicity, cytokine production, and the host immune response [88,94,95,96]. In one of their recent studies, they found that high-dose injection of NKG2D CAR T cells resulted in cytokine release syndrome-like responses in mice, which were similar to those seen in some patients given CAR T cells therapy [97].
Lehner et al. [98] and Song et al. [99], respectively, grafted the extracellular domain of NKG2D on two signaling platforms CD28-CD3ζ or 4-1BB-CD3ζ to create new NKG2D CARs. CD28 and 4-1BB (CD137) were added to improve the proliferation and persistence of CAR T cells. CD28 increased IL-2-independent proliferation, and 4-1BB served as a TNF receptor. Upon stimulation with tumor cells, both primary CD4+ T cells and CD8+ T cells expressing these two CARs were activated and demonstrated high cytotoxicity in vitro. Chang et al. [100] added DAP10 to create the fusion protein NKG2D-DAP10-CD3ζ and applied it to NK cells rather than T cells. It markedly activated NK cells, induced greater cytotoxicity against a wide spectrum of tumor subtypes, and produced more cytokine types than mock-transduced NK cells.
VanSeggelen et al. [101] constructed a fusion protein with full-length murine NKG2D and cytoplasmic murine CD3ζ, and engineered it into murine T cells. They found that by adding an adaptor protein (DAP10) to this fusion protein, the protein expression of NKG2D-CAR would be increased 7–10 fold. They also constructed a second-generation CAR that fused the extracellular domain of NKG2D with the CD8 hinge region, the CD28 transmembrane and cytoplasmic domains, and the CD3ζ cytoplasmic domain. All these NKG2D-CARs showed strain-specific differences in surface expression and in vitro functionality. After adoptive transfer of NKG2D-CAR T cells into model mice, they observed dramatic toxicity symptoms—including acute cytokine storm and mortality—in both breast tumor-bearing mice and tumor-free mice. The NKG2D-CAR with DAP10 showed most severe toxicities, which paralleled its highest NKG2D-CAR expression.

3. Discussion

Because the advanced tumor cells can intentionally reduce the expression of NKG2DLs or even shed them off to escape immunosurveillance, the NK cells’ efficiency of detecting the ligands and activating cytotoxicity to kill tumor cells becomes questionable. To overcome this problem, artificially increasing the NKG2DLs on tumor cells or in the tumor microenvironment would be a straightforward thinking. Fusing the ligands with tumor-associated antigen-specific antibodies would help the NKG2DLs to access and label tumor cells. However, this strictly depends on the expression of the antigens on the tumor cells. For instance, high expression of HER2 occurs only in 25–30% of breast cancer patients [69]. Tumor cells’ heterogeneity and evading down-regulation of the antigens also need to be considered. Fusing NKG2DLs to cytokines promotes the delivery of cytokines to NK cells and increases cytokines’ efficiency. These fusion proteins target not tumors but NK cells. The activation from both NKG2DLs and cytokines may cause overreaction of NK cells, and then systemically hyper immune responses, not to mention that the activating cytokines themselves have many side effects. For instance, high dose IL-2 can activate resting cytotoxic lymphocytes, causing wide toxicities [58]. Although the fusion protein MICA-Fas combining NK cell activation and apoptosis induction was powerful against tumor, a significant problem is the specific delivery of the fusion gene to tumor cells. Direct injection of viral vector carrying the fusion gene into the tumor is one approach, but the safety must be considered.
All the fusion proteins with NKG2D have so far used the NKG2D portion to target tumor cells. Their targeting to the tumor cells is very dependent on the expression of NKG2DLs on tumor cells. However, as we mentioned before, there are plenty of problems with NKG2DLs on tumor cells, such as expression down-regulation, shedding, and variance in tumor types and stages [102,103,104,105]. Shedding will release soluble ligands such as MICA. Binding to soluble MICAs can markedly reduce the expression of NKG2D and severely impair the responsiveness of NK cells [106]. Multiple reports showed that NKG2DLs are expressed in various healthy, noncancerous tissues [101]. Some drug treatments and anti-cancer therapies (e.g., chemotherapy) could also induce their expression in noncancerous tissues. Therefore, many factors must be taken into consideration with caution prior to the clinical applications of these NKG2D fusion proteins to avoid unwanted severe toxicities. This is not only a low-efficiency or off-target problem, but also a safety problem. A strategy to avoid the NKG2DLs side effect would be to keep the transmembrane and intercellular domains and replace the extracellular domain of NKG2D with some other proteins to target the specific antigens or proteins on tumor cells (such as antibodies). This fusion protein will utilize the activation signaling pathway of NKG2D in NK cells, but is not limited to targeting NKG2DLs.
Most of the fusion proteins we discussed are soluble, such as NKG2DLs or NKG2D fused with antibodies or cytokines. They are easy to produce and manipulate on treatment. However, in order to express fusion proteins on the tumor cells, accurate delivery of the fusion genes to the tumor cells is one of the most critical challenges. Viral (including lentiviral [107] and adenoviral [108]) delivery systems and nanoparticles [109] are available delivery methods so far. The chitosan nanoparticle is a popularly used vehicle and shows great potential in cancer therapy, because of its capability of tumor targeting and releasing drugs or genes automatically in tumor tissues [80]. However, this is still far from perfect. If not tightly regulated or controlled, the ectopic expression of these NKG2DL fusion proteins may trigger overactivation of the immune system, leading to autoimmune responses or other serious problems.
CAR therapy for cancer is one of the most promising strategies in cancer immunotherapy. It has been restricted to small clinical trials until recently. In 2017, the US FDA approved two CAR T cell therapies: one for children with the treatment of acute lymphoblastic leukemia (ALL) and one for adults with advanced lymphomas [110]. These are the first batch of approved cancer immunotherapies using fusion proteins. The CAR strategy is a personalized therapy to engineer the patient’s own immune cells to specifically recognize and kill the patient’s cancer cells. Similar to other CARs, the primary concerns of NKG2D CARs therapy should be off-tumor toxicity and cytokine storm. It requires a tricky balance between keeping the CAR T or NK cells active enough to kill tumor cells and controlling them to not be overactive and lead to autoimmune toxicity. Endogenous NKG2D, individual variability, dose control, and combination with other anti-cancer therapy are other issues that need to be taken into account as well.

Author Contributions

Hui Ding performed the literature search and wrote the paper; Xi Yang provided some suggestions and critically proof-read the paper; Yanzhang Wei provided ideas about the structure of the paper and edited it.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Groh, V.; Bahram, S.; Bauer, S.; Herman, A.; Beauchamp, M.; Spies, T. Cell stress-regulated human major histocompatibility complex class I gene expressed in gastrointestinal epithelium. Proc. Natl. Acad. Sci. USA 1996, 93, 12445–12450. [Google Scholar] [CrossRef] [PubMed]
  2. Groh, V.; Rhinehart, R.; Secrist, H.; Bauer, S.; Grabstein, K.H.; Spies, T. Broad tumor-associated expression and recognition by tumor-derived gamma delta T cells of MICA and MICB. Proc. Natl. Acad. Sci. USA 1999, 96, 6879–6884. [Google Scholar] [CrossRef] [PubMed]
  3. Long, E.O. Tumor cell recognition by natural killer cells. Semin. Cancer Biol. 2002, 12, 57–61. [Google Scholar] [CrossRef] [PubMed]
  4. Bauer, S.; Groh, V.; Wu, J.; Steinle, A.; Phillips, J.H.; Lanier, L.L.; Spies, T. Activation of NK cells and T cells by NKG2D, a receptor for stress-inducible MICA. Science 1999, 285, 727–729. [Google Scholar] [CrossRef] [PubMed]
  5. Raulet, D.H. Roles of the NKG2D immunoreceptor and its ligands. Nat. Rev. Immunol. 2003, 3, 781–790. [Google Scholar] [CrossRef] [PubMed]
  6. Jamieson, A.M.; Diefenbach, A.; McMahon, C.W.; Xiong, N.; Carlyle, J.R.; Raulet, D.H. The role of the NKG2D immunoreceptor in immune cell activation and natural killing. Immunity 2002, 17, 19–29. [Google Scholar] [CrossRef]
  7. Diefenbach, A.; Jamieson, A.M.; Liu, S.D.; Shastri, N.; Raulet, D.H. Ligands for the murine NKG2D receptor: expression by tumor cells and activation of NK cells and macrophages. Nat. Immunol. 2000, 1, 119–126. [Google Scholar] [CrossRef] [PubMed]
  8. Bahram, S.; Spies, T. The MIC gene family. Res. Immunol. 1996, 147, 328–333. [Google Scholar] [CrossRef]
  9. Cosman, D.; Müllberg, J.; Sutherland, C.L.; Chin, W.; Armitage, R.; Fanslow, W.; Kubin, M.; Chalupny, N.J. ULBPs, Novel MHC Class I–Related Molecules, Bind to CMV Glycoprotein UL16 and Stimulate NK Cytotoxicity through the NKG2D Receptor. Immunity 2001, 14, 123–133. [Google Scholar] [CrossRef]
  10. Carayannopoulos, L.N.; Naidenko, O.V.; Fremont, D.H.; Yokoyama, W.M. Cutting edge: murine UL16-binding protein-like transcript 1: a newly described transcript encoding a high-affinity ligand for murine NKG2D. J. Immunol. 2002, 169, 4079–4083. [Google Scholar] [CrossRef] [PubMed]
  11. Diefenbach, A.; Jensen, E.R.; Jamieson, A.M.; Raulet, D.H. Rae1 and H60 ligands of the NKG2D receptor stimulate tumour immunity. Nature 2001, 413, 165–171. [Google Scholar] [CrossRef] [PubMed]
  12. Cerwenka, A.; Bakker, A.B.; McClanahan, T.; Wagner, J.; Wu, J.; Phillips, J.H.; Lanier, L.L. Retinoic Acid Early Inducible Genes Define a Ligand Family for the Activating NKG2D Receptor in Mice. Immunity 2000, 12, 721–727. [Google Scholar] [CrossRef]
  13. Malarkannan, S.; Shih, P.P.; Eden, P.A.; Horng, T.; Zuberi, A.R.; Christianson, G.; Roopenian, D.; Shastri, N. The molecular and functional characterization of a dominant minor H antigen, H60. J. Immunol. 1998, 161, 3501–3509. [Google Scholar] [PubMed]
  14. Diefenbach, A.; Raulet, D.H. Strategies for target cell recognition by natural killer cells. Immunol. Rev. 2001, 181, 170–184. [Google Scholar] [CrossRef] [PubMed]
  15. Salih, H.R.; Rammensee, H.-G.; Steinle, A. Cutting Edge: Down-Regulation of MICA on Human Tumors by Proteolytic Shedding. J. Immunol. 2002, 169, 4098–4102. [Google Scholar] [CrossRef] [PubMed]
  16. Cerwenka, A.; Baron, J.L.; Lanier, L.L. Ectopic expression of retinoic acid early inducible-1 gene (RAE-1) permits natural killer cell-mediated rejection of a MHC class I-bearing tumor in vivo. Proc. Natl. Acad. Sci. USA 2001, 98, 11521–11526. [Google Scholar] [CrossRef] [PubMed]
  17. Rohner, A.; Langenkamp, U.; Siegler, U.; Kalberer, C.P.; Wodnar-Filipowicz, A. Differentiation-promoting drugs up-regulate NKG2D ligand expression and enhance the susceptibility of acute myeloid leukemia cells to natural killer cell-mediated lysis. Leuk. Res. 2007, 31, 1393–1402. [Google Scholar] [CrossRef] [PubMed]
  18. Poggi, A.; Venturino, C.; Catellani, S.; Clavio, M.; Miglino, M.; Gobbi, M.; Steinle, A.; Ghia, P.; Stella, S.; Caligaris-Cappio, F.; et al. Vδ1 T Lymphocytes from B-CLL Patients Recognize ULBP3 Expressed on Leukemic B Cells and Up-Regulated by Trans-Retinoic Acid. Cancer Res. 2004, 64, 9172–9179. [Google Scholar] [CrossRef] [PubMed]
  19. Armeanu, S.; Bitzer, M.; Lauer, U.M.; Venturelli, S.; Pathil, A.; Krusch, M.; Kaiser, S.; Jobst, J.; Smirnow, I.; Wagner, A.; et al. Natural Killer Cell–Mediated Lysis of Hepatoma Cells via Specific Induction of NKG2D Ligands by the Histone Deacetylase Inhibitor Sodium Valproate. Cancer Res. 2005, 65, 6321–6329. [Google Scholar] [CrossRef] [PubMed]
  20. Schmudde, M.; Braun, A.; Pende, D.; Sonnemann, J.; Klier, U.; Beck, J.F.; Moretta, L.; Bröker, B.M. Histone deacetylase inhibitors sensitize tumour cells for cytotoxic effects of natural killer cells. Cancer Lett. 2008, 272, 110–121. [Google Scholar] [CrossRef] [PubMed]
  21. Korrer, M.J.; Routes, J.M. Adenovirus serotype 5 E1A expressing tumor cells elicit a tumor-specific CD8+ T cell response independent of NKG2D. Results Immunol. 2015, 5, 1–5. [Google Scholar] [CrossRef] [PubMed]
  22. Routes, J.M.; Ryan, S.; Morris, K.; Takaki, R.; Cerwenka, A.; Lanier, L.L. Adenovirus serotype 5 E1A sensitizes tumor cells to NKG2D-dependent NK cell lysis and tumor rejection. J. Exp. Med. 2005, 202, 1477–1482. [Google Scholar] [CrossRef] [PubMed]
  23. Gasser, S.; Orsulic, S.; Brown, E.J.; Raulet, D.H. The DNA damage pathway regulates innate immune system ligands of the NKG2D receptor. Nature. 2005, 436, 1186–1190. [Google Scholar] [CrossRef] [PubMed]
  24. Dhanji, S.; Teh, H.-S. IL-2-activated CD8+CD44high cells express both adaptive and innate immune system receptors and demonstrate specificity for syngeneic tumor cells. J. Immunol. 2003, 171, 3442–3450. [Google Scholar] [CrossRef] [PubMed]
  25. Maasho, K.; Opoku-Anane, J.; Marusina, A.I.; Coligan, J.E.; Borrego, F. NKG2D is a costimulatory receptor for human naive CD8+ T cells. J. Immunol. 2005, 174, 4480–4484. [Google Scholar] [CrossRef] [PubMed]
  26. Zhang, C.; Zhang, J.; Niu, J.; Zhou, Z.; Zhang, J.; Tian, Z. Interleukin-12 improves cytotoxicity of natural killer cells via upregulated expression of NKG2D. Hum. Immunol. 2008, 69, 490–500. [Google Scholar] [CrossRef] [PubMed]
  27. Horng, T.; Bezbradica, J.S.; Medzhitov, R. NKG2D signaling is coupled to the interleukin 15 receptor signaling pathway. Nat. Immunol. 2007, 8, 1345–1352. [Google Scholar] [CrossRef] [PubMed]
  28. Xu, W.; Jones, M.; Liu, B.; Zhu, X.; Johnson, C.B.; Edwards, A.C.; Kong, L.; Jeng, E.K.; Han, K.; Marcus, W.D.; et al. Efficacy and mechanism-of-action of a novel superagonist interleukin-15: interleukin-15 receptor αSu/Fc fusion complex in syngeneic murine models of multiple myeloma. Cancer Res. 2013, 73, 3075–3086. [Google Scholar] [CrossRef] [PubMed]
  29. Zhu, X.; Marcus, W.D.; Xu, W.; Lee, H.-I.; Han, K.; Egan, J.O.; Yovandich, J.L.; Rhode, P.R.; Wong, H.C. Novel Human Interleukin-15 Agonists. J. Immunol. 2009, 183, 3598–3607. [Google Scholar] [CrossRef] [PubMed]
  30. Burgess, S.J.; Marusina, A.I.; Pathmanathan, I.; Borrego, F.; Coligan, J.E. IL-21 down-regulates NKG2D/DAP10 expression on human NK and CD8+ T cells. J. Immunol. 2006, 176, 1490–1497. [Google Scholar] [CrossRef] [PubMed]
  31. Zhang, C.; Zhang, J.; Sun, R.; Feng, J.; Wei, H.; Tian, Z. Opposing effect of IFNγ and IFNα on expression of NKG2 receptors: Negative regulation of IFNγ on NK cells. Int. Immunopharmacol. 2005, 5, 1057–1067. [Google Scholar] [CrossRef] [PubMed]
  32. Song, H.; Hur, D.Y.; Kim, K.-E.; Park, H.; Kim, T.; Kim, C.; Bang, S.; Cho, D.-H. IL-2/IL-18 prevent the down-modulation of NKG2D by TGF-β in NK cells via the c-Jun N-terminal kinase (JNK) pathway. Cell. Immunol. 2006, 242, 39–45. [Google Scholar] [CrossRef] [PubMed]
  33. Chen, D.; Gyllensten, U. MICA polymorphism: biology and importance in cancer. Carcinogenesis 2014, 35, 2633–2642. [Google Scholar] [CrossRef] [PubMed]
  34. Monoclonal Antibodies to Treat Cancer|American Cancer Society. Available online: https://www.cancer.org/treatment/treatments-and-side-effects/treatment-types/immunotherapy/monoclonal-antibodies.html (accessed on 4 October 2017).
  35. Flanagan, M.L.; Hu, P.; Khawli, L.A.; Epstein, A.L. Generation of H60 and Rae1β fusion proteins for the activation of murine NK cells and stimulation of innate antitumor immunity in syngeneic experimental tumor models. Proc. Am. Assoc. Cancer Res. 2005, 46, 6050. [Google Scholar]
  36. Flanagan, M.L.; Khawli, L.A.; Hu, P.; Epstein, A.L. H60/TNT-3 fusion protein activates NK cells in vitro and improves immunotherapeutic outcome in murine syngeneic tumor models. J. Immunother. 2006, 29, 274–283. [Google Scholar] [CrossRef] [PubMed]
  37. Cho, H.M.; Rosenblatt, J.D.; Tolba, K.; Shin, S.J.; Shin, D.S.; Calfa, C.; Zhang, Y.; Shin, S.-U. Delivery of NKG2D ligand using an anti-HER2 antibody-NKG2D ligand fusion protein results in an enhanced innate and adaptive antitumor response. Cancer Res. 2010, 70, 10121–10130. [Google Scholar] [CrossRef] [PubMed]
  38. Kellner, C.; Hallack, D.; Glorius, P.; Staudinger, M.; Mohseni Nodehi, S.; de Weers, M.; van de Winkel, J.G.J.; Parren, P.W.H.I.; Stauch, M.; Valerius, T.; et al. Fusion proteins between ligands for NKG2D and CD20-directed single-chain variable fragments sensitize lymphoma cells for natural killer cell-mediated lysis and enhance antibody-dependent cellular cytotoxicity. Leukemia 2012, 26, 830–834. [Google Scholar] [CrossRef] [PubMed]
  39. Xie, W.; Liu, F.; Wang, Y.; Ren, X.; Wang, T.; Chen, Z.; Tang, M.; Sun, F.; Li, Z.; Wang, M.; et al. VEGFR2 targeted antibody fused with MICA stimulates NKG2D mediated immunosurveillance and exhibits potent anti-tumor activity against breast cancer. Oncotarget 2016, 7, 16445–16461. [Google Scholar] [CrossRef] [PubMed]
  40. Germain, C.; Larbouret, C.; Cesson, V.; Donda, A.; Held, W.; Mach, J.P.; Pèlegrin, A.; Robert, B. MHC class I-related chain a conjugated to antitumor antibodies can sensitize tumor cells to specific lysis by natural killer cells. Clin. Cancer Res. 2005, 11, 7516–7522. [Google Scholar] [CrossRef] [PubMed]
  41. Wang, T.; Sun, F.; Xie, W.; Tang, M.; He, H.; Jia, X.; Tian, X.; Wang, M.; Zhang, J. A bispecific protein rG7S-MICA recruits natural killer cells and enhances NKG2D-mediated immunosurveillance against hepatocellular carcinoma. Cancer Lett. 2016, 372, 166–178. [Google Scholar] [CrossRef] [PubMed]
  42. Von Strandmann, E.P.; Hansen, H.P.; Reiners, K.S.; Schnell, R.; Borchmann, P.; Merkert, S.; Simhadri, V.R.; Draube, A.; Reiser, M.; Purr, I.; et al. A novel bispecific protein (ULBP2-BB4) targeting the NKG2D receptor on natural killer (NK) cells and CD138 activates NK cells and has potent antitumor activity against human multiple myeloma in vitro and in vivo. Blood 2006, 107, 1955–1962. [Google Scholar] [CrossRef] [PubMed]
  43. Jachimowicz, R.D.; Fracasso, G.; Yazaki, P.J.; Power, B.E.; Borchmann, P.; Engert, A.; Hansen, H.P.; Reiners, K.S.; Marie, M.; von Strandmann, E.P.; et al. Induction of in vitro and in vivo NK cell cytotoxicity using high-avidity immunoligands targeting prostate-specific membrane antigen in prostate carcinoma. Mol. Cancer Ther. 2011, 10, 1036–1045. [Google Scholar] [CrossRef] [PubMed]
  44. Rothe, A.; Jachimowicz, R.D.; Borchmann, S.; Madlener, M.; Keßler, J.; Reiners, K.S.; Sauer, M.; Hansen, H.P.; Ullrich, R.T.; Chatterjee, S.; et al. The bispecific immunoligand ULBP2-aCEA redirects natural killer cells to tumor cells and reveals potent anti-tumor activity against colon carcinoma. Int. J. Cancer 2014, 134, 2829–2840. [Google Scholar] [CrossRef] [PubMed]
  45. Hornick, J.L.; Sharifi, J.; Khawli, L.A.; Hu, P.; Biela, B.H.; Mizokami, M.M.; Yun, A.; Taylor, C.R.; Epstein, A.L. A New Chemically Modified Chimeric TNT-3 Monoclonal Antibody Directed Against DNA for the Radioimmunotherapy of Solid Tumors. Cancer Biother. Radiopharm. 1998, 13, 255–268. [Google Scholar] [CrossRef] [PubMed]
  46. Acheampong, D.O.; Tang, M.; Wang, Y.; Zhao, X.; Xie, W.; Chen, Z.; Tian, W.; Wang, M.; Zhang, J. A Novel Fusion Antibody Exhibits Antiangiogenic Activity and Stimulates NK Cell-mediated Immune Surveillance Through Fused NKG2D Ligand. J. Immunother. 2017, 40, 94–103. [Google Scholar] [CrossRef] [PubMed]
  47. Wang, W.; Erbe, A.K.; Hank, J.A.; Morris, Z.S.; Sondel, P.M. NK Cell-Mediated Antibody-Dependent Cellular Cytotoxicity in Cancer Immunotherapy. Front. Immunol. 2015, 6, 368. [Google Scholar] [CrossRef] [PubMed]
  48. Buchegger, F.; Haskell, C.M.; Schreyer, M.; Scazziga, B.R.; Randin, S.; Carrel, S.; Mach, J.P. Radiolabeled fragments of monoclonal antibodies against carcinoembryonic antigen for localization of human colon carcinoma grafted into nude mice. J. Exp. Med. 1983, 158, 413–427. [Google Scholar] [CrossRef] [PubMed]
  49. Cobleigh, M.A.; Vogel, C.L.; Tripathy, D.; Robert, N.J.; Scholl, S.; Fehrenbacher, L.; Wolter, J.M.; Paton, V.; Shak, S.; Lieberman, G.; et al. Multinational study of the efficacy and safety of humanized anti-HER2 monoclonal antibody in women who have HER2-overexpressing metastatic breast cancer that has progressed after chemotherapy for metastatic disease. J. Clin. Oncol. 1999, 17, 2639–2648. [Google Scholar] [CrossRef] [PubMed]
  50. McLaughlin, P.; Grillo-López, A.J.; Link, B.K.; Levy, R.; Czuczman, M.S.; Williams, M.E.; Heyman, M.R.; Bence-Bruckler, I.; White, C.A.; Cabanillas, F.; et al. Rituximab chimeric anti-CD20 monoclonal antibody therapy for relapsed indolent lymphoma: half of patients respond to a four-dose treatment program. J. Clin. Oncol. 1998, 16, 2825–2833. [Google Scholar] [CrossRef] [PubMed]
  51. Lehmann, C.; Zeis, M.; Uharek, L. Activation of natural killer cells with interleukin 2 (IL-2) and IL-12 increases perforin binding and subsequent lysis of tumour cells. Br. J. Haematol. 2001, 114, 660–665. [Google Scholar] [CrossRef] [PubMed]
  52. Rosenberg, S.A. IL-2: The first effective immunotherapy for human cancer. J. Immunol. 2014, 192, 5451–5458. [Google Scholar] [CrossRef] [PubMed]
  53. Robertson, M.J.; Soiffer, R.J.; Wolf, S.F.; Manley, T.J.; Donahue, C.; Young, D.; Herrmann, S.H.; Ritz, J. Response of human natural killer (NK) cells to NK cell stimulatory factor (NKSF): cytolytic activity and proliferation of NK cells are differentially regulated by NKSF. J. Exp. Med. 1992, 175, 779–788. [Google Scholar] [CrossRef] [PubMed]
  54. Brunda, M.J.; Luistro, L.; Warrier, R.R.; Wright, R.B.; Hubbard, B.R.; Murphy, M.; Wolf, S.F.; Gately, M.K. Antitumor and antimetastatic activity of interleukin 12 against murine tumors. J. Exp. Med. 1993, 178, 1223–1230. [Google Scholar] [CrossRef] [PubMed]
  55. Voest, E.E.; Kenyon, B.M.; O’Reilly, M.S.; Truitt, G.; D’Amato, R.J.; Folkman, J. Inhibition of angiogenesis in vivo by interleukin 12. J. Natl. Cancer Inst. 1995, 87, 581–586. [Google Scholar] [CrossRef] [PubMed]
  56. Tietje, A.; Li, J.; Yu, X.; Wei, Y. MULT1E/mIL-12: A novel bifunctional protein for natural killer cell activation. Gene Ther. 2014, 21, 468–475. [Google Scholar] [CrossRef] [PubMed]
  57. Tietje, A.; Yang, X.; Yu, X.; Wei, Y. MICA/IL-12: A novel bifunctional protein for killer cell activation. Oncol. Rep. 2017, 37, 1889–1895. [Google Scholar] [CrossRef] [PubMed]
  58. Ghasemi, R.; Lazear, E.; Wang, X.; Arefanian, S.; Zheleznyak, A.; Carreno, B.M.; Higashikubo, R.; Gelman, A.E.; Kreisel, D.; Fremont, D.H.; et al. Selective targeting of IL-2 to NKG2D bearing cells for improved immunotherapy. Nat. Commun. 2016, 7, 12878. [Google Scholar] [CrossRef] [PubMed]
  59. Campbell, J.A.; Trossman, D.S.; Yokoyama, W.M.; Carayannopoulos, L.N. Zoonotic orthopoxviruses encode a high-affinity antagonist of NKG2D. J. Exp. Med. 2007, 204, 1311. [Google Scholar] [CrossRef] [PubMed]
  60. Ashkenazi, A.; Pitti, R.M.; Marsters, S.A.; Lawrence, D.A.; Roy, M.; Kischkel, F.C.; Dowd, P.; Huang, A.; Donahue, C.J.; Sherwood, S.W.; et al. Genomic amplification of a decoy receptor for Fas ligand in lung and colon cancer. Nature 1998, 396, 699–703. [Google Scholar] [CrossRef]
  61. Boise, L.H.; González-García, M.; Postema, C.E.; Ding, L.; Lindsten, T.; Turka, L.A.; Mao, X.; Nuñez, G.; Thompson, C.B. bcl-x, a bcl-2-related gene that functions as a dominant regulator of apoptotic cell death. Cell 1993, 74, 597–608. [Google Scholar] [CrossRef]
  62. Ionov, Y.; Yamamoto, H.; Krajewski, S.; Reed, J.C.; Perucho, M. Mutational inactivation of the proapoptotic gene BAX confers selective advantage during tumor clonal evolution. Proc. Natl. Acad. Sci. USA 2000, 97, 10872–10877. [Google Scholar] [CrossRef] [PubMed]
  63. Soengas, M.S.; Capodieci, P.; Polsky, D.; Mora, J.; Esteller, M.; Opitz-Araya, X.; McCombie, R.; Herman, J.G.; Gerald, W.L.; Lazebnik, Y.A.; et al. Inactivation of the apoptosis effector Apaf-1 in malignant melanoma. Nature 2001, 409, 207–211. [Google Scholar] [CrossRef] [PubMed]
  64. Strand, S.; Hofmann, W.J.; Hug, H.; Müller, M.; Otto, G.; Strand, D.; Mariani, S.M.; Stremmel, W.; Krammer, P.H.; Galle, P.R. Lymphocyte apoptosis induced by CD95 (APO-1/Fas) ligand-expressing tumor cells—A mechanism of immune evasion? Nat. Med. 1996, 2, 1361–1366. [Google Scholar] [CrossRef] [PubMed]
  65. Wang, L.; Yang, J.K.; Kabaleeswaran, V.; Rice, A.J.; Cruz, A.C.; Park, A.Y.; Yin, Q.; Damko, E.; Jang, S.B.; Raunser, S.; et al. The Fas–FADD death domain complex structure reveals the basis of DISC assembly and disease mutations. Nat. Struct. Mol. Biol. 2010, 17, 1324–1329. [Google Scholar] [CrossRef] [PubMed]
  66. Strasser, A.; Jost, P.J.; Nagata, S. The Many Roles of FAS Receptor Signaling in the Immune System. Immunity 2009, 30, 180–192. [Google Scholar] [CrossRef] [PubMed]
  67. Kotturi, H.S.R.; Li, J.; Branham-O’Connor, M.; Stickel, S.L.; Yu, X.; Wagner, T.E.; Wei, Y. Tumor cells expressing a fusion protein of MULT1 and Fas are rejected in vivo by apoptosis and NK cell activation. Gene Ther. 2008, 15, 1302–1310. [Google Scholar] [CrossRef] [PubMed]
  68. Kotturi, H.S.R.; Li, J.; Branham-O’Connor, M.; Yu, X.; Wagner, T.E.; Wei, Y. In vitro and in vivo delivery of novel anticancer fusion protein MULT1E/FasTI via adenoviral vectors. Cancer Gene Ther. 2010, 17, 164–170. [Google Scholar] [CrossRef] [PubMed]
  69. Raab, S.; Steinbacher, J.; Schmiedel, B.J.; Kousis, P.C.; Steinle, A.; Jung, G.; Grosse-Hovest, L.; Salih, H.R. Fc-optimized NKG2D-Fc constructs induce NK cell antibody-dependent cellular cytotoxicity against breast cancer cells independently of HER2/neu expression status. J. Immunol. 2014, 193, 4261–4272. [Google Scholar] [CrossRef] [PubMed]
  70. Steinbacher, J.; Baltz-Ghahremanpour, K.; Schmiedel, B.J.; Steinle, A.; Jung, G.; Kübler, A.; André, M.C.; Grosse-Hovest, L.; Salih, H.R. An Fc-optimized NKG2D-immunoglobulin G fusion protein for induction of natural killer cell reactivity against leukemia. Int. J. Cancer 2015, 136, 1073–1084. [Google Scholar] [CrossRef] [PubMed]
  71. Wu, M.-R.; Cook, W.J.; Zhang, T.; Sentman, C.L. Targeting multiple types of tumors using NKG2D-coated iron oxide nanoparticles. Nanotechnology 2014, 25, 475101. [Google Scholar] [CrossRef] [PubMed]
  72. Yigit, M.V.; Moore, A.; Medarova, Z. Magnetic Nanoparticles for Cancer Diagnosis and Therapy. Pharm. Res. 2012, 29, 1180–1188. [Google Scholar] [CrossRef] [PubMed]
  73. Zhang, T.; Sentman, C.L. Cancer immunotherapy using a bispecific NK receptor fusion protein that engages both T cells and tumor cells. Cancer Res. 2011, 71, 2066–2076. [Google Scholar] [CrossRef] [PubMed]
  74. Kang, T.H.; Mao, C.-P.; He, L.; Tsai, Y.-C.; Liu, K.; La, V.; Wu, T.-C.; Hung, C.-F. Tumor-Targeted Delivery of IL-2 by NKG2D Leads to Accumulation of Antigen-Specific CD8+ T Cells in the Tumor Loci and Enhanced Anti-Tumor Effects. PLoS ONE 2012, 7, e35141. [Google Scholar] [CrossRef] [PubMed]
  75. Xia, Y.; Chen, B.; Shao, X.; Xiao, W.; Qian, L.; Ding, Y.; Ji, M.; Gong, W. Treatment With a Fusion Protein of the Extracellular Domains of NKG2D to IL-15 Retards Colon Cancer Growth in Mice. J. Immunother. 2014, 37, 257–266. [Google Scholar] [CrossRef] [PubMed]
  76. Chen, Y.; Chen, B.; Yang, T.; Xiao, W.; Qian, L.; Ding, Y.; Ji, M.; Ge, X.; Gong, W. Human fused NKG2D–IL-15 protein controls xenografted human gastric cancer through the recruitment and activation of NK cells. Cell. Mol. Immunol. 2017, 14, 293–307. [Google Scholar] [CrossRef] [PubMed]
  77. Yan, C.; Jie, L.; Yongqi, W.; Weiming, X.; Juqun, X.; Yanbing, D.; Li, Q.; Xingyuan, P.; Mingchun, J.; Weijuan, G. Delivery of human NKG2D-IL-15 fusion gene by chitosan nanoparticles to enhance antitumor immunity. Biochem. Biophys. Res. Commun. 2015. [Google Scholar] [CrossRef] [PubMed]
  78. Leonard, W.J.; Wan, C. IL-21 Signaling in Immunity. F1000Research 2016, 5, 224. [Google Scholar] [CrossRef] [PubMed]
  79. Spolski, R.; Leonard, W.J. Interleukin-21: A double-edged sword with therapeutic potential. Nat. Rev. Drug Discov. 2014, 13, 379–395. [Google Scholar] [CrossRef] [PubMed]
  80. Tan, L.; Han, S.; Ding, S.; Xiao, W.; Ding, Y.; Qian, L.; Wang, C.; Gong, W. Chitosan nanoparticle-based delivery of fused NKG2D-IL-21 gene suppresses colon cancer growth in mice. Int. J. Nanomed. 2017, 12, 3095–3107. [Google Scholar] [CrossRef] [PubMed]
  81. Jackson, H.J.; Rafiq, S.; Brentjens, R.J. Driving CAR T-cells forward. Nat. Rev. Clin. Oncol. 2016, 13, 370–383. [Google Scholar] [CrossRef] [PubMed]
  82. Eshhar, Z.; Waks, T.; Gross, G.; Schindler, D.G. Specific activation and targeting of cytotoxic lymphocytes through chimeric single chains consisting of antibody-binding domains and the gamma or zeta subunits of the immunoglobulin and T-cell receptors. Proc. Natl. Acad. Sci. USA 1993, 90, 720–724. [Google Scholar] [CrossRef] [PubMed]
  83. Savoldo, B.; Ramos, C.A.; Liu, E.; Mims, M.P.; Keating, M.J.; Carrum, G.; Kamble, R.T.; Bollard, C.M.; Gee, A.P.; Mei, Z.; et al. CD28 costimulation improves expansion and persistence of chimeric antigen receptor–modified T cells in lymphoma patients. J. Clin. Investig. 2011, 121, 1822–1826. [Google Scholar] [CrossRef] [PubMed]
  84. Lee, D.W.; Kochenderfer, J.N.; Stetler-Stevenson, M.; Cui, Y.K.; Delbrook, C.; Feldman, S.A.; Fry, T.J.; Orentas, R.; Sabatino, M.; Shah, N.N.; et al. T cells expressing CD19 chimeric antigen receptors for acute lymphoblastic leukaemia in children and young adults: A phase 1 dose-escalation trial. Lancet (Lond., Engl.) 2015, 385, 517–528. [Google Scholar] [CrossRef]
  85. Carpenito, C.; Milone, M.C.; Hassan, R.; Simonet, J.C.; Lakhal, M.; Suhoski, M.M.; Varela-Rohena, A.; Haines, K.M.; Heitjan, D.F.; Albelda, S.M.; et al. Control of large, established tumor xenografts with genetically retargeted human T cells containing CD28 and CD137 domains. Proc. Natl. Acad. Sci. USA 2009, 106, 3360–3365. [Google Scholar] [CrossRef] [PubMed]
  86. Zhang, T.; Lemoi, B.A.; Sentman, C.L. Chimeric NK-receptor-bearing T cells mediate antitumor immunotherapy. Blood 2005, 106, 1544–1551. [Google Scholar] [CrossRef] [PubMed]
  87. Zhang, T.; Barber, A.; Sentman, C.L. Generation of antitumor responses by genetic modification of primary human T cells with a chimeric NKG2D receptor. Cancer Res. 2006, 66, 5927–5933. [Google Scholar] [CrossRef] [PubMed]
  88. Zhang, T.; Barber, A.; Sentman, C.L. Chimeric NKG2D Modified T Cells Inhibit Systemic T-Cell Lymphoma Growth in a Manner Involving Multiple Cytokines and Cytotoxic Pathways. Cancer Res. 2007, 67, 11029–11036. [Google Scholar] [CrossRef] [PubMed]
  89. Barber, A.; Zhang, T.; Megli, C.J.; Wu, J.; Meehan, K.R.; Sentman, C.L. Chimeric NKG2D receptor–expressing T cells as an immunotherapy for multiple myeloma. Exp. Hematol. 2008, 36, 1318–1328. [Google Scholar] [CrossRef] [PubMed]
  90. Barber, A.; Zhang, T.; DeMars, L.R.; Conejo-Garcia, J.; Roby, K.F.; Sentman, C.L. Chimeric NKG2D Receptor-Bearing T Cells as Immunotherapy for Ovarian Cancer. Cancer Res. 2007, 67, 5003–5008. [Google Scholar] [CrossRef] [PubMed]
  91. Barber, A.; Meehan, K.R.; Sentman, C.L. Treatment of multiple myeloma with adoptively transferred chimeric NKG2D receptor-expressing T cells. Gene Ther. 2011, 18, 509–516. [Google Scholar] [CrossRef] [PubMed]
  92. Zhang, T.; Sentman, C.L. Mouse Tumor Vasculature Expresses NKG2D Ligands and Can Be Targeted by Chimeric NKG2D-Modified T Cells. J. Immunol. 2013, 190, 2455–2463. [Google Scholar] [CrossRef] [PubMed]
  93. Spear, P.; Barber, A.; Rynda-Apple, A.; Sentman, C.L. NKG2D CAR T-cell therapy inhibits the growth of NKG2D ligand heterogeneous tumors. Immunol. Cell Biol. 2013, 91, 435–440. [Google Scholar] [CrossRef] [PubMed]
  94. Barber, A.; Rynda, A.; Sentman, C.L. Chimeric NKG2D Expressing T Cells Eliminate Immunosuppression and Activate Immunity within the Ovarian Tumor Microenvironment. J. Immunol. 2009, 183, 6939–6947. [Google Scholar] [CrossRef] [PubMed]
  95. Barber, A.; Sentman, C.L. Chimeric NKG2D T cells require both T cell- and host-derived cytokine secretion and perforin expression to increase tumor antigen presentation and systemic immunity. J. Immunol. 2009, 183, 2365–2372. [Google Scholar] [CrossRef] [PubMed]
  96. Spear, P.; Barber, A.; Rynda-Apple, A.; Sentman, C.L. Chimeric Antigen Receptor T Cells Shape Myeloid Cell Function within the Tumor Microenvironment through IFN- and GM-CSF. J. Immunol. 2012, 188, 6389–6398. [Google Scholar] [CrossRef] [PubMed]
  97. Sentman, M.-L.; Murad, J.M.; Cook, W.J.; Wu, M.-R.; Reder, J.; Baumeister, S.H.; Dranoff, G.; Fanger, M.W.; Sentman, C.L. Mechanisms of Acute Toxicity in NKG2D Chimeric Antigen Receptor T Cell–Treated Mice. J. Immunol. 2016, 197, 4674–4685. [Google Scholar] [CrossRef] [PubMed]
  98. Lehner, M.; Götz, G.; Proff, J.; Schaft, N.; Dörrie, J.; Full, F.; Ensser, A.; Muller, Y.A.; Cerwenka, A.; Abken, H.; et al. Redirecting T Cells to Ewing’s Sarcoma Family of Tumors by a Chimeric NKG2D Receptor Expressed by Lentiviral Transduction or mRNA Transfection. PLoS ONE 2012, 7, e31210. [Google Scholar] [CrossRef] [PubMed]
  99. Song, D.-G.; Ye, Q.; Santoro, S.; Fang, C.; Best, A.; Powell, D.J. Chimeric NKG2D CAR-Expressing T Cell-Mediated Attack of Human Ovarian Cancer Is Enhanced by Histone Deacetylase Inhibition. Hum. Gene Ther. 2013, 24, 295–305. [Google Scholar] [CrossRef] [PubMed]
  100. Chang, Y.H.; Connolly, J.; Shimasaki, N.; Mimura, K.; Kono, K.; Campana, D. A chimeric receptor with NKG2D specificity enhances natural killer cell activation and killing of tumor cells. Cancer Res. 2013, 73, 1777–1786. [Google Scholar] [CrossRef] [PubMed]
  101. VanSeggelen, H.; Hammill, J.A.; Dvorkin-Gheva, A.; Tantalo, D.G.M.; Kwiecien, J.M.; Denisova, G.F.; Rabinovich, B.; Wan, Y.; Bramson, J.L. T Cells Engineered With Chimeric Antigen Receptors Targeting NKG2D Ligands Display Lethal Toxicity in Mice. Mol. Ther. 2015, 23, 1600–1610. [Google Scholar] [CrossRef] [PubMed]
  102. De Kruijf, E.M.; Sajet, A.; van Nes, J.G.; Putter, H.; Smit, V.T.; Eagle, R.A.; Jafferji, I.; Trowsdale, J.; Liefers, G.J.; van de Velde, C.J.; et al. NKG2D ligand tumor expression and association with clinical outcome in early breast cancer patients: an observational study. BMC Cancer 2012, 12, 24. [Google Scholar] [CrossRef] [PubMed]
  103. McGilvray, R.W.; Eagle, R.A.; Rolland, P.; Jafferji, I.; Trowsdale, J.; Durrant, L.G. ULBP2 and RAET1E NKG2D ligands are independent predictors of poor prognosis in ovarian cancer patients. Int. J. Cancer 2010, 127, 1412–1420. [Google Scholar] [CrossRef] [PubMed]
  104. Madjd, Z.; Spendlove, I.; Moss, R.; Bevin, S.; Pinder, S.E.; Watson, N.F.S.; Ellis, I.; Durrant, L.G. Upregulation of MICA on high-grade invasive operable breast carcinoma. Cancer Immun. 2007, 7, 17. [Google Scholar] [PubMed]
  105. Watson, N.F.S.; Spendlove, I.; Madjd, Z.; McGilvray, R.; Green, A.R.; Ellis, I.O.; Scholefield, J.H.; Durrant, L.G. Expression of the stress-related MHC class I chain-related protein MICA is an indicator of good prognosis in colorectal cancer patients. Int. J. Cancer 2006, 118, 1445–1452. [Google Scholar] [CrossRef] [PubMed]
  106. Groh, V.; Wu, J.; Yee, C.; Spies, T. Tumour-derived soluble MIC ligands impair expression of NKG2D and T-cell activation. Nature 2002, 419, 734–738. [Google Scholar] [CrossRef] [PubMed]
  107. Oldham, R.A.; Berinstein, E.M.; Medin, J.A. Lentiviral vectors in cancer immunotherapy. Immunotherapy 2015, 7, 271–284. [Google Scholar] [CrossRef] [PubMed]
  108. Kasala, D.; Choi, J.-W.; Kim, S.W.; Yun, C.-O. Utilizing adenovirus vectors for gene delivery in cancer. Expert Opin. Drug Deliv. 2014, 11, 379–392. [Google Scholar] [CrossRef] [PubMed]
  109. Jin, L.; Zeng, X.; Liu, M.; Deng, Y.; He, N. Current progress in gene delivery technology based on chemical methods and nano-carriers. Theranostics 2014, 4, 240–255. [Google Scholar] [CrossRef] [PubMed]
  110. CAR T Cells: Engineering Patients’ Immune Cells to Treat Their Cancers. Available online: https://www.cancer.gov/about-cancer/treatment/research/car-t-cells (accessed on 20 December 2017).
Figure 1. Schematic diagram of fusion proteins of NKG2D (natural killer group 2, member D)/NKG2D ligands (NKG2DLs) in cancer immunotherapy. Fc, fragment crystallizable region of an antibody; ADCC, antibody-dependent cell-mediated cytotoxicity; CD3, cluster of differentiation 3; DAP10, hematopoietic cell signal transducer; PI3K, phosphatidylinositol-3 kinase; Grb2, growth factor receptor-bound protein 2; Vav1, vav guanine nucleotide exchange factor 1; CAR, chimeric antigen receptors.
Figure 1. Schematic diagram of fusion proteins of NKG2D (natural killer group 2, member D)/NKG2D ligands (NKG2DLs) in cancer immunotherapy. Fc, fragment crystallizable region of an antibody; ADCC, antibody-dependent cell-mediated cytotoxicity; CD3, cluster of differentiation 3; DAP10, hematopoietic cell signal transducer; PI3K, phosphatidylinositol-3 kinase; Grb2, growth factor receptor-bound protein 2; Vav1, vav guanine nucleotide exchange factor 1; CAR, chimeric antigen receptors.
Ijms 19 00177 g001
Table 1. Fusion proteins of NKG2D/NKG2DLs in cancer immunotherapy.
Table 1. Fusion proteins of NKG2D/NKG2DLs in cancer immunotherapy.
StrategyFusion ProteinTarget/PathwayMalignancyReferences
NKG2DL + AntibodyH60-TNT3UNAYAC-1[35,36]
Rae1β-TNT3UNACT-26, LLC[36]
MICAE-Fc (conjugate with Fab)CEA/HER2/CD20CC, BC, OC, Raji[40]
ULBP2-BB4CD138MM[42]
anti-HER2 IgG3-Rae1βHER2MC[37]
ULBP2-anti-PSMA scFvPSMAPC[43]
MICAE-7D8CD20CLL, MZL, MCL[38]
ULBP2E-7D8CD20CLL, MZL, MCL[38]
ULBP2-anti-CEACEACC[44]
mAb04-MICAEVEGFRBC[39]
anti-VEGFR2 scFv-MICAEVEGFRHUVEC, K562, MDA-MB-435[46]
rG7S-MICAECD24HCC[41]
NKG2DL + CytokineMULT1E-IL-12IL-12RTC-1[56]
MICAE-IL-12IL-12RA549[57]
OMCP-mutIL-2IL-2RLLC, YAC-1[58]
NKG2DL + FasMULT1E-FasTIFasTC-1[67,68]
NKG2D + AntibodyNKG2DE-FcADCCBC[69,70]
Dap10-Fc-NKG2DERMA/RG, P815[71]
anti-CD3 scFv-NKG2DECD3RMA/RG, P815, ID8, MC-38[73]
NKG2D + CytokineNKG2DE-Fc-IL-2IL-2RTC-1[74]
dsNKG2DE-IL-15IL-15RCC[75]
dsNKG2DE-IL-21IL-21RCT-26[80]
NKG2D CARsNKG2D-CD3ζITCRMM, OC, Lymphoma, BC, etc.[86,87,88,89,90,91,92,93,94,95,96,97,101]
NKG2DE-CD28-CD3ζITCRESFT, 4T1.2[98,101]
NKG2DE-4-1BB-CD3ζITCR/CD137OC[99]
DAP10-NKG2DE-CD3ζITCROsteosarcoma, 4T1.2[100,101]
Subscript formatted E, T and I indicate extracellular domain, transmembrane domain, and intercellular domain, respectively. The NKG2D/NKG2DLs pathway was not included in the Target/Pathway column. UNA, universal nuclear antigen; LLC, Lewis lung carcinoma; CEA, carcinoembryonic antigen; HER2, human epidermal growth factor receptor 2; CC, colon carcinoma; BC, breast carcinoma; OC, ovarian carcinoma; MM, multiple myeloma; MC, mammary carcinoma; PSMA, prostate carcinoma membrane; PC, prostate carcinoma; CLL, chronic lymphocytic leukemia; MZL, marginal zone lymphoma; MCL, mantle cell lymphoma; VEGFR, antigen vascular endothelial growth factor receptor; HUVEC, human umbilical vein endothelial cell; HCC, hepatocellular carcinoma; TCR, T cell receptors; ESFT, Ewing’s sarcoma family of tumors.

Share and Cite

MDPI and ACS Style

Ding, H.; Yang, X.; Wei, Y. Fusion Proteins of NKG2D/NKG2DL in Cancer Immunotherapy. Int. J. Mol. Sci. 2018, 19, 177. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms19010177

AMA Style

Ding H, Yang X, Wei Y. Fusion Proteins of NKG2D/NKG2DL in Cancer Immunotherapy. International Journal of Molecular Sciences. 2018; 19(1):177. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms19010177

Chicago/Turabian Style

Ding, Hui, Xi Yang, and Yanzhang Wei. 2018. "Fusion Proteins of NKG2D/NKG2DL in Cancer Immunotherapy" International Journal of Molecular Sciences 19, no. 1: 177. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms19010177

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop