Skip to main content

Human Leukocyte Antigen-G (HLA-G) Expression in Cancers: Roles in Immune Evasion, Metastasis and Target for Therapy

Abstract

Aberrant induction of human leukocyte antigen-G (HLA-G) expression has been observed in various malignancies and is strongly associated with tumor immune escape, metastasis and poor prognosis. To date, great achievements have been made in understanding the underlying mechanisms of HLA-G involved in tumor progression. HLA-G could lead to tumor evasion by inhibition of immune cell cytolysis, differentiation and proliferation and inhibition of cytokine production, induction of immune cell apoptosis, generation of regulatory cells and expansion of myeloid-derived suppressive cells and by impairment of chemotaxis. Moreover, HLA-G could arm tumor cells with a higher invasive and metastatic potential with the upregulation of tumor-promoting factor expression such as matrix metalloproteinases (MMPs), indicating that ectopic HLA-G expression could render multiple effects during the progression of malignancies. In this review, we summarized the mechanisms of HLA-G involved in promoting tumor cell immune escaping, metastasis and disease progression. Special attention will be paid to its significance as an attractive therapeutic target in cancers.

Introduction

The pace of tumor growth is dictated by the continuous interaction between cancer cells and the host immune system. With the selection pressure exerted by immune system, various strategies have been developed and applied by tumor cells to avoid recognition and destruction by different immune effectors (1,2). One of the common strategies used by tumor cells to escape innate and adaptive immune response is associated with an induced aberrant expression of the nonclassical class I molecule human leukocyte antigen-G (HLA-G) (3).

HLA-G is the best characterized nonclassical HLA-class Ib molecules, which includes HLA-E, -F and -H (4). Due to its primary mRNA alternative splicing, seven HLA-G isoforms can be generated, where four of them are membrane-bound (HLA-G1 to -G4) and three soluble (HLA-G5 to -G7) (5). Moreover, another soluble isoform termed “shed HLA-G1” can be generated by proteolytic cleavage of the membrane-bound HLA-G1 by metalloproteinases (6).

HLA-G expression was first observed in cytotrophoblasts (7). In physiological conditions, HLA-G expression was also found in erythroid precursors, cornea, thymic medulla and pancreatic islets (8,911). However, HLA-G can be neo-expressed in pathological conditions including cancers, transplantation, and inflammatory and autoimmune diseases and viral infections (3,5,12,13).

In malignancies, ectopic induction of HLA-G expression has been observed in various types of tumors where it was more frequently observed in tumor lesions with advanced stage and its clinical relevance has been addressed (5,14). HLA-G can directly inhibit immune cell function through receptor binding and/or through trogocytosis and impairment of chemotaxis (15,16); moreover, HLA-G can render tumor cells with a higher invasive, metastatic potential and an unfavorable prognosis in tumor patients, indicating that HLA-G expression has multiple effects including promoting tumor cells to escape immune surveillance and enhancing their metastasis during the progression of malignancies (1719). Therefore, the role of HLA-G in malignancies has gained considerable clinical interest for the possibility of exploiting it as a molecular biomarker and a therapeutic target.

Mechanisms of HLA-G Involved in Tumor Immunology

HLA-G expression could render multiple effects during the progression of malignancies, such as inhibiting immune cell cytolysis, inducing immune cell apoptosis and the generation of regulatory cells through receptor binding and/or trogocytosis, and impairing chemotaxis of different immune effector cells (14,16). Several types of HLA-G receptors including immunoglobulin-like transcript 2 (ILT2)/cluster of differentiation 85j (CD85j), ILT4/CD85d, killer cell immunoglobulin-like receptor 2DL4 (KIR2DL4)/CD158d, CD8 and CD160 have been reported. ILT2 is expressed on B cells, some T cells and NK cells and all monocytes and dendritic cells. ILT4 is expressed only by monocytes, dendritic cells and neutrophils. KIR2DL4 is expressed on natural killer (NK) cells, and CD160 expressed by cytotoxic CD8+ T cells and NK cells, a small proportion of CD4+ T cells, as well as endothelial cells. The CD8 is predominantly expressed on the surface of cytotoxic T cells, but can also be found on NK cells (5,20). Moreover, HLA-G could enhance the expression of tumor metastasis-related factors such as matrix metalloproteinases (MMPs), providing profound effects on tumor progression (18,21,22).

HLA-G-mediated inhibition of the functions of both innate and adaptive immune cells depends on cell types and the profiles of HLA-G receptors they expressed. By binding receptors expressed on various cells, HLA-G could directly inhibit the functions of NK cells, cytotoxic T-lymphocyte (CTLs), B cells, neutrophils and dendritic cells (DCs) (5). HLA-G also has long-lasting indirect immunoregulatory activities by inducing tolerogenic cells including HLA-G-expressing T regulatory (Treg) cells, CD4low and CD8low suppressor T cells, T regulatory type 1 (Tr1) cells, myeloid-derived suppressor cells (MDSC) and DC-10 (2325). Moreover, other immunoinhibitory consequences, including impairment of chemotaxis induced by HLA-G, were also emphasized (26,27) (Figure 1).

Figure 1
figure 1

Tolerogenic functions of HLA-G through binding to receptors expressed on different types of immune cells. MICA, major histocompatibility complex class I-related chain A molecule; NKG2D, natural-killer group 2 member D.

The direct immunosuppression induced by HLA-G included inhibition of CTL and NK cell lysis, allogeneic CD4+ T-cell proliferation and the induction of the activated CD8+ T cells and CD8+ NK cell apoptosis (2831). HLA-G also affects DC maturation, migration, trafficking, antigen presentation and their cross-talk with T and NK cells, and inhibits Vγ9Vδ2 T-cell proliferation and IFN-γ production and cytotoxicity by interacting with the ILT2 inhibitory receptor (3235). We recently reported that NK cell cytolysis inhibited by HLA-G1 in an expression proportion-dependent manner, and HLA-G1 and HLA-G5 isoforms have an additive inhibitory effect on NK cytolysis (36,37). Indeed, in vitro studies with cell lines of ovarian carcinoma, hepatocellular carcinoma, glioma, melanoma and renal cell carcinoma demonstrated that, upon expression of endogenous HLA-G antigens, tumor cells were protected from lysis by allore-active NK cells and lymphokine-activated killer cells and/or antigen-specific CD8+ T cells, and that this protective effect could be reversed by masking HLA-G antigens using HLA-G or its receptor-specific antibody (18,3840). It has been demonstrated that soluble HLA-G (sHLA-G) could inhibit B-cell proliferation, differentiation and immunoglobulin (Ig) secretion in both T-cell-dependent and -independent models of B-cell activation through the receptor ILT2 (41). To be noted, HLA-G5 was shown to inhibit phagocytosis and reactive oxygen species production of neutrophils through ILT4 (42).

Indirect inhibitory immune effects of HLA-G could induce the generation of tolerant cells including Treg, DC and MDSC, or by the process of trogocytosis where activated NK cells, T cells and monocytes could also temporarily inhibit immune responses by acquiring HLA-G-containing membranes from cells in their vicinity (43,44). HLA-G-induced Treg cells were observed after allogeneic stimulation by HLA-G1-expressing antigen-presenting cells (APC), which could induce CD4+ T cell anergy and differentiate into suppressive cells (45). Tolerogenic DCs could be generated through HLA-G1 tetramers or HLA-G5 dimers via ILT4-mediated IL-6 signaling pathway and STAT3 activation. Functionally, HLA-G-induced tolerogenic DCs could induce the generation of CD4+CD25+CTLA-4+ (with CTLA being “cytotoxic T-lymphocyte-associated protein”) and CD8+CD28+ regulatory T cells (20,46).

A novel subset of tolerogenic DCs (DC-10), which secrete high levels of IL-10, and express high levels of HLA-G and its receptors ILT2, ILT4 and ILT3, has a potent specific function to induce IL-10-producing adaptive Tr1 through the ILT4/HLA-G signaling pathway. Data showed that the high expression levels of ILT4, HLA-G and IL-10 are necessary for the tolerogenic activity of DC-10 and their ability to prime T cells to become Tr1 cells (47). Recent data showed that HLA-G-expressing DC-10 and CD4+ T cells are highly represented in acute myeloid leukemia (AML) patients with HLA-G positive blasts (48).

MDSC are present in cancer patients and tumor-bearing mice and are, in part, responsible for the inhibition of the cell-mediated immune response against the tumor (49). It has been shown that infiltration of MDSCs in solid tumors is associated with poor prognosis as circulating MDSCs have a negative impact on survival and inversely correlate with the presence of functional antigen-specific T cells in patients with advanced melanoma (50,51). In ILT2-transgenic mice, HLA-G has been found to induce the emergence of CD11b+ Gr1+ MDSCs with an enhanced suppressive activity and is directly involved in the prolongation of allogeneic skin graft survival (52). In an immunocompetent HLA-G1+ M8 (a human melanoma cell line) tumor-bearing mouse model, interaction between HLA-G and the murine receptor PIR-B could expand the population of CD11b+Gr1+PIR-B+ MDSC, which could decrease NK cytotoxic activity (19). In another study, in a mouse model with murine mammary 4T1 cell line, HLA-G5 was observed to favor the CD11b+ Ly6G+ mice G-MDSC expansion in vivo (17).

Trogocytosis is a process of transferring cell-surface membrane proteins and membrane patches from one cell to another during contact. Relevance of trogocytosis is evidenced in tumor immunology such as HLA-G (53). HLA-G expressed by tumor cells can be acquired by activated cytotoxic NK cells and generates the HLA-G-positive NK cells, which behave as suppressor cells capable of protecting tumor cells from NK-mediated cytolysis (54). Similarly, resting or activated CD4+, CD8+ T and monocytes cells could acquire immunosuppressive HLA-G1 from tumor cells or APCs, which could immediately reverse their function from effectors to regulatory cells. Consequently, HLA-G-mediated immune evasion can be extended to HLA-G-negative tumor cells and further amplify HLA-G tolerogenic effect by trogocytosis (55). In multiple myeloma (MM) patients, HLA-G on malignant plasma cells was associated with a poor prognosis and HLA-G could be acquired by T cells from malignant plasma cells via trogocytosis (56).

Moreover, sHLA-G could impair expression and function of different chemokine receptors in T cells, NK cells and B cells through the receptor ILT2 (16). sHLA-G was observed to downregulate the expression of C-C motif chemokine receptor 2 (CCR2), C-X-C motif chemokine receptor 3 (CXCR3) and CXCR5 in CD4+ and follicular helper T cells, CXCR3 in CD8+ T cells, CXCR3 in Th1 clones, CXCR3 in T-cell receptor (TCR) Vδ2γ9 T cells, which was mediated by the interaction with ILT2. In addition, sHLA-G could inhibit chemotaxis of CD4+ T cells toward CCL2, CCL8, C-X-C motif chemokine ligand 10 (CXCL10) and CXCL11, CD8+ T cells toward CXCL10 and CXCL11, Th1 clones toward CXCL10, TCR Vδ2γ9 T cells toward CXCL10 and CXCL11 and follicular helper T cells toward CXCL13 (27). For NK cells, sHLA-G binding to ILT2 could also impair chemotaxis, cytokine and chemokine secretion in CD56bright and CD56dim NK cells in a dose-dependent manner. Morandi et al. (26) found that sHLA-G downmodulates expression of CXCR3, CX3CR1 and CCR2 and migration toward their specific ligands such as peripheral blood NK cells toward CXCL10, CXCL11 and CX3CL1 and CD56bright NK cells toward CCL2 and CXCL10. Also, HLA-G could down-regulate CXCR4 and CXCR5 expression on germinal center B cells (41).

Relevance of HLA-G Expression in Cancers

The expression of HLA-G in cancer was first demonstrated in the context of melanoma (57). Since then, HLA-G expression in more than 2,000 malignant samples among thirty types of tumors including both solid and hematological malignancies has been studied, where HLA-G expression was observed in different sources such as on the cell surface, secreted or in tumor-derived exosomes (14,58). Among these studies, there is a high frequency of tumor cell-surface HLA-G expression with an absence in healthy tissue, and increased sHLA-G levels has been detected in various body fluids in a variety of cancers (14). Expression of HLA-G was found to be correlated with clinical parameters such as more advanced disease stage, tumor metastasis and/or with a worse prognosis in tumor patients, indicating that HLA-G could facilitate tumor immune escape, invasiveness and metastasis; thereby HLA-G expression was found to be associated with advanced clinical stage and disease progression and HLA-G expression was also documented as an unfavorable prognostic factor for many kinds of solid malignancies, including breast cancer (5964), colorectal cancer (65,66), cervical cancer (67,68), endometrial carcinoma (69,70), esophageal squamous cell carcinoma (7174), gastric cancer (7577), glioblastoma (78), hepatocellular carcinoma (7981), lung cancer (8284), nasopharyngeal carcinoma (85), oral cavity squamous cell carcinoma (86), ovarian cancer (8789), pancreatic adenocarcinoma (90), thyroid carcinoma (91,92). These data were summarized in Table 1. Moreover, HLA-G expression was found to distinguish metastatic from nonmetastatic endometrial carcinoma (69). In our recent study, we found lesion HLA-G5/-G6 isoform expression could discriminate adenocarcinoma from squamous cell carcinoma in lung cancer patients, where tumor cell sHLA-G expression was predominately observed in adenocarcinoma lesions (84). These data indicated that lesion HLA-G or specific HLA-G isoform expression may serve as a clinical marker for tumor metastasis or types of histopathological discrimination.

Table 1 Clinical relevance of HLA-G protein expression in tumor cells.a

Although HLA-G expression in solid tumors as mentioned above was associated with an unfavorable outcome or prognosis, no clear correlation was established between HLA-G expression and unfavorable clinical outcome in hematological malignancies, thus the clinical significance of HLA-G expression in liquid tumors remains controversial (9399), as presented in Table 2.

Table 2 Clinical relevance of HLA-G protein expression in hematological malignancies.

The discrepancy in the clinical significance of HLA-G between solid and liquid tumors may be due to the nature of the tumor cells which, in malignant hematological diseases, are immune cells capable of expressing the HLA-G inhibitory receptor. In this context, studies by Naji et al. (41,100) revealed that, through interaction with ILT2, HLA-G could inhibit the proliferation, differentiation and antibody secretion of activated B cells originating from peripheral blood and secondary lymphoid organs, such as tonsils, and HLA-G could also inhibit the proliferation of ILT2-bearing neoplastic B cell lines such as human B cell lymphomas (Raji, Daudi and Ramos), multiple myelomas (OPM-2, LP1 and RPMI 8226), and B cell leukemia (BV173). These data imply that HLA-G could be correlated with a good prognosis by inhibiting the proliferation of ILT2-expressing tumor cells. Other factors may also account for these controversial results, including differences in expression and interaction of ILT2 and HLA-G by tumor cells, and effects of different chemotherapy status on HLA-G expression between the studies.

To be noted, regulation of HLA-G expression is controlled at the epigenetic, transcriptional and post-transcriptional level. The polymorphic variations in the 5’ upstream regulatory region (5’ URR) and 3’ untranslated regions (3’ UTR) in the HLA-G gene were reported to affect the HLA-G mRNA translation and HLA-G expression by microRNA binding and/or by modifying mRNA stability (101). In this context, a possible association of HLA-G gene polymorphism with susceptibility and/or clinical outcome of malignancies was documented. A 14-base pair insertion/deletion (ins/del) polymorphism (rs66554220) in the 3’ UTR was revealed; both chronic lymphocytic leukemia (CLL) and diffuse large B-cell lymphoma (DLBCL) patients with the del/del genotype had shorter survival than patients with the ins/del or ins/ins genotype (102,103). Additionally, DLBCL patients who carry the HLA-G-725CC genotype in the 5’ URR had a significantly higher probability of overall survival than individuals with other genotype combinations of HLA-G-725C/G/T (103). Furthermore, in the coding segment of the HLA-G gene, the codon 130C deletion occurrence was observed to be significantly associated with a decreased free disease and overall survival in nasopharyngeal carcinoma patients (104). However, more studies on the complete HLA-G gene and its linkage disequilibrium with other genes and on HLA-G expression at both the tumor site and plasma in a specific microenvironment are required to consolidate these findings.

Advances of Tumor-Bearing Animal Models for HLA-G Study

Recently, the development of animal models consolidated the concept that HLA-G+ tumor cells can develop and tolerize the host antitumor immune response in vivo. Although there is no murine homologue of HLA-G, in vivo studies were made possible by the fact that human HLA-G can bind and mediate a signal via the murine receptor-paired immunoglobulin-like receptor (PIR)-B, the homologue of human ILTs (105). Previous data showed that human or murine tumor cells expressing HLA-G can grow in immunocompetent host mice and that blocking HLA-G function by a specific antibody inhibits the development of the tumor, where HLA-G could impair both innate and adaptive immunity resulting in an expansion of CD11b+ Gr1+ myeloid-derived suppressor cells, loss of peripheral T cells and a cytokine balance in favor of a Th2 profile versus Th1/Th17 (17,19).

Other than the tolerant functions of HLA-G in tumor immune escaping, HLA-G was also found in association with the status of tumor invasion and metastasis in patients and preclinical models with the mechanism of upregulating the expression of MMPs (18,106) (Figure 2). MMPs are one of the central mediators of the biology of tumor invasion and metastasis with their ability to degrade basement membrane and extracellular matrix components (107). Indeed, the relationship between HLA-G and MMP expression was addressed in previous studies. Hiden et al. (108) addressed that MMP-15 expression could be induced by TNF-α only in the HLA-G positive first trimester human trophoblast cell line ACH-3P, and only the HLA-G positive extravillous trophoblasts (EVT) have the invasive potential. Much higher MMP-14 expression was also found in HLA-G positive first-trimester invasive trophoblasts (109). Both tumor cells and EVT share similar characteristics in many regards, such as the capability of proliferation, invasion, vasculogenic mimicry, angiogenesis and systemic immune modulation (110). Therefore, similar tactics may be involved in invasion and metastasis of tumor cells.

Figure 2
figure 2

HLA-G promoting tumor cell invasion and metastasis by upregulation of MMPs expression.

Using a Balb/c nu/nu model, we reported that with exogenous HLA-G expression the ovarian cancer cell HO-8910 has a higher invasion and metastasis potential and the host is much less likely to survive, suggesting for the first time that HLA-G expression is associated with tumor metastasis and with poor survival in an animal model in vivo (18). Characterization of the mechanisms by which HLA-G is involved in tumor cell invasion and metastasis shows that upregulated MMP-15 expression was in accordance with HLA-G expression and a strong correlation was also observed between HLA-G and MMP-15 expression in a cohort of ovarian cancer lesions. Furthermore, knockdown of the HLA-G-induced MMP-15 expression significantly decreased cell migration and tumor metastasis (18). In accordance with our findings, HLA-G5 was documented to stimulate primary trophoblasts and trophoblastic cell line (JAR and JEG-3) invasion and to increase MMP-2 transcripts and their activity by binding to KIR2DL4 and ILT2 (22). In another study, suppression of HLA-G expression in JEG-3 cells was observed to decrease the MMP-2 and MMP-9 mRNA and protein expression dramatically (21).

Implications for HLA-G-Based Strategies in Cancer Immunotherapy

Since HLA-G is frequently and specifically expressed on tumor cells, and its expression is associated with unfavorable prognosis in various malignancies, HLA-G is an attractive target for cancer therapies (15,111).

Taking the journey of HLA-G-expressing malignant cells and targeting HLA-G-expressing cancer cells with a delivery system based on nanoparticles carrying anticancer drugs to the tumor site would be also important in maximizing the efficacy of anticancer therapies. In this context, Zhang et al. (112) successfully developed mAb HLA-G and methotrexate (MTX)-loaded nanobubbles (mAbHLA-G/MTX/PLGA NBs [with PLGA being “poly(lactic-co-glycolic acid)”]) as an HLA-G-targeted drug carrier. Data showed that the nanobubbles could efficiently target HLA-G-positive tumor tissues both in vitro and in vivo, and that the released MTX from mAbHLA-G/MTX/ PLGA NBs could remove the residual cancer cells and inhibit the reoccurrence of tumors. Moreover, given the immune inhibitory properties of the HLA-G molecule, it would be important to suppress its expression in tumor cells. Previous studies showed that suppression or downregulation of HLA-G by interfering RNA was found to increase NK cell cytolysis against targets, that antibodies blocking HLA-G or its receptors ILT2 and/or ILT4 could restore the functions of T cells and NK cells, and that the HLA-G-derived peptide HLA-G146-154 could effectively induce peptide-specific CTLs cytotoxic activity against HLA-G-expressing HLA-A24+ renal cell carcinoma cells (15,113,114).

Conclusion

On the basis of the above observations, an HLA-G-based immunotherapy can be achieved by employing HLA-G or HLA-G receptor antibodies to block HLA-G expression or interaction between its receptors, by RNA interfering to down-regulate the HLA-G expression levels, or by using HLA-G-derived immunogenic peptide and anti-idiotype antibodies to stimulate immune cell function. Thus, HLA-G-targeted strategies in malignancies could be attractive in the future for the development of nanomedicine and noninvasive medicine for simultaneously targeted imaging, drug delivery and therapy in cancers (Figure 3). Nevertheless, in hematological tumors, HLA-G could inhibit the proliferation of B-cell malignancies, and, therefore, exogenously produced HLA-G or HLA-G-derived polypeptides can be considered as therapeutic agents for leukemia therapy.

Figure 3
figure 3

Schematic representation of HLA-G-based cancer immunotherapy strategies.

Disclosure

The authors declare that they have no competing interests as defined by Molecular Medicine, or other interests that might be perceived to influence the results and discussion reported in this paper.

References

  1. Bruttel VS, Wischhusen J. (2014) Cancer stem cell immunology: key to understanding tumorigenesis and tumor immune escape? Front Immunol. 5:360.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  2. Gajewski TF, Schreiber H, Fu YX. (2013) Innate and adaptive immune cells in the tumor microenvironment. Nat. Immunol. 14:1014–22.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Curigliano G, Criscitiello C, Gelao L, Goldhirsch A. (2013) Molecular pathways: human leukocyte antigen G (HLA-G). Clin. Cancer Res. 19:5564–71.

    Article  CAS  PubMed  Google Scholar 

  4. Kochan G, Escors D, Breckpot K, Guerrero-Setas D. (2013) Role of non-classical MHC class I molecules in cancer immunosuppression. Oncoimmunology. 2:e26491.

    Article  PubMed  PubMed Central  Google Scholar 

  5. González A, Rebmann V, LeMaoult J, Horn PA, Carosella ED, Alegre E. (2012) The immunosuppressive molecule HLA-G and its clinical implications. Crit. Rev. Clin. Lab. Sci. 49:63–84.

    Article  PubMed  CAS  Google Scholar 

  6. Rizzo R, et al. (2013) Matrix metalloproteinase-2 (MMP-2) generates soluble HLA-G1 by cell surface proteolytic shedding. Mol. Cell. Biochem. 381:243–55.

    Article  CAS  PubMed  Google Scholar 

  7. Kovats S, Main EK, Librach C, Stubblebine M, Fisher SJ, DeMars R. (1990) A class I antigen, HLA-G, expressed in human trophoblasts. Science. 248:220–3.

    Article  CAS  PubMed  Google Scholar 

  8. Menier C, Rabreau M, Challier JC, Le Discorde M, Carosella ED, Rouas-Freiss N. (2004) Erythroblasts secrete the nonclassical HLA-G molecule from primitive to definitive hematopoiesis. Blood. 104:3153–60.

    Article  CAS  PubMed  Google Scholar 

  9. Le Discorde M, Moreau P, Sabatier P, Legeais JM, Carosella ED. (2003) Expression of HLA-G in human cornea, an immune-privileged tissue. Hum. Immunol. 64:1039–44.

    Article  PubMed  CAS  Google Scholar 

  10. Crisa L, McMaster MT, Ishii JK, Fisher SJ, Salomon DR. (1997) Identification of a thymic epithelial cell subset sharing expression of the class Ib HLA-G molecule with fetal trophoblasts. J. Exp. Med. 186:289–98.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Cirulli V, et al. (2006) The class I HLA repertoire of pancreatic islets comprises the nonclassical class Ib antigen HLA-G. Diabetes. 55:1214–22.

    Article  CAS  PubMed  Google Scholar 

  12. Deschaseaux F, Delgado D, Pistoia V, Giuliani M, Morandi F, Durrbach A. (2011) HLA-G in organ transplantation: towards clinical applications. Cell. Mol. Life Sci. 68:397–404.

    Article  CAS  PubMed  Google Scholar 

  13. Rizzo R, Bortolotti D, Baricordi OR, Fainardi E. (2012) New insights into HLA-G and inflammatory diseases. Inflamm. Allergy Drug Targets. 11:448–63.

    Article  CAS  PubMed  Google Scholar 

  14. Yan WH. (2011) Human leukocyte antigen-G in cancer: are they clinically relevant Cancer Lett. 311:123–30.

    Article  CAS  PubMed  Google Scholar 

  15. Yan WH. (2011) HLA-G expression in cancers: potential role in diagnosis, prognosis and therapy. Endocr. Metab. Immune Disord. Drug Targets. 11:76–89.

    Article  PubMed  Google Scholar 

  16. Morandi F, Rouas-Freiss N, Pistoia V. (2014) The emerging role of soluble HLA-G in the control of chemotaxis. Cytokine Growth Factor Rev. 25:327–35.

    Article  CAS  PubMed  Google Scholar 

  17. Loumagne L, Baudhuin J, Favier B, Montespan F, Carosella ED, Rouas-Freiss N. (2014) In vivo evidence that secretion of HLA-G by immunogenic tumor cells allows their evasion from immunosurveillance. Int. J. Cancer. 135:2107–17.

    Article  CAS  PubMed  Google Scholar 

  18. Lin A, Xu HH, Xu DP, Zhang X, Wang Q, Yan WH. (2013) Multiple steps of HLA-G in ovarian carcinoma metastasis: alter NK cytotoxicity and induce matrix metalloproteinase-15 (MMP-15) expression. Hum. Immunol. 74:439–46.

    Article  CAS  PubMed  Google Scholar 

  19. Agaugué S, Carosella ED, Rouas-Freiss N. (2011) Role of HLA-G in tumor escape through expansion of myeloid-derived suppressor cells and cytokinic balance in favor of Th2 versus Th1/Th17. Blood. 117:7021–31.

    Article  PubMed  CAS  Google Scholar 

  20. Fainardi E, et al. (2011) Emerging topics and new perspectives on HLA-G. Cell. Mol. Life Sci. 68:433–51.

    Article  CAS  PubMed  Google Scholar 

  21. Liu H, Liu X, Jin H, Yang F, Gu W, Li X. (2013) Proteomic analysis of knock-down HLA-G in invasion of human trophoblast cell line JEG-3. Int. J. Clin. Exp. Pathol. 6:2451–9.

    PubMed  PubMed Central  Google Scholar 

  22. Guo Y, et al. (2013) Soluble human leukocyte antigen-g5 activates extracellular signal-regulated protein kinase signaling and stimulates trophoblast invasion. PLoS One. 8:e76023.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Amodio G, Gregori S. (2012) Human tolerogenic DC-10: perspectives for clinical applications. Transplant Res. 1:14.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Carosella ED, Gregori S, LeMaoult J. (2011) The tolerogenic interplay(s) among HLA-G, myeloid APCs, and regulatory cells. Blood. 118:6499–505.

    Article  CAS  PubMed  Google Scholar 

  25. Amodio G, et al. (2015) HLA-G expression levels influence the tolerogenic activity of human DC-10. Haematologica. 100:548–57.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  26. Morandi F, et al. (2011) Soluble HLA-G dampens CD94/NKG2A expression and function and differentially modulates chemotaxis and cytokine and chemokine secretion in CD56bright and CD56dim NK cells. Blood. 118:5840–50.

    Article  CAS  PubMed  Google Scholar 

  27. Morandi F, Ferretti E, Bocca P, Prigione I, Raffaghello L, Pistoia V. (2010) A novel mechanism of soluble HLA-G mediated immune modulation: downregulation of T cell chemokine receptor expression and impairment of chemotaxis. PLoS One. 5:e11763.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  28. Le Gal FA, et al. (1999) HLA-G-mediated inhibition of antigen-specific cytotoxic T lymphocytes. Int. Immunol. 11:1351–6.

    Article  PubMed  Google Scholar 

  29. Lin A, et al. (2007) HLA-G expression in human ovarian carcinoma counteracts NK cell function. Ann. Oncol. 18:1804–9.

    Article  CAS  PubMed  Google Scholar 

  30. Bainbridge DR, Ellis SA, Sargent IL. (2000) HLA-G suppresses proliferation of CD4(+) T-lymphocytes. J. Reprod. Immunol. 48:17–26.

    Article  CAS  PubMed  Google Scholar 

  31. Contini P, et al. (2003) Soluble HLA-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. 33:125–34.

    Article  CAS  PubMed  Google Scholar 

  32. Horuzsko A, Lenfant F, Munn DH, Mellor AL. (2001) Maturation of antigen-presenting cells is compromised in HLA-G transgenic mice. Int. Immunol. 13:385–94.

    Article  CAS  PubMed  Google Scholar 

  33. Liang S, Ristich V, Arase H, Dausset J, Carosella ED, Horuzsko A. (2008) Modulation of dendritic cell differentiation by HLA-G and ILT4 requires the IL-6—STAT3 signaling pathway. Proc. Natl. Acad. Sci. U.S.A. 105:8357–8362.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Gros F, Cabillic F, Toutirais O, Maux AL, Sebti Y, Amiot L. (2008) Soluble HLA-G molecules impair natural killer/dendritic cell crosstalk via inhibition of dendritic cells. Eur. J. Immunol. 38:742–9.

    Article  CAS  PubMed  Google Scholar 

  35. Lesport E, et al. (2011) Inhibition of human Vγ9Vδ2 T-cell antitumoral activity through HLA-G: implications for immunotherapy of cancer. Cell. Mol. Life Sci. 68:3385–99.

    Article  CAS  PubMed  Google Scholar 

  36. Chen BG, Xu DP, Lin A, Yan WH. (2013) NK cytolysis is dependent on the proportion of HLA-G expression. Hum. Immunol. 74:286–9.

    Article  CAS  PubMed  Google Scholar 

  37. Zhang WQ, et al. (2014) HLA-G1 and HLA-G5 isoforms have an additive effect on NK cytolysis. Hum. Immunol. 75:182–9.

    Article  CAS  PubMed  Google Scholar 

  38. Wiendl H, et al. (2002) A functional role of HLA-G expression in human gliomas: an alternative strategy of immune escape. J. Immunol. 168:4772–80.

    Article  CAS  PubMed  Google Scholar 

  39. Riteau B, et al. (2001) HLA-G1 co-expression boosts the HLA class I-mediated NK lysis inhibition. Int. Immunol. 13:193–201.

    Article  CAS  PubMed  Google Scholar 

  40. Bukur J, et al. (2003) Functional role of human leukocyte antigen-G up-regulation in renal cell carcinoma. Cancer Res. 63:4107–11.

    CAS  PubMed  Google Scholar 

  41. Naji A, et al. (2014) Binding of HLA-G to ITIM-bearing Ig-like transcript 2 receptor suppresses B cell responses. J. Immunol. 192:1536–46.

    Article  CAS  PubMed  Google Scholar 

  42. Baudhuin J, et al. (2013) Exocytosis acts as a modulator of the ILT4-mediated inhibition of neutrophil functions. Proc. Natl. Acad. Sci. U.S.A. 110:17957–62.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Carosella ED, Favier B, Rouas-Freiss N, Moreau P, Lemaoult J. (2008) Beyond the increasing complexity of the immunomodulatory HLA-G molecule. Blood. 111:4862–70.

    Article  CAS  PubMed  Google Scholar 

  44. Carosella ED, HoWangYin KY, Favier B, LeMaoult J. (2008) HLA-G-dependent suppressor cells: Diverse by nature, function, and significance. Hum. Immunol. 69:700–7.

    Article  CAS  PubMed  Google Scholar 

  45. LeMaoult J, Krawice-Radanne I, Dausset J, Carosella ED. (2004) HLA-G1-expressing antigen-presenting cells induce immunosuppressive CD4+ T cells. Proc. Natl. Acad. Sci. U.S.A. 101:7064–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Ristich V, Liang S, Zhang W, Wu J, Horuzsko A. (2005) Tolerization of dendritic cells by HLA-G. Eur. J. Immunol. 35:1133–42.

    Article  CAS  PubMed  Google Scholar 

  47. Gregori S, et al. (2010) Differentiation of type 1 T regulatory cells (Tr1) by tolerogenic DC-10 requires the IL-10-dependent ILT4/HLA-G pathway. Blood. 116:935–44.

    Article  CAS  PubMed  Google Scholar 

  48. Locafaro G, Amodio G, Tomasoni D, Tresoldi C, Ciceri F, Gregori S. (2014) HLA-G expression on blasts and tolerogenic cells in patients affected by acute myeloid leukemia. J. Immunol. Res. 2014:636292.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  49. Serafini P. (2013) Myeloid derived suppressor cells in physiological and pathological conditions: the good, the bad, and the ugly. Immunol. Res. 57:172–84.

    Article  PubMed  Google Scholar 

  50. Khaled YS, Ammori BJ, Elkord E. (2013) Myeloid-derived suppressor cells in cancer: recent progress and prospects. Immunol. Cell Biol. 91:493–502.

    Article  CAS  PubMed  Google Scholar 

  51. Weide B, et al. (2014) Myeloid-derived suppressor cells predict survival of patients with advanced melanoma: comparison with regulatory T cells and NY-ESO-1- or melan-A-specific T cells. Clin. Cancer Res. 20:1601–9.

    Article  CAS  PubMed  Google Scholar 

  52. Zhang W, Liang S, Wu J, Horuzsko A. (2008) Human inhibitory receptor immunoglobulin-like transcript 2 amplifies CD11b+Gr1+ myeloid-derived suppressor cells that promote long-term survival of allografts. Transplantation. 86:1125–34.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Carosella ED, Gregori S, Rouas-Freiss N, LeMaoult J, Menier C, Favier B. (2011) The role of HLA-G in immunity and hematopoiesis. Cell. Mol. Life Sci. 68:353–68.

    Article  CAS  PubMed  Google Scholar 

  54. Caumartin J, et al. (2007) Trogocytosis-based generation of suppressive NK cells. EMBO J. 26:1423–33.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. HoWangYin KY, Alegre E, Daouya M, Favier B, Carosella ED, LeMaoult J. (2010) Different functional outcomes of intercellular membrane transfers to monocytes and T cells. Cell. Mol. Life Sci. 67:1133–45.

    Article  CAS  PubMed  Google Scholar 

  56. Brown R, et al. (2012) CD86+ or HLA-G+ can be transferred via trogocytosis from myeloma cells to T cells and are associated with poor prognosis. Blood. 120:2055–63.

    Article  CAS  PubMed  Google Scholar 

  57. Paul P, et al. (1998) HLA-G expression in melanoma: a way for tumor cells to escape from immunosurveillance. Proc. Natl. Acad. Sci. U.S.A. 95:4510–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Riteau B, et al. (2003) Exosomes bearing HLA-G are released by melanoma cells. Hum. Immunol. 64:1064–72.

    Article  CAS  PubMed  Google Scholar 

  59. Kleinberg L, et al. (2006) Expression of HLA-G in malignant mesothelioma and clinically aggressive breast carcinoma. Virchows Arch. 449:31–9.

    Article  CAS  PubMed  Google Scholar 

  60. Chen HX, et al. (2010) Upregulation of human leukocyte antigen-G expression and its clinical significance in ductal breast cancer. Hum. Immunol. 71:892–8.

    Article  CAS  PubMed  Google Scholar 

  61. He X, et al. (2010) 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. 17:1459–69.

    Article  PubMed  Google Scholar 

  62. de Kruijf EM, et al. (2010) HLA-E and HLA-G expression in classical HLA class I-negative tumors is of prognostic value for clinical outcome of early breast cancer patients. J. Immunol. 185:7452–9.

    Article  PubMed  CAS  Google Scholar 

  63. da Silva GB, et al. (2013) Expression of the classical and nonclassical HLA molecules in breast cancer. Int. J. Breast Cancer. 2013:250435.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  64. Ramos CS, et al. (2014) Analysis of HLA-G gene polymorphism and protein expression in invasive breast ductal carcinoma. Hum. Immunol. 75:667–672.

    Article  CAS  PubMed  Google Scholar 

  65. Ye SR, Yang H, Li K, Dong DD, Lin XM, Yie SM. (2007) Human leukocyte antigen G expression: as a significant prognostic indicator for patients with colorectal cancer. Mod. Pathol. 20:375–83.

    Article  CAS  PubMed  Google Scholar 

  66. Guo ZY, et al. (2015) Predictive value of HLA-G and HLA-E in the prognosis of colorectal cancer patients. Cell. Immunol. 293:10–6.

    Article  CAS  PubMed  Google Scholar 

  67. Miranda LN, et al. (2015) Greater expression of the human leukocyte antigen-G (HLA-G) and interleukin-17 (IL-17) in cervical intraepithelial neoplasia: analytical cross-sectional study. Sao Paulo Med. J. 133:336–42.

    Article  PubMed  Google Scholar 

  68. Li XJ, Zhang X, Lin A, Ruan YY, Yan WH. (2012) Human leukocyte antigen-G (HLA-G) expression in cervical cancer lesions is associated with disease progression. Hum. Immunol. 73:946–9.

    Article  CAS  PubMed  Google Scholar 

  69. Barrier BF, Kendall BS, Sharpe-Timms KL, Kost ER. (2006) Characterization of human leukocyte antigen-G (HLA-G) expression in endometrial adenocarcinoma. Gynecol. Oncol. 103:25–30.

    Article  CAS  PubMed  Google Scholar 

  70. Bijen CB, et al. (2010) The prognostic role of classical and nonclassical MHC class I expression in endometrial cancer. Int. J. Cancer. 126:1417–27.

    CAS  PubMed  Google Scholar 

  71. Yie SM, Yang H, Ye SR, Li K, Dong DD, Lin XM. (2007) Expression of HLA-G is associated with prognosis in esophageal squamous cell carcinoma. Am. J. Clin. Pathol. 128:1002–9.

    Article  PubMed  Google Scholar 

  72. Lin A, et al. (2011) Human leukocyte antigen-G expression is associated with a poor prognosis in patients with esophageal squamous cell carcinoma. Int. J. Cancer. 129:1382–90.

    Article  CAS  PubMed  Google Scholar 

  73. Zheng J, et al. (2014) Human leukocyte antigen G is associated with esophageal squamous cell carcinoma progression and poor prognosis. Immunol. Lett. 161:13–9.

    Article  CAS  PubMed  Google Scholar 

  74. Hu J, et al. (2013) Overexpression of HLA-G Is positively associated with Kazakh esophageal squamous cell carcinoma in Xinjiang, China. Viral Immunol. 26:180–4.

    Article  CAS  PubMed  Google Scholar 

  75. Yie SM, Yang H, Ye SR, Li K, Dong DD, Lin XM. (2007) Expression of human leukocyte antigen G (HLA-G) correlates with poor prognosis in gastric carcinoma. Ann. Surg. Oncol. 14:2721–9.

    Article  PubMed  Google Scholar 

  76. Tuncel T, et al. (2013) Immunoregulatory function of HLA-G in gastric cancer. Asian Pac. J. Cancer Prev. 14:7681–4.

    Article  PubMed  Google Scholar 

  77. Du L, et al. (2011) Human leukocyte antigen-G is closely associated with tumor immune escape in gastric cancer by increasing local regulatory T cells. Cancer Sci. 102:1272–80.

    Article  CAS  PubMed  Google Scholar 

  78. Wastowski IJ, et al. (2013) Human leukocyte antigen-G is frequently expressed in glioblastoma and may be induced in vitro by combined 5-aza-2’-deoxycytidine and interferon-γ treatments: results from a multicentric study. Am. J. Pathol. 182:540–52.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Cai MY, et al. (2009) Human leukocyte antigen-G protein expression is an unfavorable prognostic predictor of hepatocellular carcinoma following curative resection. Clin. Cancer Res. 15:4686–93.

    Article  CAS  PubMed  Google Scholar 

  80. Wang Y, Ye Z, Meng XQ, Zheng SS. (2011) Expression of HLA-G in patients with hepatocellular carcinoma. Hepatobiliary Pancreat. Dis. Int. 10:158–63.

    Article  CAS  PubMed  Google Scholar 

  81. Lin A, et al. (2010) Aberrant human leucocyte antigen-G expression and its clinical relevance in hepatocellular carcinoma. J. Cell Mol. Med. 14:2162–71.

    Article  CAS  PubMed  Google Scholar 

  82. Yie SM, Yang H, Ye SR, Li K, Dong DD, Lin XM. (2007) Expression of human leucocyte antigen G (HLA-G) is associated with prognosis in non-small cell lung cancer. Lung Cancer. 58:267–74.

    Article  PubMed  Google Scholar 

  83. Lin A, et al. (2010) Clinical relevance and functional implications for human leucocyte antigen-g expression in non-small-cell lung cancer. J. Cell Mol. Med. 14:2318–29.

    Article  CAS  PubMed  Google Scholar 

  84. Yan WH, Liu D, Lu HY, Li YY, Zhang X, Lin A. (2015) 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. 19:778–85.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Cai MB, et al. (2012) Expression of human leukocyte antigen G is associated with prognosis in nasopharyngeal carcinoma. Int. J. Biol. Sci. 8:891–900.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Gonçalves AS, et al. (2014) The clinicopathologic significance of the expression of HLA-G in oral squamous cell carcinoma. Oral Surg. Oral Med. Oral Pathol. Oral Radiol. 117:361–8.

    Article  PubMed  Google Scholar 

  87. Jung YW, et al. (2009) Correlation of human leukocyte antigen-G (HLA-G) expression and disease progression in epithelial ovarian cancer. Reprod. Sci. 16:1103–11.

    Article  CAS  PubMed  Google Scholar 

  88. Rutten MJ, et al. (2014) HLA-G expression is an independent predictor for improved survival in high grade ovarian carcinomas. J. Immunol. Res. 2014:274584.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Menier C, Prevot S, Carosella ED, Rouas-Freiss N. (2009) Human leukocyte antigen-G is expressed in advanced-stage ovarian carcinoma of high-grade histology. Hum. Immunol. 70:1006–9.

    Article  CAS  PubMed  Google Scholar 

  90. Xu YF, et al. (2015) High expression of human leukocyte antigen-G is associated with a poor prognosis in patients with PDAC. Curr. Mol. Med. 15:360–7.

    Article  CAS  PubMed  Google Scholar 

  91. de Figueiredo Feitosa NL, et al. (2014) HLA-G is differentially expressed in thyroid tissues. Thyroid. 24:585–92.

    Article  PubMed  CAS  Google Scholar 

  92. Nunes LM, et al. (2013) Association between the HLA-G molecule and lymph node metastasis in papillary thyroid cancer. Hum. Immunol. 74:447–51.

    Article  CAS  PubMed  Google Scholar 

  93. Alkhouly N, Shehata I, Ahmed MB, Shehata H, Hassan S, Ibrahim T. (2013) HLA-G expression in acute lymphoblastic leukemia: a significant prognostic tumor biomarker. Med. Oncol. 30:460.

    Article  PubMed  CAS  Google Scholar 

  94. Guo QY, Chen BG, Ruan YY, Lin A, Yan WH. (2011) HLA-G expression is irrelevant to prognosis in patients with acute myeloid leukemia. Leuk. Res. 35:1350–4.

    Article  CAS  PubMed  Google Scholar 

  95. Attia MA, Nosair NA, Gawally A, Elnagar G, Elshafey EM. (2014) HLA-G expression as a prognostic indicator in B-cell chronic lymphocytic leukemia. Acta Haematol. 132:53–8.

    Article  CAS  PubMed  Google Scholar 

  96. Nückel H, Rebmann V, Dürig J, Dührsen U, Grosse-Wilde H. (2005) HLA-G expression is associated with an unfavorable outcome and immunodeficiency in chronic lymphocytic leukemia. Blood. 105:1694–8.

    Article  PubMed  CAS  Google Scholar 

  97. Erikci AA, Karagoz B, Ozyurt M, Ozturk A, Kilic S, Bilgi O. (2009) HLA-G expression in B chronic lymphocytic leukemia: a new prognostic marker? Hematology. 14:101–5.

    Article  CAS  PubMed  Google Scholar 

  98. Perez-Chacon G, et al. (2009) Prognostic irrelevance of HLA-G in B-cell chronic lymphocytic leukemia. Int. J. Lab. Hematol. 31:327–37.

    Article  CAS  PubMed  Google Scholar 

  99. Giannopoulos K, Dmoszyńska A, Bojarska-Junak A, Schmitt M, Rolifński J. (2008) Expression of HLA-G in patients with B-cell chronic lymphocytic leukemia (B-CLL). Folia Histochem. Cytobiol. 46:457–60.

    CAS  PubMed  Google Scholar 

  100. Naji A, Menier C, Maki G, Carosella ED, Rouas-Freiss N. (2012) Neoplastic B-cell growth is impaired by HLA-G/ILT2 interaction. Leukemia. 26:1889–92.

    Article  CAS  PubMed  Google Scholar 

  101. Donadi EA, Castelli EC, Arnaiz-Villena A, Roger M, Rey D, Moreau P. (2011) Implications of the polymorphism of HLA-G on its function, regulation, evolution and disease association. Cell. Mol. Life Sci. 68:369–95.

    Article  CAS  PubMed  Google Scholar 

  102. Rizzo R, et al. (2014) HLA-G is a component of the chronic lymphocytic leukemia escape repertoire to generate immune suppression: impact of the HLA-G 14 base pair (rs66554220) polymorphism. Haematologica. 99:888–96.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  103. Bielska M, et al. (2015) Human leukocyte antigen-G polymorphisms influence the clinical outcome in diffuse large B-cell lymphoma. Genes Chromosomes Cancer. 54:185–93.

    Article  CAS  PubMed  Google Scholar 

  104. Ghandri N, et al. (2011) Association of HLA-G polymorphisms with nasopharyngeal carcinoma risk and clinical outcome. Hum. Immunol. 72:150–8.

    Article  CAS  PubMed  Google Scholar 

  105. Liang S, Baibakov B, Horuzsko A. (2002) HLA-G inhibits the functions of murine dendritic cells via the PIR-B immune inhibitory receptor. Eur. J. Immunol. 32:2418–26.

    Article  CAS  PubMed  Google Scholar 

  106. Ueshima C, et al. (2015) The killer cell Ig-like receptor 2DL4 expression in human mast cells and its potential role in breast cancer invasion. Cancer Immunol. Res. 3:871–80.

    Article  CAS  PubMed  Google Scholar 

  107. Hadler-Olsen E, Winberg JO, Uhlin-Hansen L. (2013) Matrix metalloproteinases in cancer: their value as diagnostic and prognostic markers and therapeutic targets. Tumour Biol. 34:2041–51.

    Article  CAS  PubMed  Google Scholar 

  108. Hiden U, et al. (2007) The first trimester human trophoblast cell line ACH-3P: a novel tool to study autocrine/paracrine regulatory loops of human trophoblast subpopulations—TNF-alpha stimulates MMP15 expression. BMC Dev. Biol. 7:137.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  109. Hiden U, et al. (2008) MT1-MMP expression in first-trimester placental tissue is upregulated in type 1 diabetes as a result of elevated insulin and tumor necrosis factor-alpha levels. Diabetes. 57:150–7.

    Article  CAS  PubMed  Google Scholar 

  110. Holtan SG, Creedon DJ, Haluska P, Markovic SN. (2009) Cancer and pregnancy: parallels in growth, invasion, and immune modulation and implications for cancer therapeutic agents. Mayo Clin. Proc. 84:985–1000.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  111. Amiot L, Ferrone S, Grosse-Wilde H, Seliger B. (2011) Biology of HLA-G in cancer: a candidate molecule for therapeutic intervention? Cell. Mol. Life Sci. 68:417–31.

    Article  CAS  PubMed  Google Scholar 

  112. Zhang X, et al. (2014) Methotrexate-loaded PLGA nanobubbles for ultrasound imaging and synergistic targeted therapy of residual tumor during HIFU ablation. Biomaterials. 35:5148–61.

    Article  CAS  PubMed  Google Scholar 

  113. Komohara Y, et al. (2007) HLA-G as a target molecule in specific immunotherapy against renal cell carcinoma. Oncol. Rep. 18:1463–8.

    CAS  PubMed  Google Scholar 

  114. Jasinski-Bergner S, et al. (2015) Clinical relevance of miR-mediated HLA-G regulation and the associated immune cell infiltration in renal cell carcinoma. Oncoimmunology. 4:e1008805.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

Download references

Acknowledgments

This work was supported by grants from the National Natural Science Foundation of China (31170879, 31370920, 81372247), the Natural Science Foundation of Zhejiang Province (LR13H160001) and the Zhejiang Provincial program for the cultivation of high-level innovative health talents.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Wei-Hua Yan.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, and provide a link to the Creative Commons license. You do not have permission under this license to share adapted material derived from this article or parts of it.

The images or other third party material in this article are included in the article’s Creative Commons license, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons license and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder.

To view a copy of this license, visit (https://doi.org/creativecommons.org/licenses/by-nc-nd/4.0/)

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Lin, A., Yan, WH. Human Leukocyte Antigen-G (HLA-G) Expression in Cancers: Roles in Immune Evasion, Metastasis and Target for Therapy. Mol Med 21, 782–791 (2015). https://doi.org/10.2119/molmed.2015.00083

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.2119/molmed.2015.00083

Keywords