Abstract
Type I or invariant natural killer T cells belong to a unique lineage of innate T cells, which express markers of both T lymphocytes and NK cells, namely T cell receptor (TCR) and NK1.1 (CD161C), respectively. Thus, apart from direct killing of target cells like NK cells, and they also produce a myriad of cytokines which modulate the adaptive immune responses. Unlike traditional T cells which carry a conventional αβ TCR, NKT cells express semi-invariant TCR – Vα14-Jα18, coupled with Vβ8, Vβ7 and Vβ2 in mice. In humans, the invariant TCR is composed of Vα24-Jα18, coupled with Vβ11.
Keywords: NKT cell immunotherapy, Glioblastoma, T-cell receptor, T-lymphocyte, Cancer immunity, Brain tumor cells, Solid tumors
Introduction to NKT Cell Biology
NKT cell development occurs in the thymus, where they acquire effector functions as they mature [1-4]. In mice, NKT cells are quite enriched within the T lymphocyte compartments of liver and adipose tissues while lower percentages are found in spleen, blood and bone marrow. In humans however, fewer NKT cells are detectable in blood and liver. Promyelocytic leukemia zinc finger (PLZF), the master transcription factor for innate lineage is indispensable for NKT cell development and effector functions [5,6]. Mice deficient of PLZF are almost devoid of NKT cells while the CD4 T cells in T cell-specific PLZF transgenic mice show memory and effector program similar to NKT cells [5,6]. NKT cells are CD1d-restricted in that they recognize lipid antigens presented by CD1d, an MHC-I-like antigen-presenting molecule [2]. NKT cells are known to particularly recognize and react to a glycosphingolipid α-galactoceramide (αGalCer), presented by CD1d, as such, CD1d- αGalCer tetramers are used to stain and recognize NKT cells. Upon activation, either through antigen-recognition or via cytokines (IL-12, IL-18, IL-33), NKT cells have the peculiar ability to rapidly secrete explosive amounts of both Th1 and Th2 cytokines IFNγ and IL-4, apart from other cytokines including IL-17, GM-CSF, IL-2, IL-10, IL-13, TNF-α, etc [7,8]. Recent studies indicate that NKT cells function as effector subsets dedicated to producing Th1, Th2 and Th17 cytokines and are labelled NKT1, NKT2 and NKT17, respectively [7,9].
Literature Review
NKT cells residing in adipose tissues are unique in that they lack PLZF and are more anti-inflammatory in function, being able to produce IL-10 and IL-4 [7,10]. Their number was found to decrease in expanded adipose tissues from obese mice. NKT1 cells are enriched in spleen and liver, NKT2 in lungs and gut while NKT17 cells are more abundant in lymph nodes, skin, intestine and lung tissues, indicating that tissue-resident NKT cells tailor their functions and roles based in the tissues they reside in. A very recent study reported a newer NKT subset, memory follicular helper iNKT (iNKTFH) cells, which are able to generate in the absence of follicular helper T cells by presentation of lipid antigens by dendritic cells and can elicit B cell memory responses [11].
Owing to their ability to produce a myriad of cytokines and chemokines, NKT cells play a role in various diseases, infections and pathological conditions, either protective or pathogenic [12,13]. NKT cell-derived cytokines and chemokines modulate the immune responses of several different cell types, including, but not limited to, conventional CD4 T and CD8 T cells, macrophages, neutrophils, B-cells, NK-cells and dendritic cells [14,15]. Not surprisingly thus, NKT cells have been shown to play protective role in immunity against bacterial and viral pathogens, tumors, and autoimmune diseases [16,17] while being able to promote graft tolerance [18]. On the other hand, NKT cells have been described to play pathogenic roles in atherosclerosis, allergy and metabolic disorders.
NKT cells in cancer immunity
Increasing evidence from recent research indicates that innate-like T cells have significant and important role in cancers making them potential candidates for immunotherapies [19,20]. It is well-accepted that NKT cells aid in tumor immunity. Several studies in different tumor models of mice depicted NKT cell-mediated tumor surveillance [16,21,22]. In humans, reduced numbers of circulating NKT cells, lower proliferation and cytokine expression of NKT cells were demonstrated in patients suffering from different kinds of cancers [23-25]. Further, a positive correlation was reported between the number of tumor-infiltrating NKT cells and a better prognosis [24-27]. Mostly, correlative, these studies in humans corroborated the findings from mice models, pointing towards NKT cells as targets for immunotherapies.
NKT cells target tumors by means of two effective strategies. Firstly, they can kill tumor cells by direct cytolysis using perforin and granzyme B [28,29]. However, with only a few solid tumors expressing CD1d, tumors escape being recognized by NKT cells [29,30]. In fact, down-regulation of CD1d by some tumors was seen, which correlated with the disease progression [31-33]. Secondly, by modulating the recruitment and the function of other immune cells by means of secreted cytokines, NKT cells play an indirect role [20,28]. In this scenario, NKT cells may be activated by CD1d-expressing antigen presenting cells or by antigen-independent activation by means of cytokines in the tumor microenvironment. NKT cells can recruit and/or activate other effector cells like T cells, B cells, NK cells as well as DCs. Alternatively, NKT cells can act by altering the immune-suppression in the tumor microenvironment [34]. Several studies have shown that NKT cells negatively regulate or reverse the immunosuppression by myeloid-derived suppressor cells (MSDCs) and tumor-associated macrophages [34,35].
Because of the varied targets of NKT cells and the ability to boost and amplify adaptive immune responses, NKT cells are believed to be good candidates for anti-tumor therapies. Several pre-clinical and a few clinical studies support this notion of NKT-cell based immune therapies for targeting tumors [36].
NKT cells in glioblastoma
Glioblastoma multiforme (GBM) is the most common and aggressive type of primary brain tumor in adults. Median survival in these patients is less than two years, particularly low in those with high grade gliomas [37]. Despite new approaches to surgery, radiotherapy and chemotherapy, survival has increased only modestly over the last 30-50 years. This poor prognosis for GBM has been attributed to extensive cellular and genetic heterogeneity existing not only between the four major subtypes (classical, neural, pro-neural and mesenchymal) but also at intra-tumoral level [38-40].
Although brain was initially considered as an immune-privileged organ, several studies eventually demonstrated ample amount of infiltration of immune cells in patients with malignant glioma [41,42]. However, endogenous immune responses against brain tumor cells are unable to control the tumor growth [43]. Several preclinical studies have tried to utilize anti-tumor immune responses generated by innate and adoptive immune cells including NK and T cells [44,45]. In most of these cases, immune responses of NK and T cells were suppressed by glioma tumor cells. Another challenge to the development of therapies is the glioma cell-mediated immune regulation at the tumor microenvironment, which depends on several factors and on recruitment of several immune suppressive cells a like Tregs, tumor-associated macrophages and MSDCs [45].
Given the molecular heterogeneity in GBM tumors, and their profound immunosuppression, an effective immunotherapy is a big challenge. Additionally, Gliomas are poorly immunogenic. Thus, multitarget approaches and combinatorial therapies might hold a therapeutic value in fighting gliomas. Recent years have seen a surge in the number of immunotherapeutic studies and strategies against gliomas which show promise [46]. Here we outline the role of NKT-cells in gliomas and the NKT-cell mediated therapies reported thus far.
Discussion
NKT cell numbers and their functional status in glioma patients is the first and most important thing to be considered in order to evaluate the role of these cells in glioma tumor protection. As stated earlier, the NKT cell numbers and the functional responses of NKT cells were reported to be altered in patients with solid tumors [22]. In contrast, the abundance of circulating NKT cells were reported to be comparable between GBM patients and age-matched controls [47,48]. Additionally, the same study showed that NKT cells from GBM patients were equally capable of expanding in vitro in the presence of mature autologous DCs loaded with α-GalCer and were able to kill glioma target cells in a CD1d-depen dent manner. NKT cell-mediated direct tumor lysis is dependent on the expression of CD1d on these cells. However, very few non-hematopoietic solid tumors express CD1d on their surface and gliomas are one of them [31,47,49-51]. Glioma cells from adult patients with both low and high-grade gliomas were found to express CD1d on their surface. What is not yet clear is the extent of NKT cell infiltration into the glioma tumor microenvironment. Also, the in vivo role of NKT cells in glioma patients is yet to be determined.
Medulloblastoma is another type of brain cancer expressing CD1d and not surprisingly thus, medulloblastoma cell lines were effectively killed by NKT cells activated with α-GalCer [51]. Interestingly, injection of NKT cells directly at tumor site significantly extended the survival of NOS/SCID mice implanted with human medulloblastoma cell line. As promising as these results are, it would be interesting to see if the strategy can be applied for treating glioma tumors. In a preclinical model of glioblastoma, irradiated whole tumor cells pulsed with α-GalCer increased the median survival of tumor-bearing mice in a CD1d-dependent manner by prompting T cell adaptive responses [48,52]. They also showed that the irradiated whole tumor cells pulsed with α-GalCer were capable of activating NKT cells in vitro and eliciting T cell responses.
CD1d is expressed by antigen presenting cells including dendritic cells (DCs) at high levels. DCs efficiently present ligands like α-GalCer to NKT cells and activate them to in turn enhance innate and adaptive immune responses [52,53]. Ligands specific to NKT cells (α-GalCer) have been evaluated as adjuvants for dendritic cells (DC)-based vaccine immune therapy in treating breast cancer and melanoma [54,55]. DCs are also the most efficient of APCs and as such they have been tested for vaccine immune therapy by loading with tumor lysates as antigens. A few DC-based vaccines have shown some effectiveness against glioblastoma although only to a limited extent [29,56,57]. Immunization by co-delivery of a-galCer loaded DCs and tumor antigens has proved to be an effective strategy for providing NKT cell-mediated tumor-immunity [48,58-62]. In a recent study, Liu et al. reported an effective method of NKT cell based immune therapy [63]. They used exosomes, small vesicles secreted by tumor cells as antigens. The tumor-derived exosomes were co-delivered with α-GalCer in a DC-based vaccine intravenously to orthotopic glioblastoma rat models to generate a more effective therapeutic response as measured by prolonged survival rate, decreased rate of glioma growth, increased antigen-specific CTL response and strong immune-modulatory effects [63].
Conclusion
Tumor tolerance in glioma depends upon the local production of cytokines and cytokines from tumor cells and recruitment of immunosuppressive cells in tumor microenvironment. It is well established that NKT cells act by modulating the immune responses either by boosting immune responses or by suppressing immune-regulation. However, it is also reported that NKT cells play a role in immune tolerance in a few graft models [64,65]. Involvement of NKT cells in tumor tolerance is not clearly established but one study showed the presence of immune tolerant IL-10+IL-6+ NKT cells in the glioma tissues from patients and micro RNA 92a (miR-92a) secreted from glioma cells was found was found to play a critical role in the generation of these regulatory NKT cells [66]. IL-10 and IL-6 play central role in maintaining the immune suppressive microenvironment in gliomas. IL-10+IL-6+NKT cells, which were induced by co-culture of NKT and glioma cells in vitro, expressed significantly lower levels of cytokines IFN-γ, perforin and FasL and were found to suppress proliferation of CD8+ T cells.
Pending Questions and Future Perspectives
Despite the recent developments which increase our understanding about NKT cell biology and functions [28,67-69], their role in the different types of tumors is not completely understood. The studies on the role of NKT cells in gliomas is very limited. A few studies described here do show good promise for the use of NKT cells as potent immune therapy for treating glioma patients. As glioma tumors are very heterogeneous in nature, more studies are warranted in order to completely understand the role of NKT cells in gliomas. Firstly, expression and functional status of CD1d need to be screened between different grades of gliomas. Localization and activation status of NKT cells in glioma microenvironment are other aspects that may govern the efficacy of NKT cells immune therapy but not much is known. It would be interesting to see if and how NKT cells migrate to glioma tumor site. Signaling pathways involved in the migration of NKT cells to the tumor site can then be meddled with for therapeutic purposes. It is known that gliomas are hypoxic in nature. Recent study shows that the activation and function of NKT cells changes with redox state of these cells and thus it would be interesting to probe if the immune functions of the brain-resident NKT cells are modulated in the glioma microenvironment to favor the glioma growth [68,69]. MDSCs, which are potent immuno-suppressive cells express CD1d and NKT cells have been shown to negatively regulate or reverse their immunosuppression [70-72]. Any role of NKT cells in the regulation of immune suppressive activity of MDSCs in glioma models needs to be investigated. This interaction may enhance the outcome of anti-glioma immune therapy if targeted as combined therapy.
Acknowledgeable
We acknowledge the support received by K.P. in part by National Institutes of Health Grant AI121156 to Dr. Cheong-Hee Chang.
References
- 1.Bendelac A, Savage PB, Teyton L (2007) The biology of NKT cells. Annu Rev Immunol 25: 297–336. [DOI] [PubMed] [Google Scholar]
- 2.Bendelac A, Rivera MN, Park SH, Roark JH (1997) Mouse CD1-specific NK1 T cells: Development, specificity, and function. Annu Rev Immunol 15: 535–562. [DOI] [PubMed] [Google Scholar]
- 3.Godfrey DI, Stankovic S, Baxter AG (2010) Raising the NKT cell family. Nat Immunol 11: 197–206. [DOI] [PubMed] [Google Scholar]
- 4.Benlagha K, Wei DG, Veiga J, Teyton L, Bendelac A (2005) Characterization of the early stages of thymic NKT cell development. J Exp Med 202: 485–492. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Savage AK, Constantinides MG, Han J, Picard D, Martin E, et al. (2008) The transcription factor PLZF directs the effector program of the NKT cell lineage. Immunity 29: 391–403. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Boucek RJ, Noble NL, Wells DE (1976) Variations in 35 SO4 incorporation into glycosaminoglycans along canine coronary arteries. A possible index of artery wall stress. Circ Res 39: 828–832. [DOI] [PubMed] [Google Scholar]
- 7.Lee YJ, Holzapfel KL, Zhu J, Jameson SC, Hogquist KA (2013) Steady-state production of IL-4 modulates immunity in mouse strains and is determined by lineage diversity of iNKT cells. Nat Immunol 14: 1146–1154. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Viale R, Ware R, Maricic I, Chaturvedi V, Kumar V (2012) NKT cell subsets can exert opposing effects in autoimmunity, tumor surveillance and inflammation. Curr Immunol Rev 8: 287–296. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Lee YJ, Wang H, Starrett GJ, Phuong V, Jameson SC, et al. (2015) Tissue-specific distribution of iNKT cells impacts their cytokine response. Immunity 43: 566–578. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Lynch L, Nowak M, Varghese B, Clark J, Hogan AE, et al. (2012) Adipose tissue invariant NKT cells protect against diet-induced obesity and metabolic disorder through regulatory cytokine production. Immunity 37: 574–587. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Harle DG, Baldo BA (1990) Identification of penicillin allergenic determinants that bind IgE antibodies in the sera of subjects with penicillin allergy. Mol Immunol 27: 1063–1071. [DOI] [PubMed] [Google Scholar]
- 12.Berzins SP, Smyth MJ, Baxter AG (2011) Presumed guilty: Natural killer T-cell defects and human disease. Nat Rev Immunol 11: 131–142. [DOI] [PubMed] [Google Scholar]
- 13.Van Kaer L, Parekh VV, Wu L (2013) Invariant natural killer T-cells as sensors and managers of inflammation. Trends Immunol 34: 50–58. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Brennan PJ, Brigl M, Brenner MB (2013) Invariant natural killer T-cells: an innate activation scheme linked to diverse effector functions. Nat Rev Immunol 13: 101–117. [DOI] [PubMed] [Google Scholar]
- 15.Van Kaer L, Parekh VV, Wu L (2010) Invariant natural killer T cells: Bridging innate and adaptive immunity. Cell Tissue Res 343: 43–55. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Exley MA, Lynch L, Varghese B, Nowak M, Alatrakchi N, et al. (2011) Developing understanding of the roles of CD1d-restricted T cell subsets in cancer: reversing tumor-induced defects. Clin Immunol 140: 184–195. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Torina A, Guggino G, La Manna MP, Sireci G (2018) The Janus face of NKT cell function in autoimmunity and infectious diseases. Int J Mol Sci 19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Guan P, Bassiri H, Patel NP, Nichols KE, Das R (2016) Invariant natural killer T cells in hematopoietic stem cell transplantation: killer choice for natural suppression. Bone Marrow Transplant 51: 629–637. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Marcus A, Gowen BG, Thompson TW, Iannello A, Ardolino M, et al. (2014) Recognition of tumors by the innate immune system and natural killer cells. Adv Immunol 122: 91–128. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.McEwen-Smith RM, Salio M, Cerundolo V (2015) The regulatory role of invariant NKT cells in tumor immunity. Cancer Immunol Res 3: 425–435. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Cui J, Shin T, Kawano T, Sato H, Kondo E, et al. (1997) Requirement for Valpha14 NKT cells in IL-12-mediated rejection of tumors. Science 278: 1623–1626. [DOI] [PubMed] [Google Scholar]
- 22.Wolf BJ, Choi JE, Exley MA (2018) Novel approaches to exploiting invariant NKT cells in cancer immunotherapy. Front Immunol 9: 384. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Yanagisawa K, Seino K, Ishikawa Y, Nozue M, Todoroki T, et al. (2002) Impaired proliferative response of V alpha 24 NKT cells from cancer patients against alpha-galactosylceramide. J Immunol 168: 6494–6499. [DOI] [PubMed] [Google Scholar]
- 24.Tachibana T, Onodera H, Tsuruyama T, Mori A, Nagayama S, et al. (2005) Increased intratumor Valpha24-positive natural killer T cells: a prognostic factor for primary colorectal carcinomas. Clin Cancer Res 11: 7322–7327. [DOI] [PubMed] [Google Scholar]
- 25.Molling JW, Kölgen W, Van der Vliet HJ, Boomsma MF, Kruizenga H, et al. (2005) Peripheral blood IFN-gamma-secreting Valpha24+Vbeta11+ NKT cell numbers are decreased in cancer patients independent of tumor type or tumor load. Int J Cancer 116: 87–93. [DOI] [PubMed] [Google Scholar]
- 26.Schneiders FL, De Bruin RC, Van den Eertwegh AJ, Scheper RJ, Leemans CR, et al. (2012) Circulating invariant natural killer T-cell numbers predict outcome in head and neck squamous cell carcinoma: Updated analysis with 10-year follow-up. J Clin Oncol 30: 567–570. [DOI] [PubMed] [Google Scholar]
- 27.Dhodapkar MV, Geller MD, Chang DH, Shimizu K, Fujii S, et al. (2003) A reversible defect in natural killer T cell function characterizes the progression of premalignant to malignant multiple myeloma. J Exp Med 197: 1667–1676. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Nair S, Dhodapkar MV (2017) Natural killer T cells in cancer immunotherapy. Front Immunol 8: 1178. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Metelitsa LS (2011) Anti-tumor potential of type-I NKT cells against CD1d-positive and CD1d-negative tumors in humans. Clin Immunol 140: 119–129. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Haraguchi K, Takahashi T, Nakahara F, Matsumoto A, Kurokawa M, et al. (2006) CD1d expression level in tumor cells is an important determinant for anti-tumor immunity by natural killer T cells. Leuk Lymphoma 47: 2218–2223. [DOI] [PubMed] [Google Scholar]
- 31.Hix LM, Shi YH, Brutkiewicz RR, Stein PL, Wang CR, et al. (2011) CD1d-expressing breast cancer cells modulate NKT cell-mediated antitumor immunity in a murine model of breast cancer metastasis. PLoS One 6: e20702. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Spanoudakis E, Hu M, Naresh K, Terpos E, Melo V, et al. (2009) Regulation of multiple myeloma survival and progression by CD1d. Blood 113: 2498–2507. [DOI] [PubMed] [Google Scholar]
- 33.Miura S, Kawana K, Schust DJ, Fujii T, Yokoyama T, et al. (2010) CD1d, a sentinel molecule bridging innate and adaptive immunity, is downregulated by the human papillomavirus (HPV) E5 protein: A possible mechanism for immune evasion by HPV. J Virol 84: 11614–11623 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Lindau D, Gielen P, Kroesen M, Wesseling P, Adema GJ (2013) The immunosuppressive tumour network: myeloid-derived suppressor cells, regulatory T cells and natural killer T cells. Immunology 138: 105–115. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Ko HJ, Lee JM, Kim YJ, Kim YS, Lee KA, et al. (2009) Immunosuppressive myeloid-derived suppressor cells can be converted into immunogenic APCs with the help of activated NKT cells: An alternative cell-based antitumor vaccine. J Immunol 182: 1818–1828. [DOI] [PubMed] [Google Scholar]
- 36.Dashtsoodol N, Shigeura T, Tashiro T, Aihara M, Chikanishi T, et al. (2017) Natural killer T cell-targeted immunotherapy mediating long-term memory responses and strong antitumor activity. Front Immunol 8: 1206. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Zhang AS, Ostrom QT, Kruchko C, Rogers L, Peereboom DM, et al. (2017) Complete prevalence of malignant primary brain tumors registry data in the United States compared with other common cancers 2010. Neuro Oncol 19: 726–735. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.McLendon R, Friedman A, Bigner D, Van Meir EG, Brat DJ, et al. (2008) Comprehensive genomic characterization defines human glioblastoma genes and core pathways. Nature 455: 1061–1068. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Verhaak RG, Hoadley KA, Purdom E, Wang V, Qi Y, et al. (2010) Integrated genomic analysis identifies clinically relevant subtypes of glioblastoma characterized by abnormalities in PDGFRA, IDH1, EGFR, and NF1. Cancer Cell 17: 98–110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Patel AP, Tirosh I, Trombetta JJ, Shalek AK, Gillespie SM, et al. (2014) Single-cell RNA-seq highlights intratumoral heterogeneity in primary glioblastoma. Science 344: 1396–1401. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Mohammad MG, Tsai VW, Ruitenberg MJ, Hassanpour M, Li H, et al. (2014) Immune cell trafficking from the brain maintains CNS immune tolerance. J Clin Invest 124: 1228–1241. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Engelhardt B, Ransohoff RM (2012) Capture, crawl, cross: The T cell code to breach the blood-brain barriers. Trends Immunol 33: 579–589. [DOI] [PubMed] [Google Scholar]
- 43.Prins RM, Soto H, Konkankit V, Odesa SK, Eskin A, et al. (2010) Gene expression profile correlates with T-cell infiltration and relative survival in glioblastoma patients vaccinated with dendritic cell immunotherapy. Clin Cancer Res 17: 1603–1615. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Zhang C, Burger MC, Jennewein L, Genbler S, Schonfeld K, et al. (2016) ErbB2/HER2-Specific NK cells for targeted therapy of glioblastoma. J Natl Cancer Inst 108. [DOI] [PubMed] [Google Scholar]
- 45.Razavi SM, Lee KE, Jin BE, Aujla PS, Gholamin S, et al. (2016) Immune evasion strategies of glioblastoma. Front Surg 3: 11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Tivnan A, Heilinger T, Lavelle EC, Prehn JH (2016) Advances in immunotherapy for the treatment of glioblastoma. J Neurooncol 131: 1–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Dhodapkar KM, Cirignano B, Chamian F, Zagzag D, Miller DC, et al. (2004) Invariant natural killer T cells are preserved in patients with glioma and exhibit antitumor lytic activity following dendritic cell-mediated expansion. Int J Cancer 109: 893–899. [DOI] [PubMed] [Google Scholar]
- 48.Hunn MK, Farrand KJ, Broadley KW, Weinkove R, Ferguson P, et al. (2012) Vaccination with irradiated tumor cells pulsed with an adjuvant that stimulates NKT cells is an effective treatment for glioma. Clin Cancer Res 18: 6446–6459. [DOI] [PubMed] [Google Scholar]
- 49.Nowak M, Arredouani MS, Tun-Kyi A, Schmidt-Wolf I, Sanda MG, et al. (2010) Defective NKT cell activation by CD1d+ TRAMP prostate tumor cells is corrected by interleukin-12 with alpha-galactosylceramide. PLoS One 5: e11311. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Chong TW, Goh FY, Sim MY, Huang HH, Thike AA, et al. (2014) CD1d expression in renal cell carcinoma is associated with higher relapse rates, poorer cancer-specific and overall survival. J Clin Pathol 68: 200–205. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Liu D, Song L, Brawley VS, Robison N, Wei J, et al. (2013) Medulloblastoma expresses CD1d and can be targeted for immunotherapy with NKT cells. Clin Immunol 149: 55–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Hunn MK, Hermans IF (2013) Exploiting invariant NKT cells to promote T-cell responses to cancer vaccines. Oncoimmunology 2: e23789. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Cerundolo V, Silk JD, Masri SH, Salio M (2009) Harnessing invariant NKT cells in vaccination strategies. Nat Rev Immunol 9: 28–38. [DOI] [PubMed] [Google Scholar]
- 54.Fujii S, Shimizu K, Kronenberg M, Steinman RM (2002) Prolonged IFN-gamma-producing NKT response induced with alpha-galactosylceramide-loaded DCs. Nat Immunol 3: 867–874. [DOI] [PubMed] [Google Scholar]
- 55.Gebremeskel S, Clattenburg DR, Slauenwhite D, Lobert L, Johnston B (2015) Natural killer T cell activation overcomes immunosuppression to enhance clearance of postsurgical breast cancer metastasis in mice. Oncoimmunology 4:e995562. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Marsh JC, Goldfarb J, Shafman TD, Diaz AZ (2013) Current status of immunotherapy and gene therapy for high-grade gliomas. Cancer Control 20: 43–48. [DOI] [PubMed] [Google Scholar]
- 57.Jie X, Hua L, Jiang W, Feng F, Feng G, et al. (2012) Clinical application of a dendritic cell vaccine raised against heat-shocked glioblastoma. Cell Biochem Biophys 62: 91–99. [DOI] [PubMed] [Google Scholar]
- 58.Hong C, Lee H, Oh M, Kang CY, Hong S, et al. (2006) CD4+ T cells in the absence of the CD8+ cytotoxic T cells are critical and sufficient for NKT cell-dependent tumor rejection. J Immunol 177: 6747–6757. [DOI] [PubMed] [Google Scholar]
- 59.Chung Y, Qin H, Kang CY, Kim S, Kwak LW, et al. (2007) An NKT-mediated autologous vaccine generates CD4 T-cell dependent potent antilymphoma immunity. Blood 110: 2013–2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Shimizu K, Kurosawa Y, Taniguchi M, Steinman RM, Fujii S (2007) Cross-presentation of glycolipid from tumor cells loaded with alpha-galactosylceramide leads to potent and long-lived T cell mediated immunity via dendritic cells. J Exp Med 204: 2641–2653. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Petersen TR, Sika-Paotonu D, Knight DA, Dickgreber N, Farrand KJ, et al. (2010) Potent anti-tumor responses to immunization with dendritic cells loaded with tumor tissue and an NKT cell ligand. Immunol Cell Biol 88: 596–604. [DOI] [PubMed] [Google Scholar]
- 62.Ghinnagow R, De Meester J, Cruz LJ, Aspord C, Corgnac S, et al. (2017) Co-delivery of the NKT agonist alpha-galactosylceramide and tumor antigens to cross-priming dendritic cells breaks tolerance to self-antigens and promotes antitumor responses. Oncoimmunology 6: e1339855. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Liu H, Chen L, Liu J, Meng H, Zhang R, et al. (2017) Co-delivery of tumor-derived exosomes with alpha-galactosylceramide on dendritic cell-based immunotherapy for glioblastoma. Cancer Lett 411: 182–190. [DOI] [PubMed] [Google Scholar]
- 64.Seino KI, Fukao K, Muramoto K, Yanagisawa K, Takada Y, et al. (2001) Requirement for natural killer T (NKT) cells in the induction of allograft tolerance. Proc Natl Acad Sci USA 98: 2577–2581. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Hongo D, Tang X, Dutt S, Nador RG, Strober S (2011) Interactions between NKT cells and tregs are required for tolerance to combined bone marrow and organ transplants. Blood 119: 1581–1589. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Tang B, Wu W, Wei X, Li Y, Ren G, et al. (2014) Activation of glioma cells generates immune tolerant NKT cells. J Biol Chem 289: 34595–34600. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Pyaram K, Sen JM, Chang CH (2017) Temporal regulation of Wnt/beta-catenin signaling is important for invariant NKT cell development and terminal maturation. Mol Immunol 85: 47–56. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Kim YH, Kumar A, Chang CH, Pyaram K (2017) Reactive oxygen species regulate the inflammatory function of NKT cells through promyelocytic leukemia zinc finger. J Immunol 199: 3478–3487. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Kumar A, Suryadevara N, Hill TM, Bezbradica JS, Van Kaer L, et al. (2017) Natural killer T cells: An ecological evolutionary developmental biology perspective. Front Immunol 8: 1858. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Gabrilovich DI, Nagaraj S (2009) Myeloid-derived suppressor cells as regulators of the immune system. Nat Rev Immunol 9: 162–174. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.De Santo C, Salio M, Masri SH, Lee LY, Dong T, et al. (2008) Invariant NKT cells reduce the immunosuppressive activity of influenza A virus-induced myeloid-derived suppressor cells in mice and humans. J Clin Invest 118: 4036–4048. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Kusmartsev S, Cheng F, Yu B, Nefedova Y, Sotomayor E, et al. (2003) All-transretinoic acid eliminates immature myeloid cells from tumor-bearing mice and improves the effect of vaccination. Cancer Res 63: 4441–4449. [PubMed] [Google Scholar]