Next Article in Journal
Reduced T-cell Numbers and Elevated Levels of Immunomodulatory Cytokines in Metastatic Prostate Cancer Patients De Novo Resistant to Abiraterone and/or Enzalutamide Therapy
Next Article in Special Issue
Fucoxanthin-Rich Brown Algae Extract Improves Male Reproductive Function on Streptozotocin-Nicotinamide-Induced Diabetic Rat Model
Previous Article in Journal
Recent Developments in Metal-Based Drugs and Chelating Agents for Neurodegenerative Diseases Treatments
Previous Article in Special Issue
IL-36 Cytokines: Regulators of Inflammatory Responses and Their Emerging Role in Immunology of Reproduction
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

European Patent in Immunoncology: From Immunological Principles of Implantation to Cancer Treatment

by
Franziska M. Würfel
1,*,
Christoph Winterhalter
2,
Peter Trenkwalder
2,
Ralph M. Wirtz
1 and
Wolfgang Würfel
2
1
STRATIFYER Molecular Pathology GmbH, D-50935 Cologne, Werthmannstrasse 1c, 50935 Cologne, Germany
2
INTELLEXON GmbH, Keltenstrasse 27, 82343 Pöcking, Germany
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2019, 20(8), 1830; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms20081830
Submission received: 31 January 2019 / Revised: 9 April 2019 / Accepted: 9 April 2019 / Published: 12 April 2019
(This article belongs to the Special Issue Reproductive Immunology: Cellular and Molecular Biology)

Abstract

:
The granted European patent EP 2 561 890 describes a procedure for an immunological treatment of cancer. It is based on the principles of the HLA-supported communication of implantation and pregnancy. These principles ensure that the embryo is not rejected by the mother. In pregnancy, the placenta, more specifically the trophoblast, creates an “interface” between the embryo/fetus and the maternal immune system. Trophoblasts do not express the “original” HLA identification of the embryo/fetus (HLA-A to -DQ), but instead show the non-classical HLA groups E, F, and G. During interaction with specific receptors of NK cells (e.g., killer-immunoglobulin-like receptors (KIR)) and lymphocytes (lymphocyte-immunoglobulin-like receptors (LIL-R)), the non-classical HLA groups inhibit these immunocompetent cells outside pregnancy. However, tumors are known to be able to express these non-classical HLA groups and thus make use of an immuno-communication as in pregnancies. If this occurs, the prognosis usually worsens. This patent describes, in a first step, the profiling of the non-classical HLA groups in primary tumor tissue as well as metastases and recurrent tumors. The second step comprises tailored antibody therapies, which is the subject of this patent. In this review, we analyze the underlying mechanisms and describe the currently known differences between HLA-supported communication of implantation and that of tumors.

1. Introduction

The close similarities between embryos, i.e., trophoblast invasion and the invasive behavior of tumors have long been known and are the subject of many publications, observations, and hypotheses [1,2]. An editorial on this subject has been published recently [3].
The HLA system is the central focus of cellular and immunological communication; it plays an essential role in the immunological identification of cells and in communication with and within the immune system. Profiling of the HLA system is an essential diagnostic step prior to organ or bone marrow transplantations. In transplantation, there is always a direct contact between the donor organ, typified by HLA classes I and II (HLA A-C; HLA DP-DS), and the recipient’s immune system. This results in more or less severe reactions of the recipient’s immune system. Because of this, prior to transplantations, HLA matching is performed. However, even in cases of a high HLA concordance, additional immunosuppressive therapy is usually necessary.
However, conditions are different in implantation and pregnancy. The embryo/fetus itself does not come into direct contact with the maternal immune system, but the trophoblast and/or placenta does. Although the trophoblastic cells are of embryonic origin, they show a different HLA identification compared to adult epithelial tissue: no classical HLA class I and II proteins (HLA-A, HLA-B, and HLA DP-DS) from the original embryonic/fetal HLA signature are expressed, except for HLA-C [4,5,6,7]. Instead, the “non-classical” HLA groups (class Ib) such as HLA-E [8,9,10], HLA-F [10,11], and HLA-G [12,13,14,15,16,17,18] are expressed. Despite the indirect cell contact of the maternal and fetal classical HLA system, the fetus has a minimum of 50% non-matching HLA compared to the mother. The 50% HLA “miss-match” is derived from the future father; therefore, the embryo is often described as a “semi-allotransplant”. Moreover, in pregnancies after egg or embryo donation or in cases of surrogacy, the embryo is, genetically, completely foreign to the future mother, which might be regarded as “allotransplant”. However, these pregnancies are also successful, even though the mother is not related to the embryo. In these cases, as well as standard mother-to-embryo constellations, implantation and pregnancy comprise a close cellular “network” of the trophoblast and maternal tissue. Due to this special situation, the trophoblast, to maintain pregnancy, expresses the “non-classical” HLA groups in order to escape the maternal immune system. The “non-classical” HLA groups inhibit immunocompetent cells of the maternal immune system by interacting with specific receptors of NK cells (e.g., killer-immunoglobulin-like receptors (KIR)) and lymphocytes (lymphocyte-immunoglobulin-like receptors (LIL-R)). In pregnancy failures and placental disorders, because of an aberrant expression of the non-classic HLA groups, this mechanism is not working properly. Tumor cells use the same immune escape mechanism to evade immune cell elimination. In view of this context, it is necessary to examine these non-classical groups and their interaction with the receptors/receptor families of the various immunocompetent cells more closely.

2. Non-Classical HLA Groups E to G (class Ib)

2.1. HLA-E

HLA-E is the first prominent non-classical HLA class Ib members. It is located 650 kb upstream from HLA-C and flanked by HLA-A on the 3’ position on chromosome 6p21.3. Similar to HLA-B, it consists of seven exons. The first exon encodes the signal peptide, while exons 2, 3, and 4 encode the alpha domains α1, α2, and α3. Exon 5 encodes the transmembrane domain, and exons 6 and 7 encode the cytoplasmatic part of the protein [19].
HLA-E is the least polymorphous HLA antigen with 13 known alleles, resulting in five different proteins [20,21]. The alleles HLA-E*0101 and HLA-E*0103 are the two known functional alleles. They differ by an amino acid substitution (arginine for glycine) at position 107, which results in a differentiated cell surface expression, thermal stability, and peptide binding affinity [22,23]. For a stable surface expression, HLA-E needs to bind intracellular peptide sequences, exclusively derived from signal peptides of classical HLA class I proteins and HLA-G [20,24,25]. Surface expression of HLA-E therefore also depends on expression of the classical HLA groups (class Ia) and HLA-G. Reduced expression of classic HLA groups and HLA-G thus results in a reduced HLA-E expression [23,26]. Besides signal peptide sequences from other HLA groups, HLA-E also binds peptides derived from cell-stress-related proteins such as Hsp60 and pathogen-associated proteins such as the human cytomegalovirus [27,28]. In contrast to other non-classical HLA class I genes, HLA-E is not only expressed in trophoblast cells but also in a variety of nucleated cells [29]. In the trophoblast, HLA-E is mainly expressed during the first trimester of pregnancy [30].

2.2. HLA-F

HLA-F is the second but lesser known member of the non-classical HLA class I family. HLA-F is located on the terminal end of chromosome 6p21.3. Similar to the HLA class Ia genes, the HLA-F gene has eight exons, which likewise encode the signal peptide and the alpha and transmembrane domains. The functions of these exons are similar to HLA-E: the first exon encodes the signal peptide, while exons 2, 3, and 4 encode the alpha domains α1, α2, and α3. Exon 5 encodes the transmembrane domain, while exons 6 and 7 encode the cytoplasmatic part [19]. Thus far, 22 HLA-F alleles with three mRNA transcript variants—HLA-F1, -F2, and -F3—are currently known [21]. However, recent NGS analysis determined 37 haplotypes, resulting in 30 coding haplotypes on the HLA-F region [31]. Unlike the classical HLA groups, exons 7 and 8 are not translated in the transcript variants F1 and F3 [19]. The cytoplasmatic tail is shorter, compared to other HLA genes, and shows greater variation in length between the three isoforms [32]. However, the structure of the protein corresponds to the classic HLA class Ia groups and forms a peptide binding pocket using the α1 and α2 domains. In this classical conformation, HLA-F forms a complex with β2-microglobulin. Besides its ability to bind to β-microglobulin, HLA-F also forms complexes with heavy chains of other HLA class I molecules, possibly to stabilize them [33]. Apart from the classical HLA conformation and complexes with other heavy HLA chains, there is a stable open conformation (OC) of HLA-F characterized by the absence of β2-microglobulin and peptides bound in the peptide binding groove [32].
HLA-F expression is mainly restricted to the intracellular parts of the cell, specifically the endoplasmatic reticulum, but can also be expressed on the surface of trophoblastic and activated cells of lymphocytic origin, e.g., activated B-cells [34]. High expression is restricted to the extravillous trophoblast and declining in the further course of pregnancy [30].

2.3. HLA-G

HLA-G is the most prominent gene within the non-classical HLA group. Similar to other HLA genes, it is located on chromosome 6p21.3: flanked upstream by HLA-A and downstream by HLA-F. HLA-G remains the most polymorphic gene within the otherwise almost non-polymorphic non-classical HLA group. At present, 75 single nucleotide polymorphisms (SNPs) are known for the coding region of HLA-G, resulting in 53 alleles with 18 proteins and associated isoforms [21,35,36]. Similar to the HLA class Ia groups, HLA-G has 8 exons, but the numbering of exons differs between databases. The National Center for Biotechnology Information (NCBI) and the genomic browser from the University of California Santa Cruz (UCSC) start with exon 1 in the possible promoter sequence of HLA-G, resulting in a translational start for the signal peptide in exon 2. Exons 3–5 encode the alpha 1–3 domains, and exon 6 encodes the connecting peptide, the transmembrane domain, and the cytoplasmatic tail. The IMGT/HLA database denotes the exon that encodes the signal peptide as exon 1, resulting in a shifted exon numbering for the translated protein. In all databases, exon 8 remains untranslated due to the presence of a stop code in exon 7. The region of the non-translated exon 8 is designated as the “3´untranslated region (3′UTR)” [37]. Eighteen SNPs, a 14 bp insertion/deletion, and 44 haplotypes are currently described for the 3′UTR region [36], which are known to influence the translation of HLA-G proteins by reduced transcription, mRNA stability, or aberrant alternative splicing. Besides sequence variants, six micro-RNAs (miRNAs) (miR-133a, miR-148a, miR-148b, miR-152, miR-548q, and miR628-5q) are also known to downregulate HLA-G protein expression by binding to the 3′UTR. [36].
Due to alternative splicing of the primary transcript, HLA-G can be expressed as four membrane-bound isoforms (HLA-G1 to -G4) and, in contrast to classical HLA class I genes, as three soluble isoforms (HLA-G5 to -G7) [38]. HLA-G1 and the soluble HLA-G5 represent the complete extracellular protein structure composed of three alpha domains (α1–α3) and may be bound to β2-microglobulin (β2m) [39]. The other isoforms are not associated with β2m and differ in their extracellular protein structure. The soluble isoforms show a high structural similarity with the membrane bound isoforms HLA-G1 and HLA-G2 but preserve the intron 4 (HLA-G5 and -G6) [40,41,42]. HLA-G7 is the soluble structural homolog of HLA-G3 and comprises only the α1 domain. The absence of transmembrane domains is the result of the translation of intron 2, which encodes two amino acids bound to the α1 domain [37]. Besides the different known protein isoforms, proteasome-generated spliced peptides for classical HLA groups have been described, accounting for approximately one-third of the whole antigens [43]. Considering the genetic and structural similarities between classical HLA class I genes and HLA-G, further HLA-G peptides and proteins could be generated through this post-transcriptional modification. It is assumed that they have an impact on T-cell response [44].
In addition to these isoforms, HLA-G can also occur in several protein complex formations. Similar to the classic HLA proteins, HLA-G1 exists as a monomer bound with β2-microglobulin. HLA-G1 and HLA-G5 are able to form heterodimers and homotrimers by establishing disulfide bridges between the α1 domains (via cysteine 42). The remaining membranous isoforms do not form a complex with β2-microglobulin, but, similar to HLA-G1 and HLA-G5, can also form hetero- and homodimers.
Compared to HLA-E and -F, HLA-G protein expression is only restricted to trophoblast cells in pregnancy. HLA-G1 is the main isoform and is expressed in extravillous trophoblast cells (EVTs), e.g., endothelial EVTs, interstitial EVTs, and villous trophoblast cells. EVTs further express the isoforms HLA-G2, -G5, and -G6, while the villous trophoblast cells only express the soluble homolog of HLA-G1, i.e., HLA-G5, which is released into the maternal bloodstream [13,45].

3. Interaction of HLA-E to -G with Receptor Families of Immunocompetent Cells

As mentioned above, the non-classical HLA genes predominantly mediate immune evasion and immune suppression by inhibiting cells of the adaptive and innate immune system, e.g., natural killer cells (NK cells), T- and B-lymphocytes by interacting with the inhibitory leukocyte immunoglobulin-like receptors B1 and B2 (LILRB1 and LILRB2), killer cell immunoglobulin-like receptor 2DL4 (KIR2DL4), and the NK receptor group 2 (NKG2)/killer cell lecithin-like receptors (KLRs) [46,47].

3.1. Receptor Interaction of HLA-E

HLA-E interacts with the killer cell lectin-like receptor C1 (KLRC1), also known as NKG2A, -B and -C which is expressed by NK cells [48]. The NKG2A receptor belongs to the NKG2 receptor family, which represents the second group of NK cell receptors, beside KI-receptors (KIR) [8,49]. These receptors are type II transmembrane proteins that have an extracellular C-type transmembrane domain. The NKG2 receptor family can be subdivided into six subgroups: -A, -B, -C, -D, -E, and -H. Some are splicing variants of the same gene (A/B and E/H) [49]. The NKG2A receptor together with the NKG2B receptor belongs to the inhibitory receptors within the NKG2 family, which mediate an inhibitory signal to the NK cell via immunoreceptor tyrosine-based inhibition motifs (ITIMs) [50]. Upon binding of HLA-E to NKG2A, the ITIMs bind to the Src homology region 2 domain-containing phosphatase-1 (SHP-1), which dephosphorylates signaling molecules in the signaling pathway of the immunoreceptor tyrosine-based activating motifs (ITAMs) and thus sends an inhibitory signal to the cell [51]. Similar to NKG2B, -C, -E, and -H, the NKG2A receptor dimerizes with CD94 to form a stable complex [52,53]. CD94 is expressed by NK cells and plays a role in recognizing HLA class I proteins. With the exception of CD94/NKG2D, the CD94/NKG2 receptor family only recognizes HLA-E as a ligand [52]. However, the affinity of HLA-E towards single NKG2 subtypes varies. HLA-E binds preferentially to the inhibitory receptor CD94/NKG2A rather than the activating receptor CD94/NKG2C (Figure 1) [52]. Activating receptors such as CD94/NKG2C do not possess intracellular motifs. For signaling transduction, the CD94/NKG2C receptor contains a positively charged transmembrane domain with DNAX activation protein 12 (DAP-12), which has an ITAM in its cytoplasmic domain and transmits an activating signal to the cell [51,54,55]. Once activated, the signaling transduction protein DAP12 recruits the spleen tyrosine kinase (Syk) and zeta-chain-associated protein kinase 70 (ZAP70), which stimulate increased cytotoxicity and cytokine production in NK cells [56]. HLA-E inhibits NK cell activation and proliferation via the inhibitory NKG2A receptor [57]. Activation via the NKG2C receptor only occurs after binding of a restricted repertoire of peptides such as CMV, A80 and B13 [58]. However, HLA-E that has bound HLA-G leader peptide sequence engages, the activating receptor of which is CD94/NKG2C. This binding results in an increased proliferative activity and elevated antibody-dependent cytotoxicity [29].

3.2. Receptor Interaction of HLA-F

HLA-F interacts mainly with the receptors LILRB1 and LILRB2 as a tetramer belonging to the family of leukocyte-immunoglobulin (Ig)-like receptors (LILR) (Figure 2) [34,59]. They comprise five activating (A) receptors (LIRA1, -2, -4, -5 and -6), one soluble receptor (LIRA3), and five inhibitory (B; “blocking”) receptors (LILRB1–5) [60]. The LILRB1 receptor (also known as Ig-like transcript 2 (ILT2) or CD85j) is an inhibitory receptor expressed in monocytes, dendritic cells (DCs), as well as B-, T-, and NK cells. The receptor has four ITIMs in its intracellular domain and transmits an inhibitory signal to the immunocompetent cell via SHP-1 and -2. During T-cell activation, cytotoxicity is inhibited exclusively via SHP-2, which induces blockade of the mechanistic target of rapamycin (mTOR) signaling pathway [61]. The LILRB1 receptor is able to prevent the proliferation of antigen-specific γδ-T-cells by forming complexes with further LILRB1 receptors [62]. This homodimer of LILRB1 receptors induces the synthesis of interleukin-10 (IL-10) and the transformation of growth factor beta (TGF-β) and inhibits the synthesis of the proinflammatory cytokine interferon-gamma (IFN-y) [46,63,64]. The LILRB2 receptor, also known as ILT4, belongs to the inhibitory receptors and is primarily expressed by dendritic and endothelial cells [65]. Unlike LILRB1, LILRB2 has only three ITIMs, which are able to recruit SHP-1 and -2, mediating an inhibitory signal to the DCs [39]. The receptors LILRB1 and B2 bind to β2-microglobulin and to the α3 domain of the MHC Class I and recognize numerous HLA Ia and Ib molecules in addition to HLA-F.
HLA-F as an open conformer (OC) is able to bind to the activating NK cell receptor KIR3DS1, KIR2DS4 and to the inhibitory NK cell receptor KIR3DL1 and -2 [66,67,68]. The receptors KIR3DS1, KIR2DS4, KIR3DL1 and -2 belong to the family of killer cell immunoglobulin-like receptors (KIRs), which are expressed in NK cells as transmembrane glycoproteins. They are distinguished and classified by their number of extracellular domains (two or three domains, 2D or 3D) and the length of their intracellular signaling domains. KIR subtypes with short (S) intracellular domains are classified as activating receptors because their transmembrane domain contains a charged lysine residue instead of an immunoreceptor tyrosine-based inhibition motif (ITIM) [69]. KIRs such as KIR3DL1 with a long cytoplasmic domain (L) belong to the category of inhibitory receptors; their cytoplasmic domain also contains an ITIM, which mediates an inhibitory signal to NK cells after activation by the protein tyrosine phosphatases SHP-1 and SHP-2 [70,71]. However, HLA-F OC possesses the highest binding affinity for the activating receptor KIR3DS1. Via the charged transmembrane domain, KIR3DS1 dimerizes with DAP-12, which transmits an activating signal to the NK cell via ITAMs. Binding of HLA-F to KIR3DS1 results in NK cell activation [72,73].

3.3. Receptor Interaction of HLA-G

HLA-G interacts with various receptors, such as LILRB1, LILRB2, CD8, CD160 and KIR2DL4, which are expressed by cells of the adaptive and natural immune system (Figure 3) [36]. The inhibitory receptor LILRB1 is found in monocytes, DCs, and B-, T-, and NK cells: The LILRB2 receptor is only expressed in dendritic and on endothelial cells, which bind to β2-microglobulin and to the α3 domain and thus recognize numerous HLA class Ia and Ib molecules. Because of the hydrophobicity of the α3 domain in the HLA-G molecule, receptors favor HLA-G as an interaction partner for binding [74]. The binding affinity of receptors LILRB1 and LILRB2 increases upon multimerization of the HLA-G molecules [38]. However, the two receptors recognize different structure conformers of HLA-G: LILRB1 preferentially binds to HLA-G in a complex with β2-microglobulin, while LILRB2 also recognizes the free heavy alpha domains of HLA-G without bound β2-microglobulin [75,76]. The interaction of soluble and membrane-bound HLA-G with LILRB1 or LILRB2 causes direct and indirect immune-suppressive effects on immune cells [77]. The direct immune inhibitory mechanism is mediated by the direct binding of HLA-G to these receptors, which causes an inhibition of cytotoxic T-cells as well as NK cells [78,79]. As an indirect effect HLA-G inhibits the proliferation of allo-specific CD+ T-cells [80]. As the LILRB1 receptor is also expressed by DCs and B-cells, the interaction with HLA-G affects their function and maturation as well and induces the generation of HLA-G expressing and tolerogenic DCs such as DC-10 [81]. HLA-G+ APCs can induce immunosuppressive CD4+ T cells and, in the case of DC-10, mediate the generation of type 1 regulatory T cells [82,83]. The receptor KIR2DL4 (also known as CD158d) is a special type among KIR receptor subtypes. Unlike other KIR receptors, KIR2DL4 interacts within the non-classical HLA group only with HLA-G [55]. The receptor contains the two extracellular domains D0 and D1 and has only one ITIM. A charged arginine residue enables KIR2DL4 to form a complex with Fc fragment receptor γ (FcRγ). FcRγ has two ITAMs, which transmit the activation signal to the NK cells. The complex formed with FcRγ stimulates the NK cell to enhanced chemokine and cytokine production [55]. Unlike other KIR receptors, KIR2DL4 cannot be detected on quiescent peripheral NK cells with phenotype CD56dimCD16+. It is primarily expressed in an NK cell subpopulation known as decidual/uterine NK cells (uNK) with the phenotype CD56brightCD16- [84,85,86]. The membrane-bound HLA-G binds to KIR2DL4, stimulating the uNK cells to proliferate and produce IFN-γ [87].
HLA-G also interacts with CD8, a surface marker for cytotoxic T-cells. Binding of soluble HLA-G (sHLA-G) to CD8 induces apoptosis in CD8+ cytotoxic T-cells [88]. In addition to cytotoxic T-cells, CD8 is also expressed in some activated uNK cells [90]. Engagement of HLA-G with CD8 expressed on uNK cells likewise induces apoptosis in this subpopulation [91].
Besides uNK and cytotoxic T-cells, sHLA-G also induces apoptosis in activated endothelial cells through the engagement with the CD160 receptor [92]. CD160 is expressed on CD56dim CD16+ cytotoxic NK cells, CD8+ T lymphocytes, and activated endothelial cells [92]. It is a glycosylphosphatidylinositol-anchored receptor that cannot mediate signals by itself due to missing ITAMs. The receptor relies on the recruitment of adapter proteins such as phosphoinositide-3 kinase in order to provoke cytokine production and cytotoxicity [89].

3.4. HLA-C—A Special Case

Besides the non-classical HLA class Ib genes, HLA-C is the only classical HLA class Ia gene, which is also expressed in extravillous trophoblasts [93]. HLA-C also interacts, according to present knowledge, with different inhibitory receptors, such as KIR2DL1, KIR2DL2, and KIR2DL3, as well as activating receptors, such as KIR2DS2 and KIR2DS1 [69,94,95,96,97]. HLA-C is the only classical HLA class Ia gene, which binds toKIR2DL4, as the non-classical HLA gene HLA-G does. The binding of HLA-C to KIR2DL4 mediates a non-cytotoxic activation of uterine NK cells (uNK, CD 56bright), which leads to the synthesis and release of numerous cytokines or growth factors (e.g., G-CSF, GM-CSF, LIF, IL-4, and IL-10), which likewise promote trophoblast and embryo growth [6,7].

3.5. Significance of HLA-E to -G for Implantation/Pregnancy

As briefly mentioned above, the placenta or trophoblast cells create an interface between the maternal and embryonal tissue. The relevant cells interacting with the maternal immune system are trophoblast cells, which can be subdivided into cytotrophoblasts (CTs), syncytiotrophoblasts (STs), and EVTs. They emerge from the outer cell mass from early blastocysts and subsequently invade the endometrium. These trophoblast cells are the first to encounter maternal immune cells. During trophoblast invasion, the amount of uNK cells and M2 macrophages, in this context also denoted as decidual macrophages, immediately increases. The uNK cell and macrophage migration might be enhanced by the invading trophoblast cells, which secrete NK cell chemoattractants such as MCP-1 [98]. Uterine NK cells are the dominating immune cells at the beginning of pregnancy, constituting 70–90% of all leukocytes in the decidua [98]. Besides uNK cells, antigen presenting cells (APCs) are the second most abundant population of leukocytes in the decidua [99]. DCs such as DC-10 represent the majority within this population and significantly support the induction of the fetal-maternal immune tolerance. DC-10 cells represent a unique subpopulation, as they express high levels of LILRB1 and -B2 as well as HLA-G. Moreover, they secrete interleukin-10 (IL-10), an immunosuppressive cytokine that induces HLA-G expression [83]. Besides DCs, decidual macrophages also support spiral artery remodeling and placental growth by the secretion of angiogenic growth factors (such as angiogenin, keratinocyte growth factor, fibroblast growth factor B (FGF-B), vascular endothelial growth factor A (VEGF-A), and angiopoietin-1 and -2), matrix metalloproteases (MMP1, -2, -7, -9, and -10), and cytokines (interleukin (IL)-1β, -2, -4, -5, -6, -8, -10, and -13 and TNF-α) [100]. The invading trophoblast interacts with these innate immune cells and T-lymphocytes through the expression of the non-classical HLA groups, here denoted as “embryonic” HLA groups (-E to -G). The expression of the embryonic HLA groups results not only in immune cell inhibition but also in the secretion of the already mentioned growth factors, cytokines and MMPs [101,102]. The HLA expression pattern differs among CTs, STs, and EVTs [15,103]. Moreover, the HLA expression pattern also changes during gestation from implantation to the third trimester [30]. HLA-G and -E are the main HLA class Ib groups expressed during implantation and the first and second trimester to interact with cells of the innate immune system [30]. This interaction supports placentation and the induction of the immune-suppressive milieu to maintain pregnancy. The changes in HLA-G expression across gestation can be recapitulated in plasma samples of pregnant women. During the first trimenon, plasma levels of HLA-G1/G5 increase significantly and thereafter decreases continuously till childbirth [104].
However, with regard to HLA-F, the data concerning expression changes during pregnancy are inconsistent. Shoubu et al. observed that HLA-F is expressed on the surface of EVTs and increases during pregnancy [11], whereas Hackmon et al. demonstrated that HLA-F expression in EVTs decreases [30]. However, Hackmon et al. could also detect an increase of HLA-F expression in the fetal villous mesenchyme during the course of gestation [30].
HLA-E, -F, and -G have also been identified in preimplantation embryos (PIEs) and embryonic stem (ES) cells (also derived from Wharton-jelly) [105,106,107,108]. Preimplantation embryos (Day 3–5) are able to secret extracellular vesicles (EVs) with a diameter of 50–200 nm into the culture media, a phenomenon that has been recently described and named as secretome [109]. The concentration of HLA-G in these EVs is positively correlated with the implantation and pregnancy rate [109,110] (Table 1). In the case of the preimplantation embryo, mainly soluble forms of HLA-G are synthesized [16]. High levels of soluble HLA-G are believed to increase the probability of implantation and higher pregnancy rates in an IVF/ICSI program [18,111,112]. However, these data are still preliminary and a multicenter study demonstrated widely differing results [113]. Moreover, a recently published meta-analysis demonstrates the opposite: the presence of soluble HLA-G in the culture medium results in higher implantation and pregnancy rates [114]. Unfortunately, there is no information which soluble HLA-G isoforms were measured—G5, G6, or G7. However, the knowledge of individual isoform patterns (and polymorphisms) seems to be crucial to better understand the immunological interactions [115]. While HLA-G1 predominates in women with undisturbed pregnancies, women with recurrent spontaneous abortions (RSAs) show higher levels of the short membrane-associated HLA-G4 [116]. RSA patients also show lower levels of soluble HLA-G in maternal serum [117]. This seems to be particularly true for the soluble isoform G5, but not G7 [118]. Moreover, low HLA-G5 levels are often accompanied by low HLA-G1 levels [119].
One possible reason for an aberrant HLA-G protein expression might stem from variations in the genomic sequence of HLA-G. It has been observed that certain SNPs and a 14-pb insertion/deletion in the 3′UTR region influence the HLA-G protein expression. Several studies have found that variations in the 3′UTR, in particular the 3′UTR 14bp deletion [116] or insertion/deletion [115,120,121], increase the risk not only of miscarriage [122,123] but also of repetitive implantation failures (RIFs) [124]. In addition, the HLA-G alleles HLA-G*01:04 and HLA-G*01:05A are likewise accompanied by an increased risk of miscarriages [116,125]. Similar results were noted by Vargas et al. for HLA-G*01:04:01 and HLA-G*01:01:18 [9] and Hashemi et al. for the G + 3142 > C polymorphism [121]. HLA-G gene polymorphisms such as 01:06, 01:01:06, 01:01:01:06, and 01:05N (null) alleles were significantly higher in patients with RIFs in an IVF/ICSI program [126]. In addition to HLA-G polymorphisms, an association between HLA-E polymorphisms and RSAs has been observed. The HLA-E 0101 allele, compared with the HLA-E 0103 allele, was significantly higher in women suffering from RSAs [127].
Regarding HLA-F, less is known about aberrant expression levels or polymorphisms in RIFs or RSAs.

4. “Embryonic“ HLA Genes in Tumors

4.1. HLA-G Expression in Cancer

Besides trophoblasts, embryonic HLA genes are also expressed by malignant tumors to escape immune cell recognition and subsequent elimination. This assumption is based on studies investigating the most prominent non-classical HLA gene HLA-G [15,36,47,128,129]. Importantly, HLA-G mRNA and protein expression can be found in a broad spectrum of cancer types including breast cancer [130,131,132,133,134,135,136,137], ovarian cancer [79,138,139,140], endometrial cancer [141], cervical cancer [136,142,143,144,145], prostate cancer [146], lung cancer [147,148,149,150,151,152], bladder cancer [153,154,155,156], colorectal cancer [29,35,157,158,159], colon cancer [160], thyroid cancer [161,162], esophageal cancer [163,164], melanoma [165,166,167,168], lymphoma [169], Hodgkin´s lymphoma [170], renal cancer [171,172,173], glioblastoma [174,175,176,177,178], germ cell tumor (testicular) [179] and pancreatic cancer [180] (Table 2). The vast majority of publications show consistently that high expression of class Ib groups, in particular HLA-G, is associated with particularly poor prognosis [163,164,181,182,183,184,185,186]. This may be due to the immune-suppressive property of HLA-G to inhibit proliferation and cytotoxic activity of tumor infiltrated T- and NK cells. Wan et al. reported that HLA-G expression in tumors not only correlates with lower numbers of tumor infiltrating NK cells but also inhibits the cytotoxic potential of residing NK cells probably by binding to their inhibitory receptor LILRB1 [187]. This observation was also found in non-small cell lung cancer, where increased HLA-G and LILRB1 protein expression correlated with increased tumor stage [149]. However, HLA-G interacts not only with LILRB1 but also with the NK cell specific receptor KIR2DL4. Similar to the physiological function during pregnancy, an activation of the KIR2DL4 receptor via HLA-G in tumors not only results in NK cell inhibition but also stimulates the production of growth factors, cytokines, and the release of MMPs. Coexpression of HLA-G and KIR2DL4 therefore worsens cancer prognosis by mediating cancer invasion and metastatic spread, as has been reported in breast cancer [188]. Besides direct immune cell inhibition, an additional indirect mechanism has been shown for the isoforms HLA-G1 and -G5, which are able to induce the generation of HLA-G positive regulatory T-cells (Tregs) and HLA-G+ APCs, such as DC-10 as well as HLA-G positive macrophages (via trogocytosis) [189]. HLA-G+ APCs are able to inhibit the proliferation of CD4+ T-cells and induce the differentiation of CD+ T-cells to Tregs [189,190]. In addition to direct cell–dell interactions HLA-G positive, tumor-associated macrophages (TAMs), can also secrete HLA-G, thereby promoting and supporting an immune-suppressive milieu surrounding cancer cells to further enhance immune evasion. Such TAMs can built up to 50% of the total tumor mass and are consistently associated with a poor prognosis [191]. In addition, tolerogenic DC-10 supports the immune inhibiting effect of TAMs by expressing membrane-bound HLA-G and secreting IL-10. IL-10 can also be synthesized by tumor cells and, in turn, upregulates the expression of HLA-G [192]. The impact of combined expression of IL-10 and HLA-G has been evaluated in lip squamous cell carcinoma (LSCC), where high levels of HLA-G and IL-10 protein expression could be observed only in carcinoma lesions but not in normal tissues [193]. As an additional mechanism, IL-10 induces Type 1 T regulatory (Tr1) cells mediated by the IL-10-dependent ILT4/HLA-G pathway [83].
By mediating an immune-suppressive surrounding, HLA-G counteracts the anti-tumor effect of tumor-infiltrating lymphocytes (TILs). This assumption was supported by Dong et al., who demonstrated that high HLA-G expression was inversely associated with TIL infiltration in breast cancer. Strikingly, they also found that, breast cancer patients with a high HLA-G protein expression and a low TIL infiltration had a significantly higher risk of recurrence compared to patients with low HLA-G expression and a high TIL density [194]. Moreover, it could be shown that inflammatory TILs itself may express HLA-G, as has been demonstrated, e.g., in melanoma [195].
These immune-suppressive effects of HLA-G support the observation that HLA-G expression is associated with poor prognosis. In contrast, some reports have also described opposite associations of HLA-G expression with prognosis in some cancer indications. As one example, Rutten et al. reported that HLA-G protein expression was positively associated with prolonged progression-free survival and improved response to chemotherapeutical treatment in high grade ovarian cancer [139].
However, in all these studies HLA-G protein expression has been has been determined by antibodies detecting all HLA-G isoforms (i.e., clone 4H84, clone MEM-G1 and MEM-G/9). Only a few studies have investigated HLA-G protein isoform expression, by additionally using the anti-HLA-G antibody clone 5A6G7, which recognizes particularly the isoforms HLA-G5/-G6. Zhang et al. evaluated HLA-G5/-G6 expression in ovarian cancer via immunohistochemistry by applying the specific anti-HLA-G antibody clone 5A6G7. Interestingly, they did not find any association of HLA-G5/G6 expression associated with clinical outcome or other variables such as age, histological type, or FIGO stage [138]. In contrast, in non-small-cell lung cancer, immunohistochemical HLA-G5/-G6 expression did discriminate between adenocarcinomas and squamous cell carcinoma [150,196]. It has to be mentioned that that HLA-G isoform expression is not homogeneous within carcinoma entities and mRNA as well as protein expression or synthesis may vary within one tumor [171,196,197]. Besides the known HLA-G isoforms, shed isoforms may occur through proteolytic cleavage by metalloproteinases generating soluble HLA-G1 mediated [198]. Lin et al. assumed the existence of an yet unidentified HLA-G isoform, lacking the alpha 1 domain but containing intron 4 in colon carcinoma [197]. Besides HLA-G isoform expression, polymorphic variants within the DNA sequence such as the 14 bp insertion/deletion in the 3′UTR might also have an impact on the overall increase of cancer risk due to its influence on mRNA stability and protein expression. Li et al. investigated a meta-analysis to evaluate this assumption. The 14 bp insertion/deletion did not significantly correlate with cancer susceptibility, except for breast cancer [199]. A similar observation was published by Haghi et al., demonstrating that the 14 bp deletion was significantly more frequent in patients suffering stage II and III breast cancer than in patients with stage I breast cancer [200].

4.2. HLA-E Expression in Cancer

In addition to HLA-G, HLA-E expression can be observed in multiple cancer entities such as breast cancer [132], colorectal cancer [158,159,160,201], renal cancer [173,202,203], lung cancer [204], melanoma [205,206], and gastric cancer [207,208], cervical cancer (adenocarcinoma) [209], glioblastoma [174,177,210], Hepatic carcinoma (hepatocellular) [211], Hodgkin’s lymphoma [212], Thyroid cancer [213] and leukemia [214]. Table 3 shows an overview of tumors in which expression of HLA-E has been detected. Importantly, the occurrence of HLA-E/β2 microglobulin complex, which interacts with the inhibitory NKG2A receptor, has been associated with bad prognosis in colon cancer potentially by contributing to immune evasion of the tumor [159,182]. In addition, HLA-E monomers not bound to β2 microglobulin was also associated with bad prognosis [208]. In line with this, the risk of metastasis is increased in rectal cancer expressing HLA-E [158]. Moreover, the favorable effect on prognosis exerted by TILs is counteracted by a high expression of HLA-E [204,215,216,217]. However, contradictory effects have been observed. Benevolo et al. reported that high HLA-E expression is associated with good prognosis and could therefore serve, in combination with a high expression of HLA-A, as a prognostic marker. They also investigated the expression of the inhibitory NK receptor NKG2A on TILs and determined a positive correlation with HLA-E expression [201]. In renal cell carcinoma HLA-E protein expression determined by IHC was not associated with disease-specific survival, but inversely correlated with the presence of CD56+ NK cells [202]. These findings consolidate the immunogenic role of HLA-E based on its interaction with the NK cell receptors CD94/NKG2A, -B, -C, and -D, which may be also expressed in CD8+ T-lymphocytes to lesser extent [8,28,50]. However, based on these data the prognostic value of HLA-E remains to be uncertain. One possible explanation might be the missing analysis of the two functional HLA-E*0101 and HLA-E*0103 alleles, which are known to differ in their cell surface expression, thermal stability, and peptide binding affinity [22,23]. Interestingly, Wagner et al. evaluated HLA-E*0101 and HLA-E*0103 alleles in patients suffering chronic lymphocytic leukemia. They observed that patients with the HLA-E*0103 allele are in need of early treatment [218]. However, in renal cell cancer Seliger et al. found no effect on overall survival upon overexpression of HLA-E, while an inverse correlation with tumor infiltrating CD56+ NK cells could be demonstrated [202].

4.3. HLA-F Expression in Cancer

While several studies have found significant HLA-F expression in several cancer types, the data are even less conclusive. Table 4 lists studies showing HLA-F expression in tumors. HLA-F expression could be observed in breast cancer [219], gastric cancer [207,220], bladder cancer [221], nasopharyngeal cancer [222], esophageal squamous cell cancer [185], hepatocellular carcinoma [186], neuroblastoma [223] and lung cancer [186]. However, no clear correlation with a higher tumor burden or decreased disease specific survival could be drawn. As one example, Zhang et al. observed that HLA-F expression did not correlate with prognosis for gastric cancer patients [224]. However, patients with a positive coexpression of HLA-E and -F suffering gastric cancer had a significantly lower five-year survival rate and a lower postsurgical outcome [207]. In addition, in stage II breast cancer, HLA-F expression correlated with poorer outcome, compared to the HLA-F negative group [219]. The same observation could be found for non-small-cell lung cancer [186] and hepatocellular cancer [184]. One possible reason for the inconsistence of the current data might be that no study distinguishes between the individual isoforms, complex variants, open versus closed conformation and/or antisense polymorphisms. As previously described, HLA-F interacts with LILRB1 and -2 receptors of tumor infiltrating monocytes such as monocytes, DCs, and B-, T-, and NK cells when they migrate into the tumor tissue. However, few data exist regarding the immune-suppressive effect of HLA-F expressed on cancer cells with respect to TILs. Furthermore, contradictory results exist for melanoma patients, where HLA-F expression was not inversely related with the amount of TILs [225].

4.4. HLA-C Expression in Cancer

As already described, HLA-C is a special case: as part of the classic HLA complex, it is expressed on nearly every cell type. However, HLA-C is overexpressed on certain tumor cell lines, particularly Cw, which is accompanied with poor prognosis [226,227]. HLA-C primarily activates NK cells (see above), thereby possibly inducing production of cytokines and growth factors that drive malignant cell growth, resulting in tumor promotion. This phenomenon that TILs can also support tumor proliferation rather than limit tumor growth is also well known and contributes to worsen prognosis [191,228,229,230].

4.5. Soluble “Embryonic” HLAs in Cancer

The existence of secreted and/or shed embryonic HLA proteins resulting insoluble fractions detected in sera from tumor become of increasing interest. Serum HLA-G levels have been determined in high-grade ovarian, colorectal, gastric, esophageal, lung, and breast cancer, melanoma, and neuroblastoma [140,163,231,232]. Soluble HLA-G (sHLA-G) levels were significantly increased in plasma in breast cancer patients compared to healthy controls and even correlated with the histological type [233]. Interestingly, serum HLA-G levels are associated with estrogen receptor expression and disease progression at the point of diagnosis. This is of particular importance, as estrogen receptor positive “luminal” cancers are well known to have less TILs despite having higher frequencies of multiple signal transduction oncogenes such as PIK3CA compared to estrogen receptor negative tumors. The estrogen dependent HLA gene expression might serve as one underlying mechanism for this phenomenon. Moreover, the high serum level of HLA-G in extracellular vesicles (EVs) correlated with disease progression before neoadjuvant chemotherapy [232]. In lung cancer, sHLA-G in plasma discriminated non-small-cell lung cancer (NSCLC) from small-cell lung cancer (SCLC) and served as a prognostic marker with high levels of sHLA-G indicating reduced overall survival (OS). Similarly, in melanoma, sHLA-G levels were also increased in serum from melanoma patients compared to healthy controls. Furthermore, melanoma patients receiving interferon-alpha (IFN-α) treatment exhibited further increase of sHLA-G levels. Interestingly IFN-α also upregulated HLA-G cell surface expression in circulating monocytes [231]. Morandi et al. corroborated this observation through the detection of sHLA-G secreting monocytes, which had been activated by neuroblastoma tumor cells to secrete sHLA-G [234]. Morandi also evaluated soluble HLA-E (sHLA-E) and -F (sHLA-F) levels in plasma samples from patients suffering from neuroblastoma. Importantly, patients with metastatic disease had higher sHLA-E plasma levels than patients with a localized tumor. In addition to this finding, high sHLA-E and -F levels were observed in relapse-free patients and patients having better OS. [223]. Similar to the findings concerning sHLA-G, sHLA-E levels are also significantly increased in melanoma patients compared to healthy controls [206]. In addition, sHLA-E levels were also significantly elevated in patients suffering from chronic lymphocytic leukemia and were associated with advanced disease stage [218].
In summary, there is consistent evidence in the literature that these HLA groups represent an important factor in determining prognosis [129,142,143,158,160,162,163,216]. This primarily applies to HLA-G and its isoforms, while HLA-E and -F are far less investigated.

4.6. Metastases

In clinical tumor therapy, surgical removal of metastases is still an uncommon approach except in rare cases of solitary metastases or oligometastatic disease. Because of that, there are few publications on this issue with regard to HLA expression in metastatic lesions. The expression of HLA-G has been shown in cancer stem cells (CSCs) of certain types of leukemia [235] and renal cancer [236] and in lymph node metastases of thyroid cancer [162], gastric cancer metastases [237], metastases of ovarian cancer [141], malignant melanoma [165,166,205], colon cancer [157], and breast cancer [219,238]. A strong expression of HL-G in the primary tumor increases the likelihood of metastases [128]. However, little is known about the individual isoforms expressed in metastatic tumor tissue. There is evidence of the expression of G1 or the “shed” variant, primarily in colon cancer metastases [128]. Furthermore, HLA-E and -F expression has been found in metastases from breast cancer [132,219].

5. Significance for Immunoncology

General Considerations

Since trophoblast cell invasion and placentogenesis show biological features similar to those of carcinogenesis, malignant tumor invasion and growth could be mediated by similar cellular pathways [3]. It is likely that malignant cells, expressing “embryonic” HLA groups (i.e., HLA class Ib), mediate the same immunological effect as observed in implantation and pregnancy [196]. It has been proven that the expression of “embryonic” HLA groups by tumor cells leads to a similar inhibition of the innate and adaptive immune system and thereby enables tumor immune escape (therapy). This “escape phenomenon” of tumors has long been known and can also be observed in the course of neoadjuvant and adjuvant chemotherapy [22,37,160,168,170,172,178,181,183,239,240,241]. Since the immune evasion mechanism mediated by the expression of immune checkpoints has long been proposed as the major reason for chemotherapy resistance, the investigation of an immune checkpoint therapy has been established. Prominent immune checkpoints are programmed cell death 1 (PD-1) and its ligand (PD-L1), cytotoxic T-lymphocyte associated protein 4 (CTLA4), and indoleamine-2,3-dioxygenase (IDO-1). These immune checkpoints mainly target T-cell response. Anti-PD1 (prembolizumab and nivolumab), anti-PD-L1 (atezolizumab, durvalumab, and avelumab), and anti-CTLA4 (ipilimumab) therapies have been introduced to treat multiple kinds of cancer. However, while having provided substantial promise as a new treatment strategy, the modulation by immune checkpoint therapies are only successful in 15–20% of patients [241,242,243,244,245,246]. One reason for this limited activity of checkpoint inhibitors might be the simultaneous expression of embryonic HLA genes on cancer cells, which govern an independent immune inhibition mechanism. Early hints in this direction stem from studies of intraoral mucoepidermoid carcinomas, where HLA-G, HLA-E, and PD-L1 expression was determined via immunohistochemistry [247]. Similarly, Lopes et al. observed coexpression of PD-L1 and HLA-G proteins in lip carcinomas. This coexpression was associated with a higher malignancy and occurrence of systemic metastases [246]. Therefore, it is conceivable that, when confronted with protective mechanisms similar to the defense mechanisms in pregnancy, immune oncological therapeutic approaches are less effective than expected [248]. It has to be noted that, compared to trophoblastic cells, cancer cells express the classical HLA class Ia genes (HLA-A, -B, and -C) and therefore escape NK cell recognition and elimination through the interaction with KIRs [249]. These conditions are fundamentally different from pregnancy, where the embryo has foreign antigenicity and needs more intensive protection.

6. Hypotheses for an Immunological Tumor Therapy Concept (ITTC)

The patent focuses on determining the individual expression pattern of the new immune-oncological targets to dissect the cellular communication of specific malignant cell clones with the innate and adaptive immune system (Figure 4). This shall lead to tailored immune oncological treatment modalities to break the resistance of tumors refractory to check point inhibition. These assumptions are based on our detailed knowledge from implantation and pregnancy, where the communication has to be regarded as multifactorial and depends on the physiologic expression of non-classical HLA class Ib isoforms to prevent spontaneous abortions and enable implantation, growth and development of the embryo. Apparently, the communication based on non-classical HLA genes is extremely successful and is crucial to allow the genetically (semi-) foreign embryo or fetus to grow undisturbed despite close cellular interdigitation.
In their untreated state, malignant tumors are also “successful” in terms of unrestricted growth, immune escape, and “implantation” into distant organs. They certainly use multifactorial—not monofactorial—cell communication by expression of various types of immune inhibiting factors including the non-classical HLA class Ib gene, while not being restricted to check point modulators. The resulting immune inhibition and evasion is based on a landscape of multiple inhibiting receptors expressed in cells of the innate and adaptive immune system by far exceeding the check point receptors. Therefore, the identification and determination of the embryonic HLA groups expression pattern on malignant tumors (including isoforms) defines the extent of the underlying immune cell inhibition and communication. However, this in turn means that any monofactorial anti-tumor treatment may be ineffective in most situations. For this reason, an extensive HLA profiling including robust target quantitation for every malignant tumor seems to be necessary. Importantly, it is reasonable to monitor the HLA expression patterns for subsequent treatment adaptations as metastases are known to be able to modify their biology. As has been observed in breast cancer, metastasis can frequently convert from being an estrogen-receptor-positive primary tumor into an estrogen-receptor-negative metastasis [250,251]. As hormone receptors i.a. also control HLA gene expression this has to be taken into account. In line with this, HLA expression or the underlying gene activation also frequently changes during disease progression [132,165,252]. Because of that, HLA profiling including robust quantitation of individual HLAs should be repeated in cases of metastases (“metastatic surgery”).
According to the initial companion diagnostic concepts in breast cancer, where Her-2/neu status has to be determined before tailored therapy regimen are given, an adopted approach for the embryonic HLA genes seems to be reasonable. If a tumor/metastasis expresses “embryonic” HLA groups or specific isoforms treatment by tailored antibodies is a promising option. Human antibodies against HLA-E, -F, and -G are partly already commercially available [42].
Specific antibody treatment should be administered based on the results of HLA profiling. The following treatment strategies/concepts are possible:
  • Masking antibodies mask the corresponding HLA groups/isoforms, blocking the “escape mechanism” of the tumor cells and allowing the immune system to attack the tumor as normal.
  • The concept of special antibodies, e.g., coupled with receptors, weak radiation, or chemotherapy drugs, is based on activating the defenses of the immune system as well as attacking the tumor cells directly.
  • As radiation and, to a lesser extent, chemotherapy drugs can change the steric structure of an antibody, the antibody would be administered first, followed by an application of the radiation particle or the drug.
The underlying rationale of this patent offers several further treatment possibilities. Instead of applying antibodies, in situ gene editing could be performed to erase the expression of non-classic HLA groups (patent pending). Another approach is the blockage of the corresponding receptors on NK cells and lymphocytes, similar to the concept of, e.g., the PD-1/PD-1-ligand therapeutic system (patents pending). Furthermore, vaccination/immunization approaches by injecting non-viable tumor cells or their membranes (if expressing neo-antigens) could be a further therapeutic concept. The strategy of this approach is the blocking or eradication of non-classical HLA groups on the surface of the non-viable tumor cells prior to injection (patent pending) to enable recognition and antibody generation against the exposed neo-antigens. Based on the assumption that the HLA system may dominate other immune escape mechanisms, it will be intriguing to track clinical approaches evaluating individual HLA proteins as new anti-cancer targets with striking potential synergy to current check point inhibitors. However, for this purpose, a robust quantitation of the individual HLA profile seems to be crucial to reduce potential side effects and tailor the most important treatment option.

Funding

This research received no external funding.

Acknowledgments

The authors would like to thank Elke Veltrup and Getraud Würfel for excellent advisory help and support.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Murray, M.J.; Lessey, B.A. Embryo implantation and tumor metastasis: Common pathways of invasion and angiogenesis. Semin. Reprod. Endocrinol. 1999, 17, 275–290. [Google Scholar] [CrossRef] [PubMed]
  2. Perry, J.K.; Lins, R.J.; Lobie, P.E.; Mitchell, M.D. Regulation of invasive growth: Similar epigenetic mechanisms underpin tumour progression and implantation in human pregnancy. Clin. Sci. 2009, 118, 451–457. [Google Scholar] [CrossRef] [PubMed]
  3. Kurlak, L.O.; Knofler, M.; Mistry, H.D. Lumps & bumps: Common features between placental development and cancer growth. Placenta 2017, 56, 2–4. [Google Scholar] [PubMed]
  4. Hiby, S.E.; Walker, J.J.; O’Shaughnessy, K.M.; Redman, C.W.; Carrington, M.; Trowsdale, J.; Moffett, A. Combinations of maternal KIR and fetal HLA-C genes influence the risk of preeclampsia and reproductive success. J. Exp. Med. 2004, 200, 957–965. [Google Scholar] [CrossRef]
  5. Varla-Leftherioti, M. Role of a KIR/HLA-C allorecognition system in pregnancy. J. Reprod. Immunol. 2004, 62, 19–27. [Google Scholar] [CrossRef]
  6. Blais, M.E.; Dong, T.; Rowland-Jones, S. HLA-C as a mediator of natural killer and T-cell activation: Spectator or key player? Immunology 2011, 133, 1–7. [Google Scholar] [CrossRef]
  7. Chazara, O.; Xiong, S.; Moffett, A. Maternal KIR and fetal HLA-C: A fine balance. J. Leukoc. Biol. 2011, 90, 703–716. [Google Scholar] [CrossRef] [PubMed]
  8. King, A.; Allan, D.S.; Bowen, M.; Powis, S.J.; Joseph, S.; Verma, S.; Hiby, S.E.; McMichael, A.J.; Loke, Y.W.; Braud, V.M. HLA-E is expressed on trophoblast and interacts with CD94/NKG2 receptors on decidual NK cells. Eur. J. Immunol. 2000, 30, 1623–1631. [Google Scholar] [CrossRef]
  9. Vargas, R.G.; Sarturi, P.R.; Mattar, S.B.; Bompeixe, E.P.; Silva Jdos, S.; Pirri, A.; Bicalho Mda, G. Association of HLA-G alleles and 3′ UTR 14 bp haplotypes with recurrent miscarriage in Brazilian couples. Hum. Immunol. 2011, 72, 479–485. [Google Scholar] [CrossRef]
  10. Ishitani, A.; Sageshima, N.; Hatake, K. The involvement of HLA-E and -F in pregnancy. J. Reprod. Immunol. 2006, 69, 101–113. [Google Scholar] [CrossRef] [PubMed]
  11. Shobu, T.; Sageshima, N.; Tokui, H.; Omura, M.; Saitom, K.; Nagatsuka, Y.; Nakanishi, M.; Hayashi, Y.; Hatake, K.; Ishitani, A. The surface expression of HLA-F on decidual trophoblasts increases from mid to term gestation. J. Reprod. Immunol. 2006, 72, 18–32. [Google Scholar] [CrossRef]
  12. Kovats, S.; Main, E.K.; Librach, C.; Stubblebine, M.; Fisher, S.J.; DeMars, R. A class I antigen, HLA-G, expressed in human trophoblasts. Science 1990, 248, 220–223. [Google Scholar] [CrossRef]
  13. Hunt, J.S.; Petroff, M.G.; McIntire, R.H.; Ober, C. HLA-G and immune tolerance in pregnancy. FASEB J. 2005, 19, 681–693. [Google Scholar] [CrossRef]
  14. Roussev, R.G.; Coulam, C.B. HLA-G and its role in implantation (review). J. Assist. Reprod. Genet. 2007, 24, 288–295. [Google Scholar] [CrossRef]
  15. Apps, R.; Gardner, L.; Moffett, A. A critical look at HLA-G. Trends Immunol. 2008, 29, 313–321. [Google Scholar] [CrossRef]
  16. Shaikly, V.R.; Morrison, I.E.; Taranissi, M.; Noble, C.V.; Withey, A.D.; Cherry, R.J.; Blois, S.M.; Fernandez, N. Analysis of HLA-G in maternal plasma, follicular fluid, and preimplantation embryos reveal an asymmetric pattern of expression. J. Immunol. 2008, 180, 4330–4337. [Google Scholar] [CrossRef]
  17. Rizzo, R.; Andersen, A.S.; Lassen, M.R.; Sorensen, H.C.; Bergholt, T.; Larsen, M.H.; Melchiorri, L.; Stignani, M.; Baricordi, O.R.; Hviid, T.V. Soluble human leukocyte antigen-G isoforms in maternal plasma in early and late pregnancy. Am. J. Reprod. Immunol. 2009, 62, 320–338. [Google Scholar] [CrossRef]
  18. Rebmann, V.; da Silva Nardi, F.; Wagner, B.; Horn, P.A. HLA-G as a tolerogenic molecule in transplantation and pregnancy. J. Immunol. Res. 2014, 2014, 297073. [Google Scholar] [CrossRef]
  19. Foroni, I.; Couto, A.R.; Bettencourt, B.F.; Santos, M.; Lima, M.; Bruges-Armas, J. HLA-E, HLA-F and HLA-G-The Non-Classical Side of the MHC Cluster. HLA and Associated Important Diseases; InTech: Rijeka, Croatia, 2014. [Google Scholar]
  20. Kraemer, T.; Blasczyk, R.; Bade-Doeding, C. HLA-E: A novel player for histocompatibility. J. Immunol. Res. 2014, 2014, 7. [Google Scholar] [CrossRef]
  21. Robinson, J.; Halliwell, J.A.; Hayhurst, J.D.; Flicek, P.; Parham, P.; Marsh, S.G. The IPD and IMGT/HLA database: Allele variant databases. Nucleic. Acids Res. 2015, 43, D423–D431. [Google Scholar] [CrossRef]
  22. Zheng, H.; Lu, R.; Xie, S.; Wen, X.; Wang, H.; Gao, X.; Guo, L. Human leukocyte antigen-E alleles and expression in patients with serous ovarian cancer. Cancer Sci. 2015, 106, 522–528. [Google Scholar] [CrossRef]
  23. Strong, R.K.; Holmes, M.A.; Li, P.; Braun, L.; Lee, N.; Geraghty, D.E. HLA-E Allelic variants: Correlating differential expression, peptide affinities, crystal structures, and thermal stabilities. J. Biol. Chem. 2003, 278, 5082–5090. [Google Scholar] [CrossRef]
  24. Lee, N.; Goodlett, D.R.; Ishitani, A.; Marquardt, H.; Geraghty, D.E. HLA-E surface expression depends on binding of TAP-dependent peptides derived from certain HLA-F class I signal sequences. J. Immunol. 1998, 160, 4951–4960. [Google Scholar]
  25. Llano, M.; Lee, N.; Navarro, F.; Garcia, P.; Albar, J.P.; Geraghty, D.E.; Lopez-Botet, M. HLA-E-bound peptides influence recognition by inhibitory and triggering CD94/NKG2 receptors: Preferential response to an HLA-G-derived nonamer. Eur. J. Immunol. 1998, 28, 2854–2863. [Google Scholar] [CrossRef]
  26. O’Callaghan, C.A.; Tormo, J.; Willcox, B.E.; Braud, V.M.; Jakobsen, B.K.; Stuart, D.I.; McMichael, A.J.; Bell, J.I.; Jones, E.Y. Structural features impose tight peptide binding specificity in the nonclassical MHC molecule HLA-E. Mol. Cell 1998, 1, 531–541. [Google Scholar] [CrossRef]
  27. Pietra, G.; Romagnani, C.; Moretta, L.; Mingari, M.C. HLA-E and HLA-E-bound peptides: Recognition by subsets of NK and T cells. Curr. Pharm. Des. 2009, 15, 3336–3344. [Google Scholar] [CrossRef]
  28. Michaelsson, J.; Teixeira de Matos, C.; Achour, A.; Lanier, L.L.; Karre, K.; Soderstrom, K. A signal peptide derived from hsp60 binds HLA-E and interferes with CD94/NKG2A recognition. J. Exp. Med. 2002, 196, 1403–1414. [Google Scholar] [CrossRef]
  29. Rolle, A.; Meyer, M.; Calderazzo, S.; Jager, D.; Momburg, F. Distinct HLA-E peptide complexes modify antibody-driven effector functions of adaptive NK cells. Cell Rep. 2018, 24, 1967–1976. [Google Scholar] [CrossRef]
  30. Hackmon, R.; Pinnaduwage, L.; Zhang, J.; Lye, S.J.; Geraghty, D.E.; Dunk, C.E. Definitive class I human leukocyte antigen expression in gestational placentation: HLA-F, HLA-E, HLA-C, and HLA-G in extravillous trophoblast invasion on placentation, pregnancy, and parturition. Am. J. Reprod. Immunol. 2017, 77, e12643. [Google Scholar] [CrossRef]
  31. Lima, T.H.A.; Buttura, R.V.; Donadi, E.A.; Veiga-Castelli, L.C. Mendes-Junior CT3, Castelli EC4. HLA-F coding and regulatory segments variability determined by massively parallel sequencing procedures in a Brazilian population sample. Hum. Immunol. 2016, 77, 841–853. [Google Scholar] [CrossRef]
  32. Sim, M.J.W.; Sun, P.D. HLA-F: A New kid licensed for peptide presentation. Immunity 2017, 46, 972–974. [Google Scholar] [CrossRef]
  33. Goodridge, J.P.; Burian, A.; Lee, N.; Geraghty, D.E. HLA-F complex without peptide binds to MHC class I protein in the open conformer form. J. Immunol. 2010, 184, 6199–6208. [Google Scholar] [CrossRef]
  34. Lepin, E.J.; Bastin, J.M.; Allan, D.S.; Roncador, G.; Braud, V.M.; Mason, D.Y.; van der Merwe, P.A.; McMichael, A.J.; Bell, J.I.; Powis, S.H. Functional characterization of HLA-F and binding of HLA-F tetramers to ILT2 and ILT4 receptors. Eur. J. Immunol. 2000, 30, 3552–3561. [Google Scholar] [CrossRef]
  35. Castelli, E.C.; Veiga-Castelli, L.C.; Yaghi, L.; Moreau, P.; Donadi, E.A. Transcriptional and posttranscriptional regulations of the HLA-G gene. J. Immunol. Res. 2014, 2014, 15. [Google Scholar] [CrossRef]
  36. Carosella, E.D.; Rouas-Freiss, N.; Roux, D.T.-L.; Moreau, P.; LeMaoult, J. HLA-G: An immune checkpoint molecule. In Advances in Immunology; Frederick, W.A., Ed.; Academic Press: Cambridge, MA, USA, 2015; Chapter Two; pp. 33–144. [Google Scholar]
  37. Donadi, E.A.; Castelli, E.C.; Arnaiz-Villena, A.; Roger, M.; Rey, D.; Moreau, P. Implications of the polymorphism of HLA-G on its function, regulation, evolution and disease association. Cell Mol. Life Sci. 2011, 68, 369–395. [Google Scholar] [CrossRef]
  38. Ishitani, A.; Geraghty, D.E. Alternative splicing of HLA-G transcripts yields proteins with primary structures resembling both class I and class II antigens. Proc. Natl. Acad. Sci. USA 1992, 89, 3947–3951. [Google Scholar] [CrossRef]
  39. Carosella, E.D.; Favier, B.; Rouas-Freiss, N.; Moreau, P.; Lemaoult, J. Beyond the increasing complexity of the immunomodulatory HLA-G molecule. Blood 2008, 111, 4862–4870. [Google Scholar] [CrossRef]
  40. Fujii, T.; Ishitani, A.; Geraghty, D.E. A soluble form of the HLA-G antigen is encoded by a messenger ribonucleic acid containing intron 4. J. Immunol. 1994, 153, 5516–5524. [Google Scholar]
  41. Sangrouber, D.; Marcou, C.; Le Discorde, M.; Chang, C.C.; Carosella, E.D.; Moreau, P. Cellular co-localization of intron-4 containing mRNA and HLA-G soluble protein in melanoma analyzed by fluorescence in situ hybridization. J. Immunol. Methods 2007, 326, 54–62. [Google Scholar] [CrossRef]
  42. Paul, P.; Cabestre, F.A.; Ibrahim, E.C.; Lefebvre, S.; Khalil-Daher, I.; Vazeux, G.; Quiles, R.M.; Bermond, F.; Dausset, J.; Carosella, E.D. Identification of HLA-G7 as a new splice variant of the HLA-G mRNA and expression of soluble HLA-G5, -G6, and -G7 transcripts in human transfected cells. Hum. Immunol. 2000, 61, 1138–1149. [Google Scholar] [CrossRef]
  43. Liepe, J.; Marino, F.; Sidney, J.; Jeko, A.; Bunting, D.E.; Sette, A.; Kloetzel, P.M.; Stumpf, M.P.; Heck, A.J.; Mishto, M. A large fraction of HLA-F class I ligands are proteasome-generated spliced peptides. Science 2016, 354, 354–358. [Google Scholar] [CrossRef]
  44. Mishto, M.; Liepe, J. Post-translational peptide splicing and T Cell responses. Trends Immunol. 2017, 38, 904–915. [Google Scholar] [CrossRef]
  45. Hunt, J.S.; Morales, P.J.; Pace, J.L.; Fazleabas, A.T.; Langat, D.K. A commentary on gestational programming and functions of HLA-G in pregnancy. Placenta 2007, 28, S57–S63. [Google Scholar] [CrossRef]
  46. Young, N.T.; Waller, E.C.; Patel, R.; Roghanian, A.; Austyn, J.M.; Trowsdale, J. The inhibitory receptor LILRB1 modulates the differentiation and regulatory potential of human dendritic cells. Blood 2008, 111, 3090–3096. [Google Scholar] [CrossRef]
  47. Rouas-Freiss, N.; Moreau, P.; Menier, C.; Carosella, E.D. HLA-G in cancer: A way to turn off the immune system. In Seminars in Cancer Biology; Elsevier: Amsterdam, The Netherlands, 2003. [Google Scholar]
  48. Braud, V.M.; Allan, D.S.; O’Callaghan, C.A.; Soderstrom, K.; D’Andrea, A.; Ogg, G.S.; Lazetic, S.; Young, N.T.; Bell, J.I.; Phillips, J.H.; et al. HLA-E binds to natural killer cell receptors CD94/NKG2A, B and C. Nature 1998, 391, 795–799. [Google Scholar] [CrossRef]
  49. Brostjan, C.; Sobanov, Y.; Glienke, J.; Hayer, S.; Lehrach, H.; Francis, F.; Hofer, E. The NKG2 natural killer cell receptor family: Comparative analysis of promoter sequences. Genes Immun. 2000, 1, 504–508. [Google Scholar] [CrossRef]
  50. Carretero, M.; Palmieri, G.; Llano, M.; Tullio, V.; Santoni, A.; Geraghty, D.E.; Lopez-Botet, M. Specific engagement of the CD94/NKG2-A killer inhibitory receptor by the HLA-E class Ib molecule induces SHP-1 phosphatase recruitment to tyrosine-phosphorylated NKG2-A: Evidence for receptor function in heterologous transfectants. Eur. J. Immunol. 1998, 28, 1280–1291. [Google Scholar] [CrossRef]
  51. Farag, S.S.; Fehniger, T.A.; Ruggeri, L.; Velardi, A.; Caligiuri, M.A. Natural killer cell receptors: New biology and insights into the graft-versus-leukemia effect. Blood 2002, 100, 1935–1947. [Google Scholar] [CrossRef]
  52. Petrie, E.J.; Clements, C.S.; Lin, J.; Sullivan, L.C.; Johnson, D.; Huyton, T.; Heroux, A.; Hoare, H.L.; Beddoe, T.; Reid, H.H.; et al. CD94-NKG2A recognition of human leukocyte antigen (HLA-F)-E bound to an HLA-F class I leader sequence. J. Exp. Med. 2008, 205, 725–735. [Google Scholar] [CrossRef]
  53. Lazetic, S.; Chang, C.; Houchins, J.P.; Lanier, L.L.; Phillips, J.H. Human natural killer cell receptors involved in MHC class I recognition are disulfide-linked heterodimers of CD94 and NKG2 subunits. J. Immunol. 1996, 157, 4741–4745. [Google Scholar]
  54. Campbell, K.S.; Colonna, M. DAP12: A key accessory protein for relaying signals by natural killer cell receptors. Int. J. Biochem. Cell Biol. 1999, 31, 631–636. [Google Scholar] [CrossRef]
  55. Rajagopalan, S.; Long, E. KIR2DL4 (CD158d): An activation receptor for HLA-G. Front. Immunol. 2012, 3, 258. [Google Scholar] [CrossRef]
  56. Lanier, L.L. Natural killer cell receptor signaling. Curr. Opin. Immunol. 2003, 15, 308–314. [Google Scholar] [CrossRef]
  57. Shwetank; Date, O.S.; Carbone, E.; Manjunath, R. Inhibition of ERK and proliferation in NK cell lines by soluble HLA-E released from Japanese encephalitis virus infected cells. Immunol. Lett. 2014, 162, 94–100. [Google Scholar] [CrossRef]
  58. Lauterbach, N.; Wieten, L.; Popeijus, H.E.; Voorter, C.E.; Tilanus, M.G. HLA-E regulates NKG2C+ natural killer cell function through presentation of a restricted peptide repertoire. Hum. Immunol. 2015, 76, 578–586. [Google Scholar] [CrossRef]
  59. Allan, D.S.; Lepin, E.J.; Braud, V.M.; O’Callaghan, C.A.; McMichael, A.J. Tetrameric complexes of HLA-E, HLA-F, and HLA-G. J. Immunol. Methods 2002, 268, 43–50. [Google Scholar] [CrossRef]
  60. Hirayasu, K.; Arase, H. Functional and genetic diversity of leukocyte immunoglobulin-like receptor and implication for disease associations. J. Hum. Genet. 2015, 60, 703–708. [Google Scholar] [CrossRef]
  61. Ketroussi, F.; Giuliani, M.; Bahri, R.; Azzarone, B.; Charpentier, B.; Durrbach, A. Lymphocyte cell-cycle inhibition by HLA-G is mediated by phosphatase SHP-2 and acts on the mTOR pathway. PLoS ONE 2011, 6, e22776. [Google Scholar] [CrossRef]
  62. Lesport, E.; Baudhuin, J.; Sousa, S.; LeMaoult, J.; Zamborlini, A.; Rouas-Freiss, N.; Carosella, E.D.; Favier, B. Inhibition of human gamma delta [corrected] T-cell antitumoral activity through HLA-G: Implications for immunotherapy of cancer. Cell Mol. Life Sci. 2011, 68, 3385–3399. [Google Scholar] [CrossRef]
  63. Thomas, R.; Matthias, T.; Witte, T. Leukocyte immunoglobulin-like receptors as new players in autoimmunity. Clin. Rev. Allerg. Immunol. 2010, 38, 159–162. [Google Scholar] [CrossRef]
  64. Morel, E.; Bellon, T. HLA-F class I molecules regulate IFN-gamma production induced in NK cells by target cells, viral products, or immature dendritic cells through the inhibitory receptor ILT2/CD85j. J. Immunol. 2008, 181, 2368–2381. [Google Scholar] [CrossRef]
  65. Cortesini, N.S.-F.; Colovai, A.I.; Manavalan, J.S.; Galluzzo, S.; Naiyer, A.J.; Liu, J.; Vlad, G.; Kim-Schulze, S.; Scotto, L.; Fan, J.; et al. Role of regulatory and suppressor T-cells in the induction of ILT3+ ILT4+ tolerogenic endothelial cells in organ allografts. Transplant Immunol. 2004, 13, 73–82. [Google Scholar] [CrossRef]
  66. Goodridge, J.P.; Burian, A.; Lee, N.; Geraghty, D.E. HLA-F and MHC class I open conformers are ligands for NK cell Ig-like receptors. J. Immunol. 2013, 191, 3553–3562. [Google Scholar] [CrossRef]
  67. Burian, A.; Wang, K.L.; Finton, K.A.; Lee, N.; Ishitani, A.; Strong, R.K.; Geraghty, D.E. HLA-F and MHC-I open conformers bind natural killer cell Ig-like receptor KIR3DS1. PLoS ONE 2016, 11, e0163297. [Google Scholar] [CrossRef]
  68. Garcia-Beltran, W.F.; Holzemer, A.; Martrus, G.; Chung, A.W.; Pacheco, Y.; Simoneau, C.R.; Rucevic, M.; Lamothe-Molina, P.A.; Pertel, T.; Kim, T.E.; et al. Open conformers of HLA-F are high-affinity ligands of the activating NK-cell receptor KIR3DS1. Nat. Immunol. 2016, 17, 1067–1074. [Google Scholar] [CrossRef]
  69. Moradi, S.; Berry, R.; Pymm, P.; Hitchen, C.; Beckham, S.A.; Wilce, M.C.J.; Walpole, N.G.; Clements, C.S.; Reid, H.H.; Perugini, M.A.; et al. The structure of the atypical killer cell immunoglobulin-like receptor, KIR2DL4. J. Biol. Chem. 2015, 290, 10460–10471. [Google Scholar] [CrossRef]
  70. Burshtyn, D.N.; Scharenberg, A.M.; Wagtmann, N.; Rajagopalan, S.; Berrada, K.; Yi, T.; Kinet, J.P.; Long, E.O. Recruitment of tyrosine phosphatase HCP by the killer cell inhibitor receptor. Immunity 1996, 4, 77–85. [Google Scholar] [CrossRef]
  71. Yusa, S.-I.; Catina, T.L.; Campbell, K.S. SHP-1- and phosphotyrosine-independent inhibitory signaling by a killer cell Ig-like receptor cytoplasmic domain in human NK cells. J. Immunol. 2002, 168, 5047–5057. [Google Scholar] [CrossRef]
  72. Lunemann, S.; Schobel, A.; Kah, J.; Fittje, P.; Holzemer, A.; Langeneckert, A.E.; Hess, L.U.; Poch, T.; Martrus, G.; Garcia-Beltran, W.F.; et al. Interactions between KIR3DS1 and HLA-F activate natural killer cells to control HCV replication in cell culture. Gastroenterology 2018, 155, 1366–1371. [Google Scholar] [CrossRef]
  73. Kiani, Z.; Dupuy, F.P.; Bruneau, J.; Lebouche, B.; Zhang, C.X.; Jackson, E.; Lisovsky, I.; da Fonseca, S.; Geraghty, D.E.; Bernard, N.F. HLA-F on HLA-null 721.221 cells activates primary NK cells expressing the activating killer Ig-like receptor KIR3DS1. J. Immunol. 2018, 201, 113–123. [Google Scholar] [CrossRef]
  74. Brown, D.; Trowsdale, J.; Allen, R. The LILR family: Modulators of innate and adaptive immune pathways in health and disease. Tissue Antigens 2004, 64, 215–225. [Google Scholar] [CrossRef]
  75. Gonen-Gross, T.; Achdout, H.; Arnon, T.I.; Gazit, R.; Stern, N.; Horejsi, V.; Goldman-Wohl, D.; Yagel, S.; Mandelboim, O. The CD85J/leukocyte inhibitory receptor-1 distinguishes between conformed and beta 2-microglobulin-free HLA-G molecules. J. Immunol. 2005, 175, 4866–4874. [Google Scholar] [CrossRef]
  76. Shiroishi, M.; Kuroki, K.; Ose, T.; Rasubala, L.; Shiratori, I.; Arase, H.; Tsumoto, K.; Kumagai, I.; Kohda, D.; Maenaka, K. Efficient leukocyte Ig-like receptor signaling and crystal structure of disulfide-linked HLA-G dimer. J. Biol. Chem. 2006, 281, 10439–10447. [Google Scholar] [CrossRef]
  77. Nardi Fda, S.; Konig, L.; Wagner, B.; Giebel, B.; Santos Manvailer, L.F.; Rebmann, V. Soluble monomers, dimers and HLA-G-expressing extracellular vesicles: The three dimensions of structural complexity to use HLA-G as a clinical biomarker. HLA 2016, 88, 77–86. [Google Scholar] [CrossRef]
  78. Chen, B.G.; Xu, D.P.; Lin, A.; Yan, W.H. NK cytolysis is dependent on the proportion of HLA-G expression. Hum. Immunol. 2013, 74, 286–289. [Google Scholar] [CrossRef]
  79. Lin, A.; Yan, W.H.; Xu, H.H.; Gan, M.F.; Cai, J.F.; Zhu, M.; Zhou, M.Y. HLA-G expression in human ovarian carcinoma counteracts NK cell function. Ann. Oncol. 2007, 18, 1804–1809. [Google Scholar] [CrossRef]
  80. Bainbridge, D.R.; Ellis, S.A.; Sargent, I.L. HLA-G suppresses proliferation of CD4 (+) T-lymphocytes. J. Reprod. Immunol. 2000, 48, 17–26. [Google Scholar] [CrossRef]
  81. Naji, A.; Menier, C.; Morandi, F.; Agaugue, S.; Maki, G.; Ferretti, E.; Bruel, S.; Pistoia, V.; Carosella, E.D.; Rouas-Freiss, N. Binding of HLA-G to ITIM-bearing Ig-like transcript 2 receptor suppresses B cell responses. J. Immunol. 2014, 192, 1536–1546. [Google Scholar] [CrossRef]
  82. Amodio, G.; Gregori, S. Human tolerogenic DC-10: Perspectives for clinical applications. Transpl. Res. 2012, 1, 14. [Google Scholar] [CrossRef]
  83. Gregori, S.; Tomasoni, D.; Pacciani, V.; Scirpoli, M.; Battaglia, M.; Magnani, C.F.; Hauben, E.; Roncarolo, M.G. Differentiation of type 1 T regulatory cells (Tr1) by tolerogenic DC-10 requires the IL-10-dependent ILT4/HLA-G pathway. Blood 2010, 116, 935–944. [Google Scholar] [CrossRef]
  84. Djurisic, S.; Skibsted, L.; Hviid, T.V.F. A phenotypic analysis of regulatory T cells and uterine NK Cells from first trimester pregnancies and associations with HLA-G. Am. J. Reprod. Immunol. 2015, 74, 427–444. [Google Scholar] [CrossRef]
  85. Goodridge, J.P.; Lathbury, L.J.; John, E.; Charles, A.K.; Christiansen, F.T.; Witt, C.S. The genotype of the NK cell receptor, KIR2DL4, influences INFγ secretion by decidual natural killer cells. Mol. Hum. Reprod. 2009, 15, 489–497. [Google Scholar] [CrossRef]
  86. Melsen, J.E.; Lugthart, G.; Lankester, A.C.; Schilham, M.W. Human circulating and tissue-resident CD56 (bright) natural killer cell populations. Front Immunol. 2016, 7, 262. [Google Scholar] [CrossRef]
  87. van der Meer, A.; Lukassen, H.G.; van Lierop, M.J.; Wijnands, F.; Mosselman, S.; Braat, D.D.; Joosten, I. Membrane-bound HLA-G activates proliferation and interferon-gamma production by uterine natural killer cells. Mol. Hum. Reprod. 2004, 10, 189–195. [Google Scholar] [CrossRef]
  88. Contini, P.; Ghio, M.; Poggi, A.; Filaci, G.; Indiveri, F.; Ferrone, S.; Puppo, F. Soluble HLA-F-A,-B,-C and -G molecules induce apoptosis in T and NK CD8+ cells and inhibit cytotoxic T cell activity through CD8 ligation. Eur. J. Immunol. 2003, 33, 125–134. [Google Scholar] [CrossRef]
  89. Le Bouteiller, P.; Tabiasco, J.; Polgar, B.; Kozma, N.; Giustiniani, J.; Siewiera, J.; Berrebi, A.; Aguerre-Girr, M.; Bensussan, A.; Jabrane-Ferrat, N. CD160: A unique activating NK cell receptor. Immunol. Lett. 2011, 138, 93–96. [Google Scholar] [CrossRef]
  90. Burton, G. Human implantation: Cell biology and immunology. J. Anat. 1997, 190, 473–475. [Google Scholar] [CrossRef]
  91. Fournel, S.; Aguerre-Girr, M.; Huc, X.; Lenfant, F.; Alam, A.; Toubert, A.; Bensussan, A.; Le Bouteiller, P. Cutting edge: Soluble HLA-G1 triggers CD95/CD95 ligand-mediated apoptosis in activated CD8+ cells by interacting with CD8. J. Immunol. 2000, 164, 6100–6104. [Google Scholar] [CrossRef]
  92. Fons, P.; Chabot, S.; Cartwright, J.E.; Lenfant, F.; L’Faqihi, F.; Giustiniani, J.; Herault, J.-P.; Gueguen, G.; Bono, F.; Savi, P.; et al. Soluble HLA-G1 inhibits angiogenesis through an apoptotic pathway and by direct binding to CD160 receptor expressed by endothelial cells. Blood 2006, 108, 2608–2615. [Google Scholar] [CrossRef]
  93. King, A.; Burrows, T.D.; Hiby, S.E.; Bowen, J.M.; Joseph, S.; Verma, S.; Lim, P.B.; Gardner, L.; Le Bouteiller, P.; Ziegler, A.; et al. Surface expression of HLA-C antigen by human extravillous trophoblast. Placenta 2000, 21, 376–387. [Google Scholar] [CrossRef]
  94. Sun, H.S.; Liu, D.X.; Bai, Y.Y.; Hu, N.W. Disease-association of different killer cell immunoglobulin-like receptors (KIR) and HLA-C gene combinations in reactive arthritis. Mod. Rheumatol. 2018, 180, 1–7. [Google Scholar] [CrossRef]
  95. Colonna, M.; Samaridis, J. Cloning of immunoglobulin-superfamily members associated with HLA-C and HLA-F-B recognition by human natural killer cells. Science 1995, 268, 405–408. [Google Scholar] [CrossRef]
  96. Male, V.; Sharkey, A.; Masters, L.; Kennedy, P.R.; Farrell, L.E.; Moffett, A. The effect of pregnancy on the uterine NK cell KIR repertoire. Eur. J. Immunol. 2011, 41, 3017–3027. [Google Scholar] [CrossRef]
  97. Sharkey, A.M.; Gardner, L.; Hiby, S.; Farrell, L.; Apps, R.; Masters, L.; Goodridge, J.; Lathbury, L.; Stewart, C.A.; Verma, S.; et al. Killer Ig-like receptor expression in uterine NK cells is biased toward recognition of HLA-C and alters with gestational age. J. Immunol. 2008, 181, 39–46. [Google Scholar] [CrossRef]
  98. Faas, M.M.; de Vos, P. Uterine NK cells and macrophages in pregnancy. Placenta 2017, 56, 44–52. [Google Scholar] [CrossRef]
  99. Amodio, G.; Mugione, A.; Sanchez, A.M.; Vigano, P.; Candiani, M.; Somigliana, E.; Roncarolo, M.G.; Panina-Bordignon, P.; Gregori, S. HLA-G expressing DC-10 and CD4 (+) T cells accumulate in human decidua during pregnancy. Hum. Immunol. 2013, 74, 406–411. [Google Scholar] [CrossRef]
  100. Lash, G.E.; Pitman, H.; Morgan, H.L.; Innes, B.A.; Agwu, C.N.; Bulmer, J.N. Decidual macrophages: Key regulators of vascular remodeling in human pregnancy. J. Leukoc. Biol. 2016, 100, 315–325. [Google Scholar] [CrossRef]
  101. Lila, N.; Rouas-Freiss, N.; Dausset, J.; Carpentier, A.; Carosella, E.D. Soluble HLA-G protein secreted by allo-specific CD4+ T cells suppresses the allo-proliferative response: A CD4+ T cell regulatory mechanism. Proc. Natl. Acad. Sci. USA 2001, 98, 12150–12155. [Google Scholar] [CrossRef]
  102. Persson, G.; Melsted, W.N.; Nilsson, L.L.; Hviid, T.V.F. HLA class Ib in pregnancy and pregnancy-related disorders. Immunogenetics 2017, 69, 581–595. [Google Scholar] [CrossRef]
  103. Rizzo, R.; Vercammen, M.; van de Velde, H.; Horn, P.A.; Rebmann, V. The importance of HLA-G expression in embryos, trophoblast cells, and embryonic stem cells. Cell Mol. Life Sci. 2011, 68, 341–352. [Google Scholar] [CrossRef]
  104. Steinborn, A.; Varkonyi, T.; Scharf, A.; Bahlmann, F.; Klee, A.; Sohn, C. Early detection of decreased soluble HLA-G levels in the maternal circulation predicts the occurrence of preeclampsia and intrauterine growth retardation during further course of pregnancy. Am. J. Reprod. Immunol. 2007, 57, 277–286. [Google Scholar] [CrossRef]
  105. Drukker, M.; Katz, G.; Urbach, A.; Schuldiner, M.; Markel, G.; Itskovitz-Eldor, J.; Reubinoff, B.; Mandelboim, O.; Benvenisty, N. Characterization of the expression of MHC proteins in human embryonic stem cells. Proc. Natl. Acad. Sci. USA 2002, 99, 9864–9869. [Google Scholar] [CrossRef]
  106. Fuzzi, B.; Rizzo, R.; Criscuoli, L.; Noci, I.; Melchiorri, L.; Scarselli, B.; Bencini, E.; Menicucci, A.; Baricordi, O.R. HLA-G expression in early embryos is a fundamental prerequisite for the obtainment of pregnancy. Eur. J. Immunol. 2002, 32, 311–315. [Google Scholar] [CrossRef]
  107. Verloes, A.; Van de Velde, H.; LeMaoult, J.; Mateizel, I.; Cauffman, G.; Horn, P.A.; Carosella, E.D.; Devroey, P.; De Waele, M.; Rebmann, V.; et al. HLA-G expression in human embryonic stem cells and preimplantation embryos. J. Immunol. 2011, 186, 2663–2671. [Google Scholar] [CrossRef]
  108. Chen, H.; Zhang, N.; Li, T.; Guo, J.; Wang, Z.; Yang, M.; Gao, L. Human umbilical cord Wharton’s jelly stem cells: Immune property genes assay and effect of transplantation on the immune cells of heart failure patients. Cell Immunol. 2012, 276, 83–90. [Google Scholar] [CrossRef]
  109. Giacomini, E.; Vago, R.; Sanchez, A.M.; Podini, P.; Zarovni, N.; Murdica, V.; Rizzo, R.; Bortolotti, D.; Candiani, M.; Vigano, P. Secretome of in vitro cultured human embryos contains extracellular vesicles that are uptaken by the maternal side. Sci. Rep. 2017, 7, 5210. [Google Scholar] [CrossRef]
  110. Noci, I.; Fuzzi, B.; Rizzo, R.; Melchiorri, L.; Criscuoli, L.; Dabizzi, S.; Biagiotti, R.; Pellegrini, S.; Menicucci, A.; Baricordi, O.R. Embryonic soluble HLA-G as a marker of developmental potential in embryos. Hum. Reprod. 2005, 20, 138–146. [Google Scholar] [CrossRef]
  111. Jabeen, A.; Miranda-Sayago, J.M.; Obara, B.; Spencer, P.S.; Dealtry, G.B.; Hayrabedyan, S.; Shaikly, V.; Laissue, P.P.; Fernández, N. Quantified colocalization reveals heterotypic histocompatibility class I antigen associations on trophoblast cell membranes: Relevance for human pregnancy. Biol. Reprod. 2013, 89, 1–10. [Google Scholar] [CrossRef]
  112. Sher, G.; Keskintepe, L.; Nouriani, M.; Roussev, R.; Batzofin, J. Expression of sHLA-G in supernatants of individually cultured 46-h embryos: A potentially valuable indicator of ‘embryo competency’ and IVF outcome. Reprod. Biomed. Online 2004, 9, 74–78. [Google Scholar] [CrossRef]
  113. Tabiasco, J.; Perrier d’Hauterive, S.; Thonon, F.; Parinaud, J.; Leandri, R.; Foidart, J.M.; Chaouat, G.; Munaut, C.; Lombroso, R.; Selva, J.; et al. Soluble HLA-G in IVF/ICSI embryo culture supernatants does not always predict implantation success: A multicentre study. Reprod. Biomed. Online 2009, 18, 374–381. [Google Scholar] [CrossRef]
  114. Niu, Z.; Wang, L.; Pang, R.T.K.; Guo, Y.; Yeung, W.S.B.; Yao, Y. A meta-analysis of the impact of human leukocyte antigen-G on the outcomes of IVF/ICSI. Reprod. Biomed. Online 2017, 34, 611–618. [Google Scholar] [CrossRef]
  115. Mosaferi, E.; Majidi, J.; Mohammadian, M.; Babaloo, Z.; Monfaredan, A.; Baradaran, B. HLA-G expression pattern: Reliable assessment for pregnancy outcome prediction. Adv. Pharm. Bull. 2013, 3, 443–446. [Google Scholar]
  116. Koc, A.; Kirbiyik, O.; Kutbay, Y.B.; Ozyilmaz, B.; Ozdemir, T.R.; Kaya, O.O.; Kubat, G.; Koc, Z.P. Fetal HLA-G alleles and their effect on miscarriage. Adv. Clin. Exp. Med. 2018, 27, 1233–1237. [Google Scholar] [CrossRef]
  117. Jassem, R.M.; Shani, W.S.; Loisel, D.A.; Sharief, M.; Billstrand, C.; Ober, C. HLA-G polymorphisms and soluble HLA-G protein levels in women with recurrent pregnancy loss from Basrah province in Iraq. Hum. Immunol. 2012, 73, 811–817. [Google Scholar] [CrossRef]
  118. Abediankenari, S.; Farzad, F.; Rahmani, Z.; Hashemi-Soteh, M.B. HLA-G5 and G7 isoforms in pregnant women. Iran J. Allerg. Asthma Immunol. 2015, 14, 217–221. [Google Scholar]
  119. Zidi, I.; Laaribi, A.B.; Bortolotti, D.; Belhadj, M.; Mehri, A.; Yahia, H.B.; Babay, W.; Chaouch, H.; Zidi, N.; Letaief, A.; et al. HLA-E polymorphism and soluble HLA-E plasma levels in chronic hepatitis B patients. HLA-F 2016, 87, 153–159. [Google Scholar] [CrossRef]
  120. Enghelabifar, M.; Allafan, S.; Khayatzadeh, J.; Shahrokh Abadi, K.; Hasanzadeh Nazarabadi, M.; Moradi, F.; Musavifar, N.; Jalali, M.; Mojarrad, M. Association of the maternal 14-bp insertion/deletion polymorphism in the histocompatibility leukocyte antigen G gene with recurrent implantation failure. Iran J. Reprod. Med. 2014, 12, 641–646. [Google Scholar]
  121. Hashemi, M.; Mokhtari, M.; Khazaeian, S.; Bahari, G.; Rezaei, M.; Nakhaee, A.; Taheri, M. Evaluation of HLA-G 14-bp ins/del and +3142G>C polymorphisms with susceptibility to recurrent spontaneous abortion. Taiwan J. Obstet. Gynecol. 2017, 56, 276–280. [Google Scholar] [CrossRef]
  122. Amodio, G.; Canti, V.; Maggio, L.; Rosa, S.; Castiglioni, M.T.; Rovere-Querini, P.; Gregori, S. Association of genetic variants in the 3′UTR of HLA-G with recurrent pregnancy loss. Hum. Immunol. 2016, 77, 886–891. [Google Scholar] [CrossRef]
  123. Michita, R.T.; Zambra, F.M.B.; Fraga, L.R.; Sanseverino, M.T.V.; Callegari-Jacques, S.M.; Vianna, P.; Chies, J.A.B. A tug-of-war between tolerance and rejection—New evidence for 3′UTR HLA-G haplotypes influence in recurrent pregnancy loss. Hum. Immunol. 2016, 77, 892–897. [Google Scholar] [CrossRef]
  124. Fan, W.; Huang, Z.; Li, S.; Xiao, Z. The HLA-G 14-bp polymorphism and recurrent implantation failure: A meta-analysis. J. Assist. Reprod. Genet. 2017, 34, 1559–1565. [Google Scholar] [CrossRef]
  125. Aldrich, C.L.; Stephenson, M.D.; Karrison, T.; Odem, R.R.; Branch, D.W.; Scott, J.R.; Schreiber, J.R.; Ober, C. HLA-G genotypes and pregnancy outcome in couples with unexplained recurrent miscarriage. Mol. Hum. Reprod. 2001, 7, 1167–1172. [Google Scholar] [CrossRef]
  126. Kuroshli, Z.; Gourabi, H.; Bazrgar, M.; Sanati, M.; Zamani Esteki, M. The relationship between HLA-G gene polymorphisms and repeated implantation failure in infertile couples undergoing assisted reproductive technique. Iran J. Allerg. Asthma Immunol. 2015, 14, 535–542. [Google Scholar]
  127. Fotoohi, M.; Ghasemi, N.; Mirghanizadeh, S.A.; Vakili, M.; Samadi, M. Association between HLA-E gene polymorphism and unexplained recurrent spontaneous abortion (RSA) in Iranian women. Int. J. Reprod. Biomed. 2016, 14, 477–482. [Google Scholar] [CrossRef]
  128. Lin, A.; Yan, W.-H. Heterogeneity of HLA-G expression in cancers: Facing the challenges. Front. Immunol. 2018, 9, 2164. [Google Scholar] [CrossRef]
  129. Amiot, L.; Ferrone, S.; Grosse-Wilde, H.; Seliger, B. Biology of HLA-G in cancer: A candidate molecule for therapeutic intervention? Cell Mol. Life Sci. 2011, 68, 417–431. [Google Scholar] [CrossRef]
  130. Jeong, S.; Park, S.; Park, B.W.; Park, Y.; Kwon, O.J.; Kim, H.S. Human leukocyte antigen-G (HLA-G) polymorphism and expression in breast cancer patients. PLoS ONE 2014, 9, e98284. [Google Scholar] [CrossRef]
  131. Rolfsen, G.B.; Castelli, E.C.; Donadi, E.A.; Duarte, R.A.; Soares, C.P. HLA-G polymorphism and breast cancer. Int. J. Immunogenet. 2014, 41, 143–148. [Google Scholar] [CrossRef]
  132. da Silva, G.B.; Silva, T.G.; Duarte, R.A.; Neto, N.L.; Carrara, H.H.; Donadi, E.A.; Goncalves, M.A.; Soares, E.G.; Soares, C.P. Expression of the classical and nonclassical HLA-F molecules in breast cancer. Int. J. Breast Cancer 2013, 2013, 250435. [Google Scholar] [CrossRef]
  133. He, X.; Dong, D.-D.; Yie, S.-M.; Yang, H.; Cao, M.; Ye, S.-R.; Li, K.; Liu, J.; Chen, J. HLA-G expression in human breast cancer: Implications for diagnosis and prognosis, and effect on allocytotoxic lymphocyte response after hormone treatment in vitro. Ann. Surg. Oncol. 2010, 17, 1459–1469. [Google Scholar] [CrossRef]
  134. Kleinberg, L.; Florenes, V.A.; Skrede, M.; Dong, H.P.; Nielsen, S.; McMaster, M.T.; Nesland, J.M.; Shih Ie, M.; Davidson, B. Expression of HLA-G in malignant mesothelioma and clinically aggressive breast carcinoma. Virchows Arch. 2006, 449, 31–39. [Google Scholar] [CrossRef]
  135. Palmisano, G.L.; Pistillo, M.P.; Fardin, P.; Capanni, P.; Nicolo, G.; Salvi, S.; Spina, B.; Pasciucco, G.; Ferrara, G.B. Analysis of HLA-G expression in breast cancer tissues. Hum. Immunol. 2002, 63, 969–976. [Google Scholar] [CrossRef]
  136. Ferguson, R.; Ramanakumar, A.V.; Koushik, A.; Coutlee, F.; Franco, E.; Roger, M. Human leukocyte antigen G polymorphism is associated with an increased risk of invasive cancer of the uterine cervix. Int. J. Cancer 2012, 131, E312–E319. [Google Scholar] [CrossRef]
  137. Singer, G.; Rebmann, V.; Chen, Y.C.; Liu, H.T.; Ali, S.Z.; Reinsberg, J.; McMaster, M.T.; Pfeiffer, K.; Chan, D.W.; Wardelmann, E.; et al. HLA-G is a potential tumor marker in malignant ascites. Clin. Cancer Res. 2003, 9, 4460–4464. [Google Scholar]
  138. Zhang, X.; Han, Q.Y.; Li, J.B.; Ruan, Y.Y.; Yan, W.H.; Lin, A. Lesion HLA-G5/-G6 isoforms expression in patients with ovarian cancer. Hum. Immunol. 2016, 77, 780–784. [Google Scholar] [CrossRef]
  139. Rutten, M.J.; Dijk, F.; Savci-Heijink, C.D.; Buist, M.R.; Kenter, G.G.; van de Vijver, M.J.; Jordanova, E.S. HLA-G expression is an independent predictor for improved survival in high grade ovarian carcinomas. J. Immunol. Res. 2014, 2014, 274584. [Google Scholar] [CrossRef]
  140. Lin, A.; Xu, H.H.; Xu, D.P.; Zhang, X.; Wang, Q.; Yan, W.H. Multiple steps of HLA-G in ovarian carcinoma metastasis: Alter NK cytotoxicity and induce matrix metalloproteinase-15 (MMP-15) expression. Hum. Immunol. 2013, 74, 439–446. [Google Scholar] [CrossRef]
  141. Bijen, C.B.; Bantema-Joppe, E.J.; de Jong, R.A.; Leffers, N.; Mourits, M.J.; Eggink, H.F.; van der Zee, A.G.; Hollema, H.; de Bock, G.H.; Nijman, H.W. The prognostic role of classical and nonclassical MHC class I expression in endometrial cancer. Int. J. Cancer 2010, 126, 1417–1427. [Google Scholar] [CrossRef]
  142. Gimenes, F.; Teixeira, J.J.; de Abreu, A.L.; Souza, R.P.; Pereira, M.W.; da Silva, V.R.; Boer, C.G.; Maria-Engler, S.S.; Bonini, M.G.; Borelli, S.D.; et al. Human leukocyte antigen (HLA-F)-G and cervical cancer immunoediting: A candidate molecule for therapeutic intervention and prognostic biomarker? Biochim. Biophys. Acta 2014, 1846, 576–589. [Google Scholar] [CrossRef]
  143. Li, X.J.; Zhang, X.; Lin, A.; Ruan, Y.Y.; Yan, W.H. Human leukocyte antigen-G (HLA-G) expression in cervical cancer lesions is associated with disease progression. Hum. Immunol. 2012, 73, 946–949. [Google Scholar] [CrossRef]
  144. Rodriguez, J.A.; Galeano, L.; Palacios, D.M.; Gomez, C.; Serrano, M.L.; Bravo, M.M.; Combita, A.L. Altered HLA-F class I and HLA-G expression is associated with IL-10 expression in patients with cervical cancer. Pathobiology 2012, 79, 72–83. [Google Scholar] [CrossRef]
  145. Zheng, N.; Wang, C.X.; Zhang, X.; Du, L.T.; Zhang, J.; Kan, S.F.; Zhu, C.B.; Dong, Z.G.; Wang, L.L.; Wang, S.; et al. Up-regulation of HLA-G expression in cervical premalignant and malignant lesions. Tissue Antigens 2011, 77, 218–224. [Google Scholar] [CrossRef]
  146. Langat, D.K.; Sue Platt, J.; Tawfik, O.; Fazleabas, A.T.; Hunt, J.S. Differential expression of human leukocyte antigen-G (HLA-G) messenger RNAs and proteins in normal human prostate and prostatic adenocarcinoma. J. Reprod. Immunol. 2006, 71, 75–86. [Google Scholar] [CrossRef]
  147. Montilla, D.; Perez, M.; Borges, L.; Bianchi, G.; Cova, J.A. Soluble human leukocyte antigen-G in the bronchoalveolar lavage of lung cancer patients. Arch. Bronconeumol. 2016, 52, 420–424. [Google Scholar] [CrossRef]
  148. Urosevic, M.; Kurrer, M.O.; Kamarashev, J.; Mueller, B.; Weder, W.; Burg, G.; Stahel, R.A.; Dummer, R.; Trojan, A. Human leukocyte antigen G up-regulation in lung cancer associates with high-grade histology, human leukocyte antigen class I loss and interleukin-10 production. Am. J. Pathol. 2001, 159, 817–824. [Google Scholar] [CrossRef]
  149. Wisniewski, A.; Kowal, A.; Wyrodek, E.; Nowak, I.; Majorczyk, E.; Wagner, M.; Pawlak-Adamska, E.; Jankowska, R.; Slesak, B.; Frydecka, I.; et al. Genetic polymorphisms and expression of HLA-G and its receptors, KIR2DL4 and LILRB1, in non-small cell lung cancer. Tissue Antigens 2015, 85, 466–475. [Google Scholar] [CrossRef]
  150. Yan, W.H.; Liu, D.; Lu, H.Y.; Li, Y.Y.; Zhang, X.; Lin, A. Significance of tumour cell HLA-G5/-G6 isoform expression in discrimination for adenocarcinoma from squamous cell carcinoma in lung cancer patients. J. Cell Mol. Med. 2015, 19, 778–785. [Google Scholar] [CrossRef]
  151. Yie, S.M.; Yang, H.; Ye, S.R.; Li, K.; Dong, D.D.; Lin, X.M. Expression of human leucocyte antigen G (HLA-G) is associated with prognosis in non-small cell lung cancer. Lung Cancer 2007, 58, 267–274. [Google Scholar] [CrossRef]
  152. Zhang, Y.; Zhao, J.; Qiu, L.; Zhang, P.; Li, J.; Yang, D.; Wei, X.; Han, Y.; Nie, S.; Sun, Y. Co-expression of ILT4/HLA-G in human non-small cell lung cancer correlates with poor prognosis and ILT4-HLA-G interaction activates ERK signaling. Tumour. Biol. 2016, 37, 11187–11198. [Google Scholar] [CrossRef]
  153. Carosella, E.D.; Ploussard, G.; LeMaoult, J.; Desgrandchamps, F. A systematic review of immunotherapy in urologic cancer: Evolving roles for targeting of CTLA-4, PD-1/PD-L1, and HLA-G. Eur. Urol. 2015, 68, 267–279. [Google Scholar] [CrossRef]
  154. Castelli, E.C.; Mendes-Junior, C.T.; Viana de Camargo, J.L.; Donadi, E.A. HLA-G polymorphism and transitional cell carcinoma of the bladder in a Brazilian population. Tissue Antigens 2008, 72, 149–157. [Google Scholar] [CrossRef]
  155. El-Chennawi, F.A.; Auf, F.A.; El-Diasty, A.M.; El-Daim, M.A.; El-Sherbiny, S.M.; Ali, A.; El-Baz, M.; El-Hameed, M.A.; Paul, P.; Ibrahim, E.C.; et al. Expression of HLA-G in cancer bladder. Egypt J. Immunol. 2005, 12, 57–64. [Google Scholar]
  156. Gan, L.H.; Huang, L.F.; Zhang, X.; Lin, A.; Xu, D.P.; Wang, Q.; Wang, T.J.; Yan, W.H. Tumor-specific upregulation of human leukocyte antigen-G expression in bladder transitional cell carcinoma. Hum. Immunol. 2010, 71, 899–904. [Google Scholar] [CrossRef]
  157. Swets, M.; Konig, M.H.; Zaalberg, A.; Dekker-Ensink, N.G.; Gelderblom, H.; van de Velde, C.J.; van den Elsen, P.J.; Kuppen, P.J. HLA-G and classical HLA-F class I expression in primary colorectal cancer and associated liver metastases. Hum. Immunol. 2016, 77, 773–779. [Google Scholar] [CrossRef]
  158. Guo, Z.Y.; Lv, Y.G.; Wang, L.; Shi, S.J.; Yang, F.; Zheng, G.X.; Wen, W.H.; Yang, A.G. Predictive value of HLA-G and HLA-E in the prognosis of colorectal cancer patients. Cell Immunol. 2015, 293, 10–16. [Google Scholar] [CrossRef]
  159. Reimers, M.S.; Engels, C.C.; Putter, H.; Morreau, H.; Liefers, G.J.; van de Velde, C.J.; Kuppen, P.J. Prognostic value of HLA class I, HLA-E, HLA-G and tregs in rectal cancer: A retrospective cohort study. BMC Cancer 2014, 14, 486. [Google Scholar] [CrossRef]
  160. Zeestraten, E.C.; Reimers, M.S.; Saadatmand, S.; Goossens-Beumer, I.J.; Dekker, J.W.; Liefers, G.J.; van den Elsen, P.J.; van de Velde, C.J.; Kuppen, P.J. Combined analysis of HLA-F class I, HLA-E and HLA-G predicts prognosis in colon cancer patients. Br. J. Cancer 2014, 110, 459–468. [Google Scholar] [CrossRef]
  161. Dardano, A.; Rizzo, R.; Polini, A.; Stignani, M.; Tognini, S.; Pasqualetti, G.; Ursino, S.; Colato, C.; Ferdeghini, M.; Baricordi, O.R.; et al. Soluble human leukocyte antigen-G and its insertion/deletion polymorphism in papillary thyroid carcinoma: Novel potential biomarkers of disease? J. Clin. Endocrinol. Metab. 2012, 97, 4080–4086. [Google Scholar] [CrossRef]
  162. Nunes, L.M.; Ayres, F.M.; Francescantonio, I.C.; Saddi, V.A.; Avelino, M.A.; Alencar Rde, C.; Silva, R.C.; Meneghini, A.J.; Wastowski, I.J. Association between the HLA-G molecule and lymph node metastasis in papillary thyroid cancer. Hum. Immunol. 2013, 74, 447–451. [Google Scholar] [CrossRef]
  163. Cao, M.; Yie, S.M.; Liu, J.; Ye, S.R.; Xia, D.; Gao, E. Plasma soluble HLA-G is a potential biomarker for diagnosis of colorectal, gastric, esophageal and lung cancer. Tissue Antigens 2011, 78, 120–128. [Google Scholar] [CrossRef]
  164. Lin, A.; Zhang, X.; Zhou, W.J.; Ruan, Y.Y.; Xu, D.P.; Wang, Q.; Yan, W.H. Human leukocyte antigen-G expression is associated with a poor prognosis in patients with esophageal squamous cell carcinoma. Int. J. Cancer 2011, 129, 1382–1390. [Google Scholar] [CrossRef]
  165. Degenhardt, Y.; Huang, J.; Greshock, J.; Horiates, G.; Nathanson, K.; Yang, X.; Herlyn, M.; Weber, B. Distinct MHC gene expression patterns during progression of melanoma. Genes Chromosom. Cancer 2010, 49, 144–154. [Google Scholar]
  166. Bezuhly, M.; Howlett, A.; Colp, P.; Conrad, D.M.; Walsh, N.; Rowden, G.; Morris, S.F.; Langley, R.G. Quantitative HLA-G expression in metastasising and non-metastasising primary thin cutaneous melanomas. Dermatology 2008, 217, 281–283. [Google Scholar] [CrossRef]
  167. Paul, P.; Cabestre, F.A.; Le Gal, F.A.; Khalil-Daher, I.; Le Danff, C.; Schmid, M.; Mercier, S.; Avril, M.F.; Dausset, J.; Guillet, J.G.; et al. Heterogeneity of HLA-G gene transcription and protein expression in malignant melanoma biopsies. Cancer Res. 1999, 59, 1954–1960. [Google Scholar]
  168. Paul, P.; Rouas-Freiss, N.; Khalil-Daher, I.; Moreau, P.; Riteau, B.; Le Gal, F.A.; Avril, M.F.; Dausset, J.; Guillet, J.G.; Carosella, E.D. HLA-G expression in melanoma: A way for tumor cells to escape from immunosurveillance. Proc. Natl. Acad. Sci. USA 1998, 95, 4510–4515. [Google Scholar] [CrossRef]
  169. Urosevic, M.; Willers, J.; Mueller, B.; Kempf, W.; Burg, G.; Dummer, R. HLA-G protein up-regulation in primary cutaneous lymphomas is associated with interleukin-10 expression in large cell T-cell lymphomas and indolent B-cell lymphomas. Blood 2002, 99, 609–617. [Google Scholar] [CrossRef]
  170. Diepstra, A.; Poppema, S.; Boot, M.; Visser, L.; Nolte, I.M.; Niens, M.; Te Meerman, G.J.; van den Berg, A. HLA-G protein expression as a potential immune escape mechanism in classical Hodgkin’s lymphoma. Tissue Antigens 2008, 71, 219–226. [Google Scholar] [CrossRef]
  171. Tronik-Le Roux, D.; Renard, J.; Verine, J.; Renault, V.; Tubacher, E.; LeMaoult, J.; Rouas-Freiss, N.; Deleuze, J.F.; Desgrandschamps, F.; Carosella, E.D. Novel landscape of HLA-G isoforms expressed in clear cell renal cell carcinoma patients. Mol. Oncol. 2017, 11, 1561–1578. [Google Scholar] [CrossRef]
  172. Ibrahim, E.C.; Guerra, N.; Lacombe, M.J.; Angevin, E.; Chouaib, S.; Carosella, E.D.; Caignard, A.; Paul, P. Tumor-specific up-regulation of the nonclassical class I HLA-G antigen expression in renal carcinoma. Cancer Res. 2001, 61, 6838–6845. [Google Scholar]
  173. Hanak, L.; Slaby, O.; Lauerova, L.; Kren, L.; Nenutil, R.; Michalek, J. Expression pattern of HLA-F class I antigens in renal cell carcinoma and primary cell line cultures: Methodological implications for immunotherapy. Med. Sci. Monit. 2009, 15, CR638–CR643. [Google Scholar]
  174. Kren, L.; Muckova, K.; Lzicarova, E.; Sova, M.; Vybihal, V.; Svoboda, T.; Fadrus, P.; Smrcka, M.; Slaby, O.; Lakomy, R.; et al. Production of immune-modulatory nonclassical molecules HLA-G and HLA-E by tumor infiltrating ameboid microglia/macrophages in glioblastomas: A role in innate immunity? J. Neuroimmunol. 2010, 220, 131–135. [Google Scholar] [CrossRef]
  175. Kren, L.; Slaby, O.; Muckova, K.; Lzicarova, E.; Sova, M.; Vybihal, V.; Svoboda, T.; Fadrus, P.; Lakomy, R.; Vanhara, P.; et al. Human leukocyte antigen-G is frequently expressed in glioblastoma and may be induced in vitro by combined 5-aza-2′-deoxycytidine and interferon-gamma treatments: Results from a multicentric study. Am. J. Pathol. 2013, 182, 540–552. [Google Scholar]
  176. Wastowski, I.J.; Simoes, R.T.; Yaghi, L.; Donadi, E.A.; Pancoto, J.T.; Poras, I.; Lechapt-Zalcman, E.; Bernaudin, M.; Valable, S.; Carlotti, C.G., Jr.; et al. Human leukocyte antigen-G is frequently expressed in glioblastoma and may be induced in vitro by combined 5-aza-2′-deoxycytidine and interferon-gamma treatments: Results from a multicentric study. Am. J. Pathol. 2013, 182, 540–552. [Google Scholar] [CrossRef]
  177. Wiendl, H.; Mitsdoerffer, M.; Hofmeister, V.; Wischhusen, J.; Bornemann, A.; Meyermann, R.; Weiss, E.H.; Melms, A.; Weller, M. A functional role of HLA-G expression in human gliomas: An alternative strategy of immune escape. J. Immunol. 2002, 168, 4772–4780. [Google Scholar] [CrossRef]
  178. Wischhusen, J.; Waschbisch, A.; Wiendl, H. Immune-refractory cancers and their little helpers—An extended role for immunetolerogenic MHC molecules HLA-G and HLA-E? Semin. Cancer Biol. 2007, 17, 459–468. [Google Scholar] [CrossRef]
  179. Karagoz, B.; Haholu, A.; Ozgun, A.; Bilgi, O.; Tuncel, T.; Emirzeoglu, L.; Celik, S.; Demirel, D. HLA-G in testicular germ cell tumors. Oncol. Res. Treat. 2014, 37, 245–248. [Google Scholar] [CrossRef]
  180. Zhou, L.; Niu, Z.Y.; Liang, Z.Y.; Zhou, W.X.; You, L.; Wang, M.Y.; Yao, L.T.; Liao, Q.; Zhao, Y.P. HLA-G impairs host immune response and predicts poor prognosis in pancreatic cancer. Am. J. Transl. Res. 2015, 7, 2036–2044. [Google Scholar]
  181. Sheu, J.; Shih Ie, M. HLA-G and immune evasion in cancer cells. J. Formos. Med. Assoc. 2010, 109, 248–257. [Google Scholar] [CrossRef]
  182. Bossard, C.; Bézieau, S.; Matysiak-Budnik, T.; Volteau, C.; Laboisse, C.L.; Jotereau, F.; Mosnier, J.-F. HLA-E/β2 microglobulin overexpression in colorectal cancer is associated with recruitment of inhibitory immune cells and tumor progression. Int. J. Cancer 2012, 131, 855–863. [Google Scholar] [CrossRef]
  183. Curigliano, G.; Criscitiello, C.; Gelao, L.; Goldhirsch, A. Molecular pathways: Human leukocyte antigen G (HLA-G). Clin. Cancer Res. 2013, 19, 5564–5571. [Google Scholar] [CrossRef]
  184. Xu, Y.; Han, H.; Zhang, F.; Lv, S.; Li, Z.; Fang, Z. Lesion human leukocyte antigen-F expression is associated with a poor prognosis in patients with hepatocellular carcinoma. Oncol. Lett. 2015, 9, 300–304. [Google Scholar] [CrossRef]
  185. Zhang, X.; Lin, A.; Zhang, J.G.; Bao, W.G.; Xu, D.P.; Ruan, Y.Y.; Yan, W.H. Alteration of HLA-F and HLA-F I antigen expression in the tumor is associated with survival in patients with esophageal squamous cell carcinoma. Int. J. Cancer 2013, 132, 82–89. [Google Scholar] [CrossRef]
  186. Lin, A.; Zhang, X.; Ruan, Y.Y.; Wang, Q.; Zhou, W.J.; Yan, W.H. HLA-F expression is a prognostic factor in patients with non-small-cell lung cancer. Lung Cancer 2011, 74, 504–509. [Google Scholar] [CrossRef]
  187. Wan, R.; Wang, Z.W.; Li, H.; Peng, X.D.; Liu, G.Y.; Ou, J.M.; Cheng, A.Q. Human leukocyte antigen-g inhibits the anti-tumor effect of natural killer cells via immunoglobulin-like transcript 2 in gastric cancer. Cell Physiol. Biochem. 2017, 44, 1828–1841. [Google Scholar] [CrossRef]
  188. Ueshima, C.; Kataoka, T.R.; Hirata, M.; Furuhata, A.; Suzuki, E.; Toi, M.; Tsuruyama, T.; Okayama, Y.; Haga, H.; Ueshima, C.; et al. The killer cell Ig-like receptor 2DL4 expression in human mast cells and its potential role in breast cancer invasion. Cancer Immunol. Res. 2015, 3, 871–880. [Google Scholar] [CrossRef]
  189. LeMaoult, J.; Caumartin, J.; Daouya, M.; Favier, B.; Le Rond, S.; Gonzalez, A.; Carosella, E.D. Immune regulation by pretenders: Cell-to-cell transfers of HLA-G make effector T cells act as regulatory cells. Blood 2007, 109, 2040–2048. [Google Scholar] [CrossRef]
  190. LeMaoult, J.; Krawice-Radanne, I.; Dausset, J.; Carosella, E.D. HLA-G1-expressing antigen-presenting cells induce immunosuppressive CD4+ T cells. Proc. Natl. Acad. Sci. USA 2004, 101, 7064–7069. [Google Scholar] [CrossRef]
  191. Solinas, G.; Germano, G.; Mantovani, A.; Allavena, P. Tumor-associated macrophages (TAM) as major players of the cancer-related inflammation. J. Leukoc. Biol. 2009, 86, 1065–1073. [Google Scholar] [CrossRef]
  192. Moreau, P.; Adrian-Cabestre, F.; Menier, C.; Guiard, V.; Gourand, L.; Dausset, J.; Carosella, E.D.; Paul, P. IL-10 selectively induces HLA-G expression in human trophoblasts and monocytes. Int. Immunol. 1999, 11, 803–811. [Google Scholar] [CrossRef]
  193. Goncalves, A.S.; Oliveira, J.P.; Oliveira, C.F.; Silva, T.A.; Mendonca, E.F.; Wastowski, I.J.; Batista, A.C. Relevance of HLA-G, HLA-E and IL-10 expression in lip carcinogenesis. Hum. Immunol. 2016, 77, 785–790. [Google Scholar] [CrossRef]
  194. Dong, D.D.; Yie, S.M.; Li, K.; Li, F.; Xu, Y.; Xu, G.; Song, L.; Yang, H. Importance of HLA-G expression and tumor infiltrating lymphocytes in molecular subtypes of breast cancer. Hum. Immunol. 2012, 73, 998–1004. [Google Scholar] [CrossRef]
  195. Ibrahim, E.C.; Aractingi, S.; Allory, Y.; Borrini, F.; Dupuy, A.; Duvillard, P.; Carosella, E.D.; Avril, M.F.; Paul, P. Analysis of HLA-F antigen expression in benign and malignant melanocytic lesions reveals that upregulation of HLA-G expression correlates with malignant transformation, high inflammatory infiltration and HLA-F-A1 genotype. Int. J. Cancer 2004, 108, 243–250. [Google Scholar] [CrossRef]
  196. Hofmeister, V. Expression und Immunmodulatorische Funktion von HLA-G und Seinen Verkürzten Isoformen in Tumorzellinien. Thesis 2004, 1, 1. [Google Scholar]
  197. Lin, A.; Zhang, X.; Zhang, R.-L.; Zhang, J.-G.; Zhou, W.-J.; Yan, W.-H. Clinical significance of potential unidentified HLA-G isoforms without α1 domain but containing intron 4 in colorectal cancer patients. Front. Oncol. 2018, 8, 361. [Google Scholar] [CrossRef]
  198. Rizzo, R.; Trentini, A.; Bortolotti, D.; Manfrinato, M.C.; Rotola, A.; Castellazzi, M.; Melchiorri, L.; Di Luca, D.; Dallocchio, F.; Fainardi, E.; et al. Matrix metalloproteinase-2 (MMP-2) generates soluble HLA-G1 by cell surface proteolytic shedding. Mol. Cell. Biochem. 2013, 381, 243–255. [Google Scholar] [CrossRef]
  199. Li, T.; Huang, H.; Liao, D.; Ling, H.; Su, B.; Cai, M. Genetic polymorphism in HLA-G 3′UTR 14-bp ins/del and risk of cancer: A meta-analysis of case-control study. Mol. Genet. Genom. 2015, 290, 1235–1245. [Google Scholar] [CrossRef]
  200. Haghi, M.; Hosseinpour Feizi, M.A.; Sadeghizadeh, M.; Lotfi, A.S. 14-bp insertion/deletion polymorphism of the HLA-G gene in breast cancer among women from North Western Iran. Asian Pac. J. Cancer Prev. 2015, 16, 6155–6158. [Google Scholar] [CrossRef]
  201. Benevolo, M.; Mottolese, M.; Tremante, E.; Rollo, F.; Diodoro, M.G.; Ercolani, C.; Sperduti, I.; Lo Monaco, E.; Cosimelli, M.; Giacomini, P. High expression of HLA-E in colorectal carcinoma is associated with a favorable prognosis. J. Transl. Med. 2011, 9, 184. [Google Scholar] [CrossRef]
  202. Seliger, B.; Jasinski-Bergner, S.; Quandt, D.; Stoehr, C.; Bukur, J.; Wach, S.; Legal, W.; Taubert, H.; Wullich, B.; Hartmann, A. HLA-E expression and its clinical relevance in human renal cell carcinoma. Oncotarget 2016, 7, 67360–67372. [Google Scholar] [CrossRef]
  203. Kren, L.; Valkovsky, I.; Dolezel, J.; Capak, I.; Pacik, D.; Poprach, A.; Lakomy, R.; Redova, M.; Fabian, P.; Krenova, Z.; et al. HLA-G and HLA-E specific mRNAs connote opposite prognostic significance in renal cell carcinoma. Diagn. Pathol. 2012, 7, 58. [Google Scholar] [CrossRef]
  204. Talebian Yazdi, M.; van Riet, S.; van Schadewijk, A.; Fiocco, M.; van Hall, T.; Taube, C.; Hiemstra, P.S.; van der Burg, S.H. The positive prognostic effect of stromal CD8+ tumor-infiltrating T cells is restrained by the expression of HLA-E in non-small cell lung carcinoma. Oncotarget 2016, 7, 3477–3488. [Google Scholar]
  205. Tremante, E.; Ginebri, A.; Lo Monaco, E.; Benassi, B.; Frascione, P.; Grammatico, P.; Cappellacci, S.; Catricala, C.; Arcelli, D.; Natali, P.G.; et al. A melanoma immune response signature including human leukocyte antigen-E. Pigment Cell Melanoma Res. 2014, 27, 103–112. [Google Scholar] [CrossRef]
  206. Allard, M.; Oger, R.; Vignard, V.; Percier, J.M.; Fregni, G.; Perier, A.; Caignard, A.; Charreau, B.; Bernardeau, K.; Khammari, A.; et al. Serum soluble HLA-E in melanoma: A new potential immune-related marker in cancer. PLoS ONE 2011, 6, e21118. [Google Scholar] [CrossRef]
  207. Ishigami, S.; Arigami, T.; Okumura, H.; Uchikado, Y.; Kita, Y.; Kurahara, H.; Maemura, K.; Kijima, Y.; Ishihara, Y.; Sasaki, K.; et al. Human leukocyte antigen (HLA-F)-E and HLA-F expression in gastric cancer. Anticancer Res. 2015, 35, 2279–2285. [Google Scholar]
  208. Sasaki, T.; Ravindranath, M.H.; Terasaki, P.I.; Freitas, M.C.; Kawakita, S.; Jucaud, V. Gastric cancer progression may involve a shift in HLA-E profile from an intact heterodimer to β2-microglobulin-free monomer. Int. J. Cancer 2014, 134, 1558–1570. [Google Scholar] [CrossRef]
  209. Spaans, V.M.; Peters, A.A.; Fleuren, G.J.; Jordanova, E.S. HLA-E expression in cervical adenocarcinomas: Association with improved long-term survival. J. Transl. Med. 2012, 10, 184. [Google Scholar] [CrossRef]
  210. Wolpert, F.; Roth, P.; Lamszus, K.; Tabatabai, G.; Weller, M.; Eisele, G. HLA-E contributes to an immune-inhibitory phenotype of glioblastoma stem-like cells. J. Neuroimmunol. 2012, 250, 27–34. [Google Scholar] [CrossRef]
  211. Chen, A.; Shen, Y.; Xia, M.; Xu, L.; Pan, N.; Yin, Y.; Miao, F.; Shen, C.; Xie, W.; Zhang, J. Expression of the nonclassical HLA-F class I and MICA/B molecules in human hepatocellular carcinoma. Neoplasma 2011, 58, 371–376. [Google Scholar] [CrossRef]
  212. Kren, L.; Fabian, P.; Slaby, O.; Janikova, A.; Soucek, O.; Sterba, J.; Krenova, Z.; Michalek, J.; Kral, Z. Multifunctional immune-modulatory protein HLA-E identified in classical Hodgkin lymphoma: Possible implications. Pathol. Res. Pract. 2012, 208, 45–49. [Google Scholar] [CrossRef]
  213. Zanetti, B.R.; Carvalho-Galano, D.F.; Feitosa, N.L.; Hassumi-Fukasawa, M.K.; Miranda-Camargo, F.A.; Maciel, L.M.; Ribeiro-Silva, A.; Soares, E.G. Differential expression of immune-modulatory molecule HLA-E in non-neoplastic and neoplastic lesions of the thyroid. Int. J. Immunopathol. Pharmacol. 2013, 26, 889–896. [Google Scholar] [CrossRef]
  214. Xu, Y.P.; Wieten, L.; Wang, S.X.; Cai, Y.; Olieslagers, T.; Zhang, L.; He, L.M.; Tilanus, M.G.J.; Hong, W.X. Clinical significance of HLA-E genotype and surface/soluble expression levels between healthy individuals and patients with acute leukemia. Leuk. Lymphoma 2018, 60, 1–8. [Google Scholar] [CrossRef]
  215. Pietra, G.; Romagnani, C.; Manzini, C.; Moretta, L.; Mingari, M.C. The emerging role of HLA-E-restricted CD8+ T lymphocytes in the adaptive immune response to pathogens and tumors. J. Biomed. Biotechnol. 2010, 2010, 907092. [Google Scholar] [CrossRef]
  216. Gooden, M.; Lampen, M.; Jordanova, E.S.; Leffers, N.; Trimbos, J.B.; van der Burg, S.H.; Nijman, H.; van Hall, T. HLA-E expression by gynecological cancers restrains tumor-infiltrating CD8 (+) T lymphocytes. Proc. Natl. Acad. Sci. USA 2011, 108, 10656–10661. [Google Scholar] [CrossRef]
  217. Derre, L.; Corvaisier, M.; Charreau, B.; Moreau, A.; Godefroy, E.; Moreau-Aubry, A.; Jotereau, F.; Gervois, N. Expression and release of HLA-E by melanoma cells and melanocytes: Potential impact on the response of cytotoxic effector cells. J. Immunol. 2006, 177, 3100–3107. [Google Scholar] [CrossRef]
  218. Wagner, B.; da Silva Nardi, F.; Schramm, S.; Kraemer, T.; Celik, A.A.; Durig, J.; Horn, P.A.; Duhrsen, U.; Nuckel, H.; Rebmann, V. HLA-E allelic genotype correlates with HLA-E plasma levels and predicts early progression in chronic lymphocytic leukemia. Cancer 2017, 123, 814–823. [Google Scholar] [CrossRef]
  219. Harada, A.; Ishigami, S.; Kijima, Y.; Nakajo, A.; Arigami, T.; Kurahara, H.; Kita, Y.; Yoshinaka, H.; Natsugoe, S. Clinical implication of human leukocyte antigen (HLA-F)-F expression in breast cancer. Pathol. Int. 2015, 65, 569–574. [Google Scholar] [CrossRef]
  220. Ishigami, S.; Arigami, T.; Setoyama, T.; Okumura, H.; Sasaki, K.; Uchikado, Y.; Kurahara, H.; Kijima, Y.; Nishizono, Y.; Nakajo, A.; et al. Clinical-pathological implication of human leukocyte antigen-F-positive gastric adenocarcinoma. J. Surg. Res. 2013, 184, 802–806. [Google Scholar] [CrossRef]
  221. Li, C.; Wang, Z.; Feng, N.; Dong, J.; Deng, X.; Yue, Y.; Guo, Y.; Hou, J. Human HLA-F F adjacent transcript 10 promotes the formation of cancer initiating cells and cisplatin resistance in bladder cancer. Mol. Med. Rep. 2018, 18, 308–314. [Google Scholar] [CrossRef]
  222. Wu, B.; Yang, H.; Ying, S.; Lu, H.; Wang, W.; Lv, J.; Xiong, H.; Hu, W. High HLA-F expression is a poor prognosis factor in patients with nasopharyngeal carcinoma. Anal. Cell Pathol. 2018, 2018, 7691704. [Google Scholar] [CrossRef]
  223. Morandi, F.; Cangemi, G.; Barco, S.; Amoroso, L.; Giuliano, M.; Gigliotti, A.R.; Pistoia, V.; Corrias, M.V. Plasma levels of soluble HLA-E and HLA-F at diagnosis may predict overall survival of neuroblastoma patients. BioMed. Res. Int. 2013, 2013, 9. [Google Scholar] [CrossRef]
  224. Zhang, J.G.; Zhang, X.; Lin, A.; Yan, W.H. Lesion HLA-F expression is irrelevant to prognosis for patients with gastric cancer. Hum. Immunol. 2013, 74, 828–832. [Google Scholar] [CrossRef]
  225. Melsted, W.N.; Johansen, L.L.; Lock-Andersen, J.; Behrendt, N.; Eriksen, J.O.; Bzorek, M.; Scheike, T.; Hviid, T.V.F. HLA-F class Ia and Ib molecules and FOXP3+ TILs in relation to the prognosis of malignant melanoma patients. Clin. Immunol. 2017, 183, 191–197. [Google Scholar] [CrossRef]
  226. Campillo, J.A.; Martinez-Escribano, J.A.; Moya-Quiles, M.R.; Marin, L.A.; Muro, M.; Guerra, N.; Parrado, A.; Campos, M.; Frias, J.F.; Minguela, A.; et al. Natural killer receptors on CD8 T cells and natural killer cells from different HLA-C phenotypes in melanoma patients. Clin. Cancer Res. 2006, 12, 4822–4831. [Google Scholar] [CrossRef]
  227. Rios, A.; Rodriguez, J.M.; Moya, M.R.; Galindo, P.J.; Canteras, M.; Alvarez, M.R.; Parrilla, P. Frequency of HLA-C alleles in differentiated thyroid carcinoma in Southeastern Spain. Cancer 2004, 100, 264–269. [Google Scholar] [CrossRef]
  228. Laoui, D.; Movahedi, K.; Van Overmeire, E.; Van den Bossche, J.; Schouppe, E.; Mommer, C.; Nikolaou, A.; Morias, Y.; De Baetselier, P.; Van Ginderachter, J.A. Tumor-associated macrophages in breast cancer: Distinct subsets, distinct functions. Int. J. Dev. Biol. 2011, 55, 861–867. [Google Scholar] [CrossRef]
  229. Quatromoni, J.G.; Eruslanov, E. Tumor-associated macrophages: Function, phenotype, and link to prognosis in human lung cancer. Am. J. Transl. Res. 2012, 4, 376–389. [Google Scholar]
  230. Marchesi, M.; Andersson, E.; Villabona, L.; Seliger, B.; Lundqvist, A.; Kiessling, R.; Masucci, G.V. HLA-F-dependent tumour development: A role for tumour associate macrophages? J. Transl. Med. 2013, 11, 247. [Google Scholar] [CrossRef]
  231. Ugurel, S.; Rebmann, V.; Ferrone, S.; Tilgen, W.; Grosse-Wilde, H.; Reinhold, U. Soluble human leukocyte antigen—G serum level is elevated in melanoma patients and is further increased by interferon-alpha immunotherapy. Cancer 2001, 92, 369–376. [Google Scholar] [CrossRef]
  232. Konig, L.; Kasimir-Bauer, S.; Hoffmann, O.; Bittner, A.K.; Wagner, B.; Manvailer, L.F.; Schramm, S.; Bankfalvi, A.; Giebel, B.; Kimmig, R.; et al. The prognostic impact of soluble and vesicular HLA-G and its relationship to circulating tumor cells in neoadjuvant treated breast cancer patients. Hum. Immunol. 2016, 77, 791–799. [Google Scholar] [CrossRef]
  233. Provatopoulou, X.; Kalogera, E.; Sagkriotis, A.; Zagouri, F.; Nonni, A.; Zografos, G.C.; Gounaris, A. Soluble human leukocyte antigen-G expression in patients with ductal and lobular breast malignancy. Anticancer Res. 2012, 32, 1021–1026. [Google Scholar]
  234. Morandi, F.; Levreri, I.; Bocca, P.; Galleni, B.; Raffaghello, L.; Ferrone, S.; Prigione, I.; Pistoia, V. Human neuroblastoma cells trigger an immunosuppressive program in monocytes by stimulating soluble HLA-G release. Cancer Res. 2007, 67, 6433–6441. [Google Scholar] [CrossRef]
  235. Locafaro, G.; Amodio, G.; Tomasoni, D.; Tresoldi, C.; Ciceri, F.; Gregori, S. HLA-G expression on blasts and tolerogenic cells in patients affected by acute myeloid leukemia. J. Immunol. Res. 2014, 2014, 636292. [Google Scholar] [CrossRef]
  236. Grange, C.; Tapparo, M.; Tritta, S.; Deregibus, M.C.; Battaglia, A.; Gontero, P.; Frea, B.; Camussi, G. Role of HLA-G and extracellular vesicles in renal cancer stem cell-induced inhibition of dendritic cell differentiation. BMC Cancer 2015, 15, 1009. [Google Scholar] [CrossRef]
  237. Zhou, K.; Xia, M.; Tang, B.; Yang, D.; Liu, N.; Tang, D.; Xie, H.; Wang, X.; Zhu, H.; Liu, C.; et al. Isolation and comparison of mesenchymal stem celllike cells derived from human gastric cancer tissues and corresponding ovarian metastases. Mol. Med. Rep. 2016, 13, 1788–1794. [Google Scholar] [CrossRef]
  238. Santana-Codina, N.; Carretero, R.; Sanz-Pamplona, R.; Cabrera, T.; Guney, E.; Oliva, B.; Clezardin, P.; Olarte, O.E.; Loza-Alvarez, P.; Mendez-Lucas, A.; et al. A transcriptome-proteome integrated network identifies endoplasmic reticulum thiol oxidoreductase (ERp57) as a hub that mediates bone metastasis. Mol. Cell Proteom. 2013, 12, 2111–2125. [Google Scholar] [CrossRef]
  239. Rouas-Freiss, N.; Moreau, P.; Ferrone, S.; Carosella, E.D. HLA-G proteins in cancer: Do they provide tumor cells with an escape mechanism? Cancer Res. 2005, 65, 10139–10144. [Google Scholar] [CrossRef]
  240. Kochan, G.; Escors, D.; Breckpot, K.; Guerrero-Setas, D. Role of non-classical MHC class I molecules in cancer immunosuppression. Oncoimmunology 2013, 2, e26491. [Google Scholar] [CrossRef]
  241. Allen, C.T.; Clavijo, P.E.; Van Waes, C.; Chen, Z. Anti-tumor immunity in head and neck cancer: Understanding the evidence, how tumors escape and immunotherapeutic approaches. Cancers 2015, 7, 2397–2414. [Google Scholar] [CrossRef]
  242. Liu, Y.; Cao, X. Immunosuppressive cells in tumor immune escape and metastasis. J. Mol. Med. 2016, 94, 509–522. [Google Scholar] [CrossRef]
  243. Rosa, B.; de Jesus, J.P.; de Mello, E.L.; Cesar, D.; Correia, M.M. Effectiveness and safety of monoclonal antibodies for metastatic colorectal cancer treatment: Systematic review and meta-analysis. Ecancermedicalscience 2015, 9, 582. [Google Scholar] [CrossRef]
  244. Shaza, L.; Charette, N.; Hendlisz, A. Colorectal cancer: Ten years of illusion of progress but advances are on the horizon. Rev. Med. Brux 2015, 36, 263–266. [Google Scholar]
  245. Tchekmedyian, N.; Gray, J.E.; Creelan, B.C.; Chiappori, A.A.; Beg, A.A.; Soliman, H.; Perez, B.A.; Antonia, S.J. Propelling immunotherapy combinations into the clinic. Oncology 2015, 29, 990–1002. [Google Scholar]
  246. Lopes, M.; Gonzaga, A.K.G.; Mosconi, C.; Palomino, G.M.; Mendonca, E.F.; Batista, A.C.; Silveira, E. Immune response and evasion mechanisms in lip carcinogenesis: An immunohistochemical study. Arch. Oral. Biol. 2019, 98, 99–107. [Google Scholar] [CrossRef]
  247. Mosconi, C.; Arantes, D.A.C.; Goncalves, A.S.; Alencar, R.C.G.; Oliveira, J.C.; Silva, T.A.; Mendonca, E.F.; Batista, A.C. Immunohistochemical investigations on the expression of programmed cell death ligand 1, human leukocyte antigens G and E, and granzyme B in intraoral mucoepidermoid carcinoma. Arch. Oral. Biol. 2017, 83, 55–62. [Google Scholar] [CrossRef]
  248. Rodriguez, J.A. HLA-F-mediated tumor escape mechanisms that may impair immunotherapy clinical outcomes via T-cell activation. Oncol. Lett. 2017, 14, 4415–4427. [Google Scholar] [CrossRef]
  249. Koepsell, S.A.; Miller, J.S.; McKenna, D.H., Jr. Natural killer cells: A review of manufacturing and clinical utility. Transfusion 2013, 53, 404–410. [Google Scholar] [CrossRef]
  250. Stefanovic, S.; Wirtz, R.; Deutsch, T.M.; Hartkopf, A.; Sinn, P.; Varga, Z.; Sobottka, B.; Sotiris, L.; Taran, F.A.; Domschke, C.; et al. Tumor biomarker conversion between primary and metastatic breast cancer: mRNA assessment and its concordance with immunohistochemistry. Oncotarget 2017, 8, 51416–51428. [Google Scholar] [CrossRef]
  251. O’Brien, C.S.; Farnie, G.; Howell, S.J.; Clarke, R.B. Breast cancer stem cells and their role in resistance to endocrine therapy. Horm. Cancer 2011, 2, 91–103. [Google Scholar] [CrossRef]
  252. Campoli, M.; Ferrone, S. HLA-F antigen changes in malignant cells: Epigenetic mechanisms and biologic significance. Oncogene 2008, 27, 5869–5885. [Google Scholar]
Figure 1. Receptor interaction of HLA-E with NKG2A, -B/CD94 and NKG2C/CD94. HLA-E binds to the inhibiting receptors NKG2A, -B and activating receptor NKG2C, belonging to the killer cell lectin-like receptor C1 (KLRC1) family, expressed on NK cells. The NKG2A and -B receptors mediate an inhibitory signal to the NK cell via immunoreceptor tyrosine-based inhibition motifs (ITIMs) [51]. The activating receptor NKG2C does not possess an intracellular immunoreceptor tyrosine-based activating motif (ITAM), but contains a positively charged transmembrane domain and dimerize with DNAX activation protein 12 (DAP-12), which has an ITAM in its cytoplasmic domain and transmits an activating signal to the cell [51,54,55].
Figure 1. Receptor interaction of HLA-E with NKG2A, -B/CD94 and NKG2C/CD94. HLA-E binds to the inhibiting receptors NKG2A, -B and activating receptor NKG2C, belonging to the killer cell lectin-like receptor C1 (KLRC1) family, expressed on NK cells. The NKG2A and -B receptors mediate an inhibitory signal to the NK cell via immunoreceptor tyrosine-based inhibition motifs (ITIMs) [51]. The activating receptor NKG2C does not possess an intracellular immunoreceptor tyrosine-based activating motif (ITAM), but contains a positively charged transmembrane domain and dimerize with DNAX activation protein 12 (DAP-12), which has an ITAM in its cytoplasmic domain and transmits an activating signal to the cell [51,54,55].
Ijms 20 01830 g001
Figure 2. Receptor interaction of HLA-F with KIR3DL1, -2, KIR3DS1 and -S4 as well as LILRB1 and -B2. The inhibiting receptors KIR3DL1 and -2 and activating receptors KIR3DS1 and KIR2DS4 belong to the family of killer cell immunoglobulin-like receptors (KIRs), which are expressed on NK cells. The inhibiting receptors KIR3DL1 and -2 have a long cytoplasmatic tail (L) with immunoreceptor tyrosine-based inhibition motifs (ITIMs). The activating receptors KIR3DS1 and KIR2DS4 are classified by their number of extracellular domains (two or three domains, 2D or 3D) and short (S) intracellular cytoplasmatic tail, which contains a charged lysine residue instead of an immunoreceptor tyrosine-based inhibition motif (ITIM). They dimerize with DNAX activation protein 12 (DAP-12), which has an immunoreceptor tyrosine-based activating motif (ITAM). The inhibitory leukocyte-immunoglobulin (Ig)-like receptors (LILR) LILRB1 (also known as Ig-like transcript 2; ILT2) and -B2 (also known as ILT4) are expressed on monocytes, dendritic cells (DCs), as well as on B-, T-, and NK cells and mediate an inhibitory signal via their ITIMs.
Figure 2. Receptor interaction of HLA-F with KIR3DL1, -2, KIR3DS1 and -S4 as well as LILRB1 and -B2. The inhibiting receptors KIR3DL1 and -2 and activating receptors KIR3DS1 and KIR2DS4 belong to the family of killer cell immunoglobulin-like receptors (KIRs), which are expressed on NK cells. The inhibiting receptors KIR3DL1 and -2 have a long cytoplasmatic tail (L) with immunoreceptor tyrosine-based inhibition motifs (ITIMs). The activating receptors KIR3DS1 and KIR2DS4 are classified by their number of extracellular domains (two or three domains, 2D or 3D) and short (S) intracellular cytoplasmatic tail, which contains a charged lysine residue instead of an immunoreceptor tyrosine-based inhibition motif (ITIM). They dimerize with DNAX activation protein 12 (DAP-12), which has an immunoreceptor tyrosine-based activating motif (ITAM). The inhibitory leukocyte-immunoglobulin (Ig)-like receptors (LILR) LILRB1 (also known as Ig-like transcript 2; ILT2) and -B2 (also known as ILT4) are expressed on monocytes, dendritic cells (DCs), as well as on B-, T-, and NK cells and mediate an inhibitory signal via their ITIMs.
Ijms 20 01830 g002
Figure 3. Receptor interaction of HLA-G with CD8, KIR2DL4, LILRB1, LILRB2 and CD160. CD8 is a marker for cytotoxic T-cells and consists of an extracellular alpha and beta domain [88]. The receptor KIR2DL4 belongs to the family of killer cell immunoglobulin-like receptors (KIRs), which are expressed on NK cells. The receptor contains two extracellular domains D0 and D1 and has only one immunoreceptor tyrosine-based inhibition motifs (ITIM). A charged arginine residue in its cytoplasmatic tail enables KIR2DL4 to form a complex with Fc fragment receptor γ (FcRγ), which stimulates cytokine production in the NK cell [55]. The inhibitory leukocyte-immunoglobulin (Ig)-like receptors (LILR) LILRB1 (also known as Ig-like transcript 2; ILT2) and -B2 (also known as ILT4) are expressed on monocytes, dendritic cells (DCs), as well as on B-, T-, and NK cells and mediate an inhibitory signal via their ITIMs [36,39,74]. CD160 is a glycosylphosphatidylinositol-anchored receptor and does not contain an immunoreceptor tyrosine-based activating motif (ITAM) [89].
Figure 3. Receptor interaction of HLA-G with CD8, KIR2DL4, LILRB1, LILRB2 and CD160. CD8 is a marker for cytotoxic T-cells and consists of an extracellular alpha and beta domain [88]. The receptor KIR2DL4 belongs to the family of killer cell immunoglobulin-like receptors (KIRs), which are expressed on NK cells. The receptor contains two extracellular domains D0 and D1 and has only one immunoreceptor tyrosine-based inhibition motifs (ITIM). A charged arginine residue in its cytoplasmatic tail enables KIR2DL4 to form a complex with Fc fragment receptor γ (FcRγ), which stimulates cytokine production in the NK cell [55]. The inhibitory leukocyte-immunoglobulin (Ig)-like receptors (LILR) LILRB1 (also known as Ig-like transcript 2; ILT2) and -B2 (also known as ILT4) are expressed on monocytes, dendritic cells (DCs), as well as on B-, T-, and NK cells and mediate an inhibitory signal via their ITIMs [36,39,74]. CD160 is a glycosylphosphatidylinositol-anchored receptor and does not contain an immunoreceptor tyrosine-based activating motif (ITAM) [89].
Ijms 20 01830 g003
Figure 4. Hypotheses for an immunological tumor therapy concept (ITTC). The patent focuses on determining the individual expression pattern of the “embryonic” HLA genes on primary tumors to create individual therapy approaches such as antibodies drug conjugates, vaccination/immunization approaches and in situ gene editing to block and downregulate HLA class Ib expression in order to overcome immune evasion mediated by the non-classical HLA groups. The patent also implies the monitoring of the HLA expression patterns on recurrent tumors and metastases for subsequent treatment adaptations.
Figure 4. Hypotheses for an immunological tumor therapy concept (ITTC). The patent focuses on determining the individual expression pattern of the “embryonic” HLA genes on primary tumors to create individual therapy approaches such as antibodies drug conjugates, vaccination/immunization approaches and in situ gene editing to block and downregulate HLA class Ib expression in order to overcome immune evasion mediated by the non-classical HLA groups. The patent also implies the monitoring of the HLA expression patterns on recurrent tumors and metastases for subsequent treatment adaptations.
Ijms 20 01830 g004
Table 1. Overview of proven occurrences of HLA groups E-G and their locations.
Table 1. Overview of proven occurrences of HLA groups E-G and their locations.
LocalizationHLA GeneAuthors
CTs, STs, EVTsHLA-E, HLA-F, HLA-GApps et al., 2008 [15]
Hackmon et al., 2017 [30]
Rizzo et al., 2011 [103]
Shobu et al., 2006 [11]
ESHLA-E, HLA-GDrukker et al., 2002 [105]
PIEHLA-GFuzzi et al., 2002 [106]
Shaikly et al., 2008 [16]
sHLA-GSher, 2004 [112]
Tabiasco et al., 2009 [113]
PIE/ESHLA-GRizzo et al., 2011 [103]
Verloes et al., 2011 [107]
ES (Wharton-jelly)HLA-E, HLA-F, HLA-GChen et al., 2012 [108]
Culture medium of PIEsHLA-GNoci et al., 2005 [110]
CTs = cytotrophoblasts, ES = embryonic stem cells, EVTs = extravillous trophoblasts, PIE = preimplantation embryo, STs = syncytiotrophoblasts.
Table 2. Selected literature on expression of HLA-G in malignant tumors.
Table 2. Selected literature on expression of HLA-G in malignant tumors.
CarcinomaAuthors
Bladder cancerCastelli et al., 2008 [154]
El-Chennawi et al., 2005 [155]
Gan et al., 2010 [156]
Breast cancerJeong et al., 2014 [130]
Rolfsen et al., 2013 [131]
Da Silva et al.,2013 [132]
He et al.,2010 [133]
Kleinberg et al., 2006 [134]
Palmisano et al., 2002 [135]
Singer et al., 2003 [136]
de Kruijf et al., 2010 [137]
Cervical cancerGimenes et al., 2014 [142]
Li et al., 2012 [143]
Rodriguez et al., 2012 [144]
Zheng et al., 2011 [145]
Ferguson et al., 2012 [136]
Colon cancerZeestraten et al., 2014 [160]
Endometrial cancerBijen et al., 2010 [141]
Esophageal cancerCao et al., 2011 [163]
Lin et al., 2011b [164]
Germ cell tumor (testicular)Karagoz et al., 2014 [179]
GlioblastomaKren et al., 2010 [174]
Kren et al.,2011 [175]
Wastowski et al., 2013 [176]
Wiendl et al., 2002 [177]
Wischhusen et al., 2007 [178]
Hodgkin’s lymphomaDiepstra et al., 2008 [170]
LymphomaUrosevic et al., 2002 [169]
Lung cancerMontilla et al., 2016 [147]
Urosevic et al., 2001 [148]
Wisniewski et al., 2015 [149]
Yan et al., 2015 [150]
Yie et al., 2007 [151]
Zhang et al., 2016 [152]
Malignant melanomaDegenhardt et al., 2010 [165]
Bezuhly et al., 2008 [166]
Paul et al., 1999 [167]
Paul et ak., 1998 [168]
Ovarian cancerZhang et al., 2016 [138]
Rutten et al., 2014 [139]
Lin et al., 2013 [140]
Pancreatic cancerZhou et al., 2015 [180]
Prostate cancerLangat et al., 2006 [146]
Rectal cancerReimers et al., 2014 [163]
Guo et al., 2015 [162]
Renal cancerTronik-Le Roux, 2017 [171]
Ibrahim et al., 2001 [172]
Hanak et al., 2009 [173]
Thyroid cancerDardano et al., 2011 [161]
Nunes et al., 2013 [162]
Table 3. Selected literature proving expression of HLA-E on malignant tumors.
Table 3. Selected literature proving expression of HLA-E on malignant tumors.
CarcinomaAuthors
Breast cancerda Silva et al, 2013 [132]
Cervical carcinoma (adenocarcinoma)Spaans et al., 2012 [213]
Colorectal cancerGuo et al., 2015 [158]
Reimer et al., 2014 [159]
Zeestraten et al., 2014 [160]
Benevolo et al., 2011 [201]
Gastric cancerIshigami et al., 2015 [207]
Sasaki et al., 2014 [208]
GlioblastomaKren et al., 2011 [174]
Wischhusen et al., 2007 [178]
Wolpert et al., 2012 [210]
Hepatic carcinoma (hepatocellular)Chen et al., 2011 [211]
Hodgkin’s lymphomaKren et al., 2012 [212]
LeukemiaXu et al., 2018 [214]
Lung cancerTalebian et al., 2015 [204]
MelanomaTremante et al., 2014 [205]
Allard et al., 2011 [206]
Renal cancerHanak et al., 2009 [173]
Seliger et al., 2016 [202]
Kren et al., 2012 [203]
Thyroid cancerZanetti et al., 2013 [213]
Table 4. Studies showing expression of HLA-F in malignant tumors.
Table 4. Studies showing expression of HLA-F in malignant tumors.
CarcinomaAuthor
Breast cancerHarada et al., 2015 [219]
Bladder cancerLi et al., 2018 [221]
Esophageal cancerZhang et al., 2013 [185]
Gastric cancerIshigami et al., 2015 [207]
Ishigami et al., 2013 [220]
Zhang et al., 2013 [224]
Hepatic cancer (hepatocellular)Xu et al., 2015 [184]
Lung cancerLin et al. 2011a [186]
NeuroblastomaMorandi et al., 2013 [223]

Share and Cite

MDPI and ACS Style

Würfel, F.M.; Winterhalter, C.; Trenkwalder, P.; Wirtz, R.M.; Würfel, W. European Patent in Immunoncology: From Immunological Principles of Implantation to Cancer Treatment. Int. J. Mol. Sci. 2019, 20, 1830. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms20081830

AMA Style

Würfel FM, Winterhalter C, Trenkwalder P, Wirtz RM, Würfel W. European Patent in Immunoncology: From Immunological Principles of Implantation to Cancer Treatment. International Journal of Molecular Sciences. 2019; 20(8):1830. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms20081830

Chicago/Turabian Style

Würfel, Franziska M., Christoph Winterhalter, Peter Trenkwalder, Ralph M. Wirtz, and Wolfgang Würfel. 2019. "European Patent in Immunoncology: From Immunological Principles of Implantation to Cancer Treatment" International Journal of Molecular Sciences 20, no. 8: 1830. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms20081830

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