Available online at www.sciencedirect.com
ScienceDirect The transition from HLA-I positive to HLA-I negative primary tumors: the road to escape from T-cell responses Natalia Aptsiauri2,3, Francisco Ruiz-Cabello1,2,3 and Federico Garrido1,2,3 MHC/HLA class I loss in cancer is one of the main mechanisms of tumor immune escape from T-cell recognition and destruction. Tumor infiltration by T lymphocytes (TILs) and by other immune cells was first described many years ago, but has never been directly and clearly linked to the destruction of HLAI positive and selection of HLA-I negative tumor cells. The degree and the pattern of lymphocyte infiltration in a tumor nest may depend on antigenicity and the developmental stages of the tumors. In addition, it is becoming evident that HLA-I expression and tumor infiltration have a direct correlation with tumor tissue reorganization. We observed that at early stages (permissive Phase I) tumors are heterogeneous, with both HLAI positive and HLA-negative cancer cells, and are infiltrated by TILs and M1 macrophages as a part of an active anti-tumor Th1 response. At later stages (encapsulated Phase II), tumor nests are mostly HLA-I negative with immune cells residing in the peri-tumoral stroma, which forms a granuloma-like encapsulated tissue structure. All these tumor characteristics, including tumor HLA-I expression pattern, have an important clinical prognostic value and should be closely and routinely investigated in different types of cancer by immunologists and by pathologists. In this review we summarize our current viewpoint about the alterations in HLA-I expression in cancer and discuss how, when and why tumor HLA-I losses occur. We also provide evidence for the negative impact of tumor HLA-I loss in current cancer immunotherapies, with the focus on reversible (‘soft’) and irreversible (‘hard’) HLA-I defects. Addresses 1 Servicio de Analisis Clinicos e Inmunologia, UGC Laboratorio Clinico, Hospital Universitario Virgen de las Nieves, 18014 Granada, Spain 2 Instituto de Investigacion Biosanitaria ibs, 18014 Granada, Spain 3 Departamento de Bioquimica, Biologia Molecular e Inmunologia III, Facultad de Medicina, Universidad de Granada, Spain Corresponding author: Garrido, Federico (federico.garrido.
[email protected]) Current Opinion in Immunology 2018, 51:123–132 This review comes from a themed issue on Tumour immunology Edited by Jewett
https://doi.org/10.1016/j.coi.2018.03.006 0952-7915/ã 2018 Elsevier Ltd. All rights reserved.
www.sciencedirect.com
Introduction After decades of investigation of different types of MHC-I alterations in human and experimental tumors by different research groups, including ours [1–10], it has become evident that the MHC-I/HLA-I altered phenotypes frequently observed in cancer cells are not just a random epiphenomenon associated with tumor progression, but represent a crucial event that determines the fate of tumor development: either its rejection or escape from T-cell mediated response. It is well established that cytotoxic T lymphocytes play a major role in controlling and destroying nascent tumors [11,12]. These effector cells are in the focus of the clinical responses to the traditional and more recent types of immunotherapy, including monoclonal antibodies against immune checkpoint inhibitors involved in the regulation of T-cell cytotoxicity [13,14,15,16,17]. T cells require interaction with tumor MHC-I molecules to recognize tumor antigens processed and presented as small peptides. Therefore, any alteration in the expression of MHC-I undoubtedly will have a profound implication in the primary tumor growth and in the metastatic colonization. A Darwinian type of T-cell mediated immune selection is responsible for the escape of HLA-I deficient tumor cells. The classical idea of ‘Generation of Diversity [GOD] and Selection’ can be also applied to the tumor development. An explosion of MHC-I diversity takes place in the primary tumor and is followed by an immune selection and destruction of tumor cells by cytotoxic T-lymphocytes via recognition of the MHC-I/tumor peptide complex [18]. As a result, primary tumor becomes composed mostly of MHC-I negative/deficient tumor cells. During this process of cancer progression tumor tissue is undergoing remodeling and is being re-shaped by the immune microenvironment. Cancer immunotherapy is currently the most rapidly advancing area of clinical oncology due to durable responses observed in some patients treated with different types of monoclonal antibodies directed against molecules regulating T-cell activity. However, little attention is being paid to the status of tumor HLA-I expression before, during and after immunotherapy. The success of cancer immunotherapy relies on better understanding the mechanisms of cancer immune escape and the dynamic Current Opinion in Immunology 2018, 51:123–132
124 Tumour immunology
changes of the tumor microenvironment during the transition from pre-existing immune response to a therapyinduced immune response. For many years our laboratory has been actively engaged in defining altered HLA-I phenotypes in human tumors and the underlying molecular mechanisms of HLA loss or downregulation. We also have been investigating how these alterations influence the response to cancer immunotherapy by correlating them with the regression or progression of post-therapy metastatic lesions [19,20]. Recently, we observed in lung carcinoma that positive HLA-I expression on tumor cells correlate with massive diffuse infiltration of tumor mass with T-lymphocytes, while loss of tumor HLA-I expression is accompanied by the absence of CD8+ T cells within the primary tumor, which is surrounded by a fibrous stroma resembling a granuloma-like structure [21,22]. We strongly believe that it is necessary to analyze tumor HLA-I expression and monitor HLA-I changes taking place during immunotherapy to understand how, when and why the MHC/HLA alterations occur, as well as to improve the efficacy of immunotherapy and minimize resistance to treatment.
How the MHC/HLA-I alterations develop It is well established that there are multiple molecular mechanisms responsible for the generation of HLA-I altered tumor escape phenotypes. In theory, any step required for the assembly of a functional HLA-I molecule can be altered causing HLA-I loss and providing an escape route from T-cell recognition. Nevertheless, there are some common well characterized mechanisms shared by different types of malignancy and specific alterations distinctive for some particular cancers. Loss of heterozygosity (LOH) in chromosome 6 and 15 (LOH-6 and LOH-15), which include the HLA and the b2-microglobulin (b2m) regions, respectively, is the most widespread mechanism. It is responsible for HLA-I haplotype loss, which leads to expression of only three HLA alleles on the tumor cell surface. We have described this mechanism in tumors of different histological type, including lung carcinoma [21,23–28]. In some tumors (melanoma, MSI-H colorectal carcinoma and bladder cancer) this chromosome instability is a frequent and early event that can later be complemented by other abnormalities, such as HLA-I transcriptional downregulation or b2m mutation, altogether leading to a total HLA-I loss [24,29,30]. We favor the idea that the frequency of LOH-6 and LOH-15 affecting the HLA and b2m genes is greatly underestimated. Recent data obtained in our laboratory using GCH arrays indicate that tumor cells with apparently ‘normal’ HLA expression, yet harbor HLA gene microdeletions that had been previously undetected by immunohistology and classical microsatellite amplification analysis (unpublished data). In addition, the modern NGS technology that detects HLA-I and b2m mutations in recurrent metastatic lesions, is not informative in case Current Opinion in Immunology 2018, 51:123–132
of macro or micro deletions of genetic material in chromosome 6 and 15. Different altered HLA-I phenotypes observed in human tumors were summarized long time ago by our group [5,7,31] and recently revisited [32]. They include: Tumor HLA phenotype no. I: HLA class I total loss. It is observed in all analyzed tumors with different frequencies and different molecular mechanisms. In melanoma and in MSI-H colorectal carcinoma it has been reported in around 15% of the tumors and is associated with b2m loss due to LOH-15 and a mutation in the second allele of the b2m gene. Similar mechanisms have been reported in B-cell lymphomas [33]. In MSS colorectal cancer the molecular mechanisms appears to be different and caused by a downregulation of the antigen presentation machinery (APM) components [34]. In breast and prostate carcinoma total HLA-I loss is observed in more than 50% of tumor samples analyzed [35,36]. HLA-I loss associated with a coordinated transcriptional downregulation of APM and HLA-I/ b2m genes has been observed in prostate, bladder and lung carcinomas [36,37,21]. Tumor HLA phenotype no. II: HLA haplotype loss. It frequently occurs as a result of the loss of genetic material at chromosome 6 (LOH-6) [28]. Often a duplication of the remaining chromosome masks the loss of heterozygosity, which in that case can be detected only by a direct HLA typing using DNA isolated from microdissected tumor tissue. This is a common molecular mechanism that occurs at early stages of tumor development and has been reported in about 30% of different types of malignancy, including melanoma, colorectal, lung, pancreas, laryngeal, breast, and bladder cancer [23–28]. These are irreversible (‘hard’) alterations, which cannot be restored with any type of conventional therapy, including immunotherapy. Tumor HLA phenotype no. III: selective HLA-A, HLA-B or HLA-C locus downregulation. It is a common mechanism observed in different human tumors and tumor cell lines [2,38]. Generally, it can be reverted by cytokines, which increase HLA-I gene transcription and lead to a subsequent upregulation on cell surface. Tumor HLA phenotype no. IV: single HLA-I allele losses. Mutations in genes coding for HLA-I alleles produce loss of the cell surface expression of one single HLA-A, HLA-B or HLA-C allele. It has been described in cervical [10], prostate [39], colorectal [40] tumors and in melanoma cell lines [41]. Tumor HLA phenotype no. V: a compound phenotype. It is a frequent phenotype observed in human tumors, when more than one molecular mechanism contribute to the generation of the aberrant HLA-I expression. In certain conditions, a combination of HLA haplotype loss with a transcriptional downregulation of HLA locus www.sciencedirect.com
The transition from HLA-I positive to HLA-I negative primary tumors: the road to escape from T-cell responses Aptsiauri, Ruiz-Cabello and Garrido 125
leaves only a single HLA-I allele on cancer cell surface [5,7]. We discovered that HLA-I haplotype loss together with a transcriptional downregulation of HLA-A, HLA-B and HLA-C genes is a common mechanism responsible for a total HLA-I loss in approximately 60% of small cell lung carcinoma samples studied [21]. Tumor HLA phenotype no. VI: Resistance to Interferon. IFN-g secretion by cytotoxic T-cells is critical for tumor rejection and can induce tumor HLA-I upregulation. Sometimes cancer cells develop resistance to IFN due to a variety of genetic lesions affecting the IFN-g or IFN-a signaling pathway. This alteration has been described in different tumor cell lines [42] and in recurrent melanoma lesions after immunotherapy [43]. Based on the underlying molecular mechanism and the ability to recover normal HLA-I expression by cytokines or after immunotherapy, these different molecular mechanisms responsible for HLA-I alterations can be classified in two major groups, namely, reversible (‘soft’) or irreversible (‘hard’) alterations. This classification has profound clinical implications in the prediction of patients’ response to cancer immunotherapy [44]. We have reported that melanoma regression was more frequent in patients with reversible regulatory tumor HLA-I alterations than in those with irreversible HLA-I gene defects [19]. Cytokine-mediated reversion of tumor HLA-I expression can restore immunogenicity and contribute to T-cell mediated rejection. In some cases interferons work together with other anti-cancer drugs in upregulating HLA-I expression. On this context, an upregulation of HLA-I expression by interferon and MEK1/2 inhibitor selumetinib was recently reported in papillary thyroid tumors with ‘soft’ mechanism of HLA loss [45]. Despite the enormous progress, there is a long way to go before the different molecular mechanisms responsible for HLA-I alterations are precisely defined in different tumor types. For example, the mechanisms responsible for total HLA-I loss [phenotype no. I] in about 60% of breast cancer patients, in 50% of prostate cancer patients, in 15% of patient with laryngeal cancer, or in 40% of patients with gastric cancer is still to be identified [5,7,32]. We have recently reported that the tumor suppressor Fhit positively regulates MHC-I expression on cancer cells [46]. Another molecule, a transcription factor NLRC5 recently reported as a candidate responsible for HLA-I expression regulation, has been identified in different tumors [47]. These transcriptional regulators could be used to correct HLA-I ‘soft’ lesions.
When the HLA-I alterations occur Based on our earlier observations, we believe that HLA-I loss is an early event that occurs when the primary tumor is only a few millimeters in diameter and has already www.sciencedirect.com
induced T-cell mediated immune selection. However, this hypothesis still needs a complete experimental and clinical confirmation [21,48,49]. It is becoming apparent that the immunological status of the host plays a major role in selecting the MHC/HLA-deficient tumor cells. In this context, we have data obtained in a H-2 MHC-I negative clone of the GR9 mouse fibrosarcoma indicating that spontaneous lung metastasis produce different MHC-I phenotypes depending on the host immune system: H-2-negative lesions in immune competent mice and H-2-positive metastases in mice lacking T-cells [50,51]. It is likely that recruitment of T lymphocytes and tumor infiltration is influenced by many factors, including tumor antigenicity. The co-existence of HLA-I positive, heterogeneous and completely HLA-I negative tumor lesions in the same cancer patient could represent different stages during tumor development indicating an ongoing T-cell mediated immune selection.
Why the MHC/HLA-I alterations occur: is the transition from HLA-I positive to HLA-I negative tumors during cancer progression inevitable? Generation of MHC-I deficient tumor cells is believed to be a consequence of T-cell immune selection in primary heterogeneous tumors. During this process highly antigenic MHC-I positive tumor cells are eliminated, while cells with low or negative MHC-I expression are selected for survival. We observed a diversity of MHC-I phenotypes in mouse fibrosarcomas induced by methylcholanthrene and adapted to tissue culture without a single passage [52]. This initial stage of the ‘explosion of MHC-I diversity’ shifts to the outgrowth of tumor lesions composed only of MHC-I negative cancer. This transition is directed by the T-cell mediated host immune response [18]. Similar findings were nicely demonstrated by Boesen and colleagues in T-cell deficient mice [11]. In a case of metastatic melanoma we were able to illustrate a chronological sequence of the immune escape of HLAI-negative tumor cells with LOH-15 and b2m gene mutation in several successive lesions. This case clearly demonstrates a transition from HLA-I positive/heterogeneous to HLA-I negative tumor phenotype, which was linked to the presence of tumor infiltrating CD8+ T-cells [53]. Similarly, another melanoma study also demonstrated how poorly immunogenic tumor phenotypes with HLA-I defects evolve during the disease progression due to the early emergence of an inactivating mutation in one allele of the b2m gene and the simultaneous loss of the other allele by a deletion in chromosome 15 (LOH-15) [54]. T-cell immune selection may not be always responsible for the appearance of HLA-I negative tumor cells. It is well established that different viral or cellular oncogene products can also cause HLA-I downregulation as a mechanism responsible for cancer escape and growth, Current Opinion in Immunology 2018, 51:123–132
126 Tumour immunology
as it has been shown in cervical or breast carcinoma [55,56]. In these malignancies, HLA-I loss probably cannot be exclusively attributed to T-cell-mediated immune selection, and may explain the observed poor T-cell infiltration. Similarly, tumor immune escape implicated in renal cell carcinoma (RCC) is likely to be T-cell independent, since according to some reports RCC cells express HLA-I molecules while the renal tubules, normal tissue from which the tumor derive, are HLA-I negative [57]. Nevertheless, most tumors derive from HLA-I positive normal epithelia [3]. The absence of HLA-I expression in the majority of tumors can be defined as a mandatory step during tumor development since the frequency of this phenomenon is very high, reaching up to 95% of analyzed cases according to published literature [3,5,10]. It is also widely acknowledged that T-lymphocytes are playing a crucial role in destroying nascent tumors, and without MHC/HLA-I expression this destruction cannot take place [11,22].
Changes in tumor tissue architecture following anti-tumor T-cell mediated responses: from HLA-I-positive/Th1-type to HLA-I-negative/Th2-type granuloma-like tumor structure The tumor microenvironment, including the immune infiltrate and tissue organization, undergoes dynamic changes during tumor growth and progression. As a result of the T-cell anti-tumor immune response and cancer immunoediting, there is a transition from a highly immunogenic to a low immunogenic tumor phenotype. This includes a gradual loss of antigen-presentation ability of tumor cells due to the accumulation of HLA-I alterations. It is also characterized by changes in the immunological composition, the density, functional state and distribution of the leukocyte infiltrate of the tumor, all leading to certain tissue re-organization. Tumor-infiltrating lymphocytes have been associated with favorable clinical outcomes in various solid tumors [58]. We have recently reported a positive correlation between tumor HLA-I expression, immune infiltration, and tumor tissue architecture in lung cancer [21,22,32]. Figure 1 summarizes our current view on these changes. It shows a schematic representation of the process of a gradual loss of tumor HLA-I expression and tumor tissue reorganization linked to the presence of cytotoxic T-cells. At the beginning, tumor cells are MHC/HLA-I positive and are densely infiltrated by T-lymphocytes normally associated with a Th1 type of cytokine production (‘permissive’ Phase I) (Figure 2a). This immune infiltration is responsible for the destruction of HLA-I positive cancer cells and selection of MHC/HLA-I negative cells to proliferate. When T-cell response fails to destroy all tumor cells, the host tries to isolate tumor by building up a stromal barrier around it, which restrains the ‘jobless’ T-lymphocytes Current Opinion in Immunology 2018, 51:123–132
and other immune cells, such as macrophages, from entering tumor mass. This encapsulated tumor structure resembles a classical granuloma formed in response to infection with bacteria or parasites, which is characterized by a Th2 type of immune response and M1/M2 macrophage polarization (non-permissive/encapsulated Phase II) [59,60] (Figure 2b). We found a similar M1/M2 macrophage polarization in lung cancer typical for a Th2 type immune response [21,32]. Different authors, including ourselves, reported that the degree of tumor infiltration with T-lymphocytes directly correlates with the number of HLA-I positive tumor cells in lung and pancreatic cancer [21,61,62], suggesting that there is a continuous T-cell activation and selection of HLA-I negative tumor cells (Figure 3). An important question arises as how widespread among different tumors is the transition from the ‘permissive’ Phase I to the ‘encapsulated’ Phase II, or a transition from a Th1 type response in HLA-I positive tumors to a Th2 type immune landscape characterized by a formation of stromal capsule around HLA-I negative tumor nest promoting M2 polarization (Figures 1 and 2). To answer this question further studies deciphering the precise and sequential cellular and molecular events that lead to this transition are necessary.
T-cell mediated immunotherapy selects cancer cells harboring irreversible/‘hard’ HLA-I defects Cancer immunotherapy has historically used a variety of biological products that are potentially able to boost antitumor T-cell mediated immune responses, which include BCG, IL-2, autologous tumor vaccines, dendritic cells loaded with peptides, tumor peptides alone, polysaccharide K[PSK], adoptive cell therapy with TILs, or checkpoint inhibitors [63,64,65]. All these therapies have demonstrated various degrees of clinical benefit in metastatic cancer patients, including complete and partial responses with both regressing and progressing lesions. In most cases it is not possible to define why some metastatic lesions progress and others regress. We analyzed in detail the HLA-I expression in progressing and regressing melanoma metastatic lesions in patients treated with autologous vaccination [19,20] and found a strong and direct correlation between high tumor HLA-I expression and regression of the metastasis, whereas the progressing lesions were harboring irreversible HLA-I defects. Furthermore, genome-wide gene expression analysis of regressing and progressing melanoma lesions showed that only 146 genes were differentially expressed in both types of lesions. These genes include IFN and HLA genes suggesting the crucial role they play in tumor rejection as a part of an immunological constant of rejection [20,66]. The predictive value of the molecular mechanisms leading to HLA-I aberrant www.sciencedirect.com
The transition from HLA-I positive to HLA-I negative primary tumors: the road to escape from T-cell responses Aptsiauri, Ruiz-Cabello and Garrido 127
Figure 1
Transition from HLA-I-positive to HLA-I-negative pattern during tumor developement PERMISSIVE PHASE I
Tumor Initiation
T cell infiltration
T cell immuno-selection of HLA-I-negative tumor cells
Th-1 like Response HLA-I-positive
HLA-I positive Tumor Cell
HLA-I-positive
HLA-I negative Tumor Cell
ENCAPSULATED PHASE II
Th-2 like Response HLA-I-heterogeneous HLA-I + / –
HLA-I-negative Granuloma-like tumor structure
CTL Current Opinion in Immunology
Tumor tissue reorganization during T-cell mediated immune response. In phase I the tumor is heavily infiltrated by T-cells, including CD4+, CD3+, CD8+ lymphocytes and CD206+ M1 macrophages. Tumor is heterogeneous with both HLA-I positive and HLA-negative cells, and has a diffused structure of the parenquima and stroma typical for a Th1-type immune response and T-cell mediated cytotoxicity. In phase II the process of T-cell mediated destruction of HLA-I positive tumor cell finishes and the ‘jobless’ T-cells reside in the stroma that surrounds the tumor nest. In this phase the escaped tumor cells are all HLA-I negative and are encapsulated by the stroma. This structure resembles a granuloma that is formed around pathogens (bacteria or parasites) as a part of a Th2-type of immune response.
expression becomes essential for circumventing cancer resistance to immunotherapy. Interestingly, we have just found that opposite to tumor HLA-I phenotype, PDL-1 expression in cancer cells is not guided by T-cell immunoselection, and its expression is random in the analyzed lung cancer tissue samples [unpublished data]. HLA-I expression may be upregulated by immunomodulating treatment only in the case of reversible/‘soft’ molecular HLA-I aberrations [67]. In this context, we have recent indications that antibodies against EGFR used in experimental tumor models can upregulate MHC-I molecules [68]. In contrast, HLA-loss variants with irreversible/ ‘hard’ defects escape from T-cell recognition and develop progressing therapy-resistant metastasis. This assumption has been recently confirmed in a study in which b2m mutation was identified among frequent mutations associated with acquired resistance to PD-1 blockade [43]. Considering that NK cells are capable of killing MHC-I negative tumors, a question arises ‘why NK cells are not www.sciencedirect.com
killing these MHC-I-negative targets?’ This question still does not have an answer. We know that NK cells do not infiltrate MHC-I negative tumors; they remain outside the tumor nest, which suggests that these tumors develop inhibitory mechanisms that block NK cell activation and migration [22]. Most NK cells are defective in cancer patients, even at early stages of tumorigenesis, which implies that MHC-I-negative tumor clones are not able to be fully eliminated. Loss of both NK and T cell functions which occurs during the cancer progression will likely contribute to the survival of tumors with no/low MHC-I. The common ‘hard’ alterations include HLA-I haplotype loss associated with LOH-6 or LOH-15 together with mutations/deletions in the remaining copy of b2m gene, or mutations in IFN signaling pathways [44]. The recovery of MHC/HLA-I expression in immunotherapy-resistant recurrent tumors with ‘hard’ HLA-I Current Opinion in Immunology 2018, 51:123–132
128 Tumour immunology
Figure 2
(a)
HLA-I
HLA-I positive tumor
CD3
(b)
HLA-I
CD45
CD8
HLA-I negative tumor
CD3
CD45
CD8
Current Opinion in Immunology
Immunohistochemistry of a lung carcinoma tissue immunostained with anti-HLA-I, anti-CD45, anti-CD3 and anti-CD8 antibodies: (a) HLA-I positive tumor with diffused tumor architecture and tumor infiltrating T-cells (Permissive Phase I). (b) HLA-I negative encapsulated tumor (Phase II) with tumor T-cells in the stroma surrounding tumor nests.
alterations or the development of novel strategies to target HLA-I deficient cancer cells is a major challenge for the future of cancer treatment. One possibility could be gene therapy aimed to restore tumor HLA-I expression and T-cell recognition by replacing defective b2m of HLA-I heavy chain genes [69–71].
Current Opinion in Immunology 2018, 51:123–132
Conclusions Tumor immunotherapy has reached fascinating times, similar to when BCG was first locally instilled in 1976 to treat superficial bladder tumors [72], or when IL-2 was used for the first time in 1987 in metastatic melanoma [63], or when the first tumor peptide was systemically injected in melanoma patients alone or as www.sciencedirect.com
The transition from HLA-I positive to HLA-I negative primary tumors: the road to escape from T-cell responses Aptsiauri, Ruiz-Cabello and Garrido 129
Figure 3
Correlation between HLA class I expression and tumor infiltrating T Lymphocytes in tumor tissue nests 100 90 80
Tumor infiltrating T lymphocytes
70 60 50 40
(mean cells/field) 30 20 10 0
Positive
Heterogeneous
Negative
HLA class I expression
0
TIME Current Opinion in Immunology
Correlation between tumor HLA-I expression and infiltration with T-lymphocytes. HLA-I antigen expression in tumors directly correlates with the degree of tumor T-cell infiltration inside the tumor nests.
a part of a dendritic cell vaccine [13,14]. Current immune checkpoint therapies against cancer are in the center of attention of oncologists and immunologist due to their clear clinical benefit and positive responses in patients [15,16,17], although relapses occur and the reason for that is still a ‘black box’ in many clinical trials. A growing awareness exists of the fact that the success of immunotherapy depends on the induction of a strong and longlasting anticancer immune response leading to tumor eradication. However, tumor cells can evade immune surveillance and destruction by cytotoxic T-cells by various strategies, including the aberrant expression of HLAI antigens (due to accumulating mutations in HLA-I/b2m genes) and defective IFN signal transduction pathway [44,53,54]. In order to eliminate cancer, novel immunotherapy strategies must correct these defects and restore T-cell recognition and elimination of the tumor cells. Importantly, all types of immunotherapy, including checkpoint inhibitors, depend on the level of HLA-I expression on cancer cells, but the majority of current cancer immunotherapy protocols do not include routine examination of tumor HLA-I expression and anti-tumor activated T-cells remain unable to see HLA-I negative tumor targets. It is difficult to understand why HLA-I molecules are not properly analyzed in post-therapy recurrent cancer lesions, since it is already accepted that HLA-I-negative tumor cell variants are T-cell immunoselected and that irreversible/‘hard’ HLA-I alterations accumulate in the recurrent progressing metastases www.sciencedirect.com
[43,44,53,54]. This paradox is hard to explain considering the amount of resources that are dedicated to improve our understanding of the tumor immune escape mechanisms [73,74]. HLA analysis in tumor tissues is a missing parameter that should be implemented [75]. Cooperation of pathologists with clinical oncologists, surgeons and immunologist is necessary to explore the immune contexture of tumors and to analyze tumor HLA-I defects in order to have reliable predictive biomarkers that tailor individual cancer treatment strategies and to monitor a response to anticancer therapies. However, it must be acknowledged that this is not an easy task due to the complexity of the HLA system and difficulties to collect fresh frozen tumor samples before, during and after the therapy. We should remember that MHC was first discovered in mice immunized with allogenic tumors in 1937 [76]. Now, 80 years later, the role of MHC in cancer is being revisited and requires urgent attention in order to override immune escape mechanisms and improve clinical efficacy of cancer immunotherapy.
Acknowledgements We would like to thank Dr Monica Bernal for helping to prepare the figures, and Dr Francisco Perea for providing the images of tumor tissue immunolabeling. This work was supported by grants from the Instituto de Salud Carlos III co-financed by FEDER funds (EU) (PI11/01386, PI14/ 01978, PI 16/00752, RETIC RD 06/020, RD09/0076/00165, PT13/0010/ 0039] and Junta de Andalucı´a in Spain [Group CTS-143]. Current Opinion in Immunology 2018, 51:123–132
130 Tumour immunology
References and recommended reading Papers of particular interest, published within the period of review, have been highlighted as: of special interest of outstanding interest 1.
Festenstein H, Garrido F: MHC antigens and malignancy. Nature 1986, 322:502-503.
2.
Lopez-Nevot MA, Esteban F, Ferron A, Gutierrez J, Oliva MR, Romero C, Huelin C, Ruiz-Cabello F, Garrido F: HLA class I gene expression in human primary tumors and autologous metastasis: demonstration of selective losses of HLA antigens on colorectal, gastric and laryngeal carcinomas. Br J Cancer 1989, 59:221-226.
3.
Garrido F, Cabrera T, Concha A, Glew S, Ruiz-Cabello F, Stern PL: Natural history of HLA expression during tumor development. Immunol Today 1993, 14:491-499.
4.
5.
Bodmer W, Browning MJ, Krausa P, Rowan A, Bicknell DC, Bodmer J: Tumour escape from immune response by variation in HLA expression and other mechanisms. Ann N Y Acad Sci 1993, 690:42-49. Garrido F, Ruiz-Cabello F, Cabrera T, Perez-Villar JJ, LopezBotet M, Duggan-Keen M, Stern PL: Implications for immunosurveillance of altered HLA class I phenotypes in human tumors. Immunol Today 1997, 18:89-95.
6.
Marincola FM, Jafee EM, Hicklin DJ, Ferrone S: Escape of human solid tumors from T cell recognition: molecular mechanisms and functional significance. Adv Immunol 2000, 74:181-273.
7.
Garrido F, Algarra I: MHC antigens and tumor escape from immune surveillance. Adv Cancer Res 2001, 83:117-158.
8.
Seliger B, Cabrera T, Garrido F, Ferrone S: HLA class I antigen abnormalities and immune escape by malignant cells. Semin Cancer Biol 2002, 12:3-13.
9.
Garrido F, Klein E: MHC antigen expression. I. Human tumors. In Seminars in Cancer Biology, vol 2-1. Edited by Klein E, Garrido F . W.B. Saunders Scientific Publications; 1991:1-2.
10. Koopman LA, Corver WE, Van del Slik AR, Giphart MJ, Fleuren GJ: Multiple genetic alterations cause frequent and heterogeneous human histocompatibility leukocyte antigen class I loss in cervical cancer. J Exp Med 2000, 191:961-976.
16. Robert L, Tsoi J, Wang X, Emerson R, Homet B, Chodon T, Mok S, Huang RR, Cochran A, Comin-Anduix B et al.: CTLA4 blockade broadens the peripheral T cell receptor repertoire. Clin Cancer Res 2014, 20:2424-2432. 17. Buferne M, Chasson L, Grange M, Mas A, Arnoux F, Bertuzzi M, Naquet P, Leserman L, Schmitt-Verhulst AM, Auphan-Anezin N: IFN-g producing CD8+ cells modified to resist major immune checkpoints induce regression of MHC class I-deficient melanomas. Oncoimmunology 2015, 4:e974759. This paper reveals that regression of experimental melanoma with reduced MHC-I expression can be achieved by CD8+ T cells transfected with STAT5CA, which produce IFN-g and subsequently increase tumor MHC-I. These CD8+ T cells also express PD-1 and upregulate PDL-1 on melanoma cells. Despite upregulation of this immunosuppressive pathway, efficient IFN-g production in the melanoma microenvironment was found associated with resistance of STAT5CA-expressing CD8+ T cells to inhibition both by PD-1/PDL-1 engagement. 18. Garrido F, Romero I, Aptsiauri N, Garcia-Lora AM: Generation of MHC class I diversity in primary tumors and selection of the malignant phenotype. Int J Cancer 2016, 138:271-280. 19. Carretero R, Romero JM, Ruiz-Cabello F, Maleno I, Rodriguez F, Camacho FM, Real LM, Garrido F, Cabrera T: Analysis of HLA class I expression in progressing and regressing metastatic melanoma lesions after immunotherapy. Immunogenetics 2008, 60:439-447. 20. Carretero R, Wang E, Rodriguez AI, Reinboth J, Ascierto ML, Engle AM, Liu H, Camacho F, Marincola FM, Garrido F et al.: Regression of melanoma metastases after immunotherapy is associated with activation of antigen presentation and interferon-mediated rejection genes. Int J Cancer 2012, 131:387-395. This paper shows that regression of metastatic lesions after immunotherapy is associated with an increase transcription of genes involved in antigen presentation, HLA and IFN. These data were obtained when a whole genome transcription analysis was performed comparing progressing and regressing melanoma metastatic lesions in a mixed responder patient. 21. Perea F, Bernal M, Sanchez-Palencia A, Carretero J, Torres C, Bayarri C, Gomez-Morales, Garrido F, Ruiz-Cabello F: The absence of HLA class I expression in non-small cell lung cancer correlates with the tumor tissue structure and the pattern of T cell infiltration. Int J Cancer 2017, 140:888-899. This paper indicates that the tumor tissue structure change dramatically when there are TILs in HLA-I positive tumors as compare when there are not in HLA-I negative tumor tissue samples.
11. Boesen M, Svane IM, Engel AM, Rygaard J, Thomsen AR, Werdelin O: CD8+ T cells are crucial for the ability of congenic normal mice to reject highly immunogenic sarcomas induced in nude mice with 3-methylcholantrene. Clin Exp Immunol 2000, 121:210-215.
22. Garrido F, Perea F, Bernal M, Sanchez-Palencia A, Aptsiauri N, Ruiz-Cabello F: The escape of cancer from T cell mediated immune surveillance: HLA class I loss and tumor tissue architecture. Vaccines 2017, 5:7.
12. Romero P, Coulie P: Adaptive T-cell immunity and tumor antigen recognition. In Tumor Immunology and Immunotherapy. Edited by Rees RC. Oxford University Press; 2014:1-14.
23. Maleno I, Cabrera CM, Cabrera T, Paco L, Lopez-Nevot MA, Collado A, Ferron A, Garrido F: Distribution of HLA class I altered phenotypes in colorectal carcinomas: high frequency of HLA haplotype loss associated with loss of heterozygosity in chromosome region 6p21. Immunogenetics 2004, 56:244-253.
13. Marchand M, Van Baren N, Weynan P, Brichard V, Dreno B, Tessier M-H, Rankin E, Parmiani G, Arienti F, Humblet Y et al.: Tumor regressions observed in patients with metastatic melanoma treated with an antigenic peptide encoded by gene MAGE-3 and presented by HLA-A1. Int J Cancer 1999, 80:219-230. 14. Nestle FO, Alijagic S, Gilliet M, Sun Y, Grabbe S, Dummer R, Burg G, Shadendorf D: Vaccination of melanoma patients with peptide or tumor lysate-pulsed dendritic cells. Nat Med 1998, 4:328-332. 15. Kvistborg P, Philips D, Kelderman S, Hageman L, Ottensmeier C, Joseph-Pietras D, Welters M, van de Burg S, Kapiteijn E, Michielin O et al.: Anti CTL-4 therapy broadens the melanomareactive CD8+ T cell response. Sci Transl Med 2014, 6:1-9. This paper provides a strong evidence of anti-CTLA-4 therapy-enhanced T cell priming as a component of the clinical mode of action. Comparison of pre-treatment and post-treatment T cell melanoma peptide-specific reactivity in peripheral blood mononuclear cell samples from melanoma patients demonstrated that the anti-CTLA-4 treatment induces a significant increase in the number of melanoma-specific CD8 T-cell responses, but does not boost the pre-existing virus-specific and melanoma-specific T cell. Current Opinion in Immunology 2018, 51:123–132
24. Maleno I, Aptsiauri N, Cabrera T, Gallego A, Paschen A, LopezNevot MA, Garrido F: Frequent loss of heterozygosity in the b2microglobulin region of chromosome 15 in primary human tumors. Immunogenetics 2011, 63:65-71. 25. Maleno I, Lopez-Nevot MA, Cabrera T, Salinero J, Garrido F: Multiple mechanisms generate HLA class I altered phenotypes in laryngeal carcinomas: high frequency of HLA haplotype loss associated with loss of heterozigocity in chromosome region 6p21. Cancer Immunol Immunother 2002, 51:389-396. 26. Maleno I, Romero JM, Cabrera T, Paco L, Aptsiauri N, Cozar JM, Tallada M, Lopez-Nevot MA, Garrido F: LOH at 6p21.3 region and HLA class I altered phenotypes in bladder carcinomas. Immunogenetics 2006, 58:503-510. 27. Ramal LM, Feenstra M, van der Zwan AW, Collado A, LopezNevot MA, Tilanus M, Garrido F: Criteria to define HLA haplotype loss in human tumors. Tissue Antigens 2000, 55:443-448. 28. Jimenez P, Canton J, Collado A, Cabrera T, Serrano A, Real LM, Garcia A, Ruiz-Cabello F, Garrido F: Chromosome loss is the www.sciencedirect.com
The transition from HLA-I positive to HLA-I negative primary tumors: the road to escape from T-cell responses Aptsiauri, Ruiz-Cabello and Garrido 131
most frequent mechanism contributing to HLA haplotype loss in human tumors. Int J Cancer 1999, 83:91-97. 29. del Campo AB, Kyte JA, Carretero J, Zinchencko S, Mendez R, Gonzalez-Aseguinolaza G, Ruiz-Cabello F, Aamdal S, Gaudernack G, Garrido F et al.: Immune escape of cancer cells with b2-microglobulin loss over the course of metastatic melanoma. Int J Cancer 2014, 134:102-113. 30. Paschen A, Mendez RM, Jimenez P, Sucker A, Ruiz-Cabello F, Song M, Garrido F, Schadendorf D: Complete loss of HLA class I antigen expression on melanoma cells: a result of successive mutational events. Int J Cancer 2003, 103:759-767. 31. Garrido F, Cabrera T, Lopez-Nevot MA, Ruiz-Cabello F: HLA class I antigens in human tumors. Adv Cancer Res 1995, 67:155-195. 32. Garrido F, Ruiz-Cabello F, Aptsiauri N: Rejection versus escape: the tumor MHC dilemma. Cancer Immunol Immunother 2017, 66:259-271. 33. Challa-Malladi M, Lieu YK, Califano O, Holmes AB, Bhagat G, Murty VV, Dominguez-Sola D, Pasqualucci L, Dalla Favera R: Combine genetic inactivation of b2 microglobuline and CD58 reveals frequent escape from immune recognition in diffuse large B cell lymphoma. Cancer Cell 2011, 20:728-740. 34. Cabrera C, Jimenez P, Cabrera T, Esparza C, Ruiz-Cabello F, Garrido F: Total loss of MHC class I antigens in colorectal tumors can be explained by two molecular pathways: b2 microglobuline inactivation in MSI-positive tumors and LMP7/ TAP2 downregulation in MSI-negative tumors. Tissue Antigens 2003, 61:211-219. 35. Cabrera T, Fernandez MA, Sierra A, Garrido A, Herruzo A, Escobedo A, Fabra A, Garrido F: High frequency of altered HLA class I phenotypes in invasive breast carcinomas. Hum Immunol 1996, 50:127-134. 36. Carretero FJ, Del Campo AB, Flores-Martı´n JF, Mendez R, Garcı´aLopez C, Cozar JM, Adams V, Ward S, Cabrera T, Ruiz-Cabello F, Garrido F, Aptsiauri N: Frequent HLA class I alterations in human prostate cancer: molecular mechanisms and clinical relevance. Cancer Immunol Immunother 2016, 65:47-59. 37. Romero JM, Jimenez P, Cabrera T, Cozar JM, Pedrinaci S, Tallada M, Garrido F, Ruiz-Cabello F: Coordinated downregulation of the antigen presentation machinery and HLA class I/b2-microglobuline complex is responsible for HLA-ABC loss in bladder cancer. Int J Cancer 2005, 113:605-610. 38. Me´ndez R, Rodrı´guez T, Del Campo A, Monge E, Maleno I, Aptsiauri N, Jime´nez P, Pedrinaci S, Pawelec G, Ruiz-Cabello F, Garrido F: Characterization of HLA class I altered phenotypes in a panel of human melanoma cell lines. Cancer Immunol Immunother 2008, 57:719-729. 39. Jimenez P, Cabrera T, Mendez R, Esparza C, Cozar JM, Tallada M, Lopez-Nevot MA, Ruiz-Cabello F, Garrido F: A nucleotide insertion in exon 4 is responsible for the absence of expression of an HLA-A *0301 allele in a prostate carcinoma cell line. Immunogenetics 2001, 53:606-610. 40. Browning M, Petronzelli F, Bicknell D, Krausa P, Rowan A, Tonks S, Murray N, Bodmer J, Bodmer W: Mechanisms of loss HLA-I expression in colorectal tumor cells. Tissue Antigens 1996, 47:364-371. 41. Wang Z, Marincola F, Rivoltini L, Parmiani G, Ferrone S: Selective histocompatibility leucocyte antigen (HLA)-A2 loss caused by aberrant pre-mRNA splicing in 624MEL28 melanoma cells. J Exp Med 1999, 190:205-215. 42. Abril E, Mendez RE, Garcia A, Serrano A, Cabrera T, Garrido F, Ruiz-Cabello F: Characterization of a gastric tumor cell line defective in MHC class I inducibility by both alpha- and gamma-interferon. Tissue Antigens 1996, 47:391-398. 43. Zaretsky JM, Garcia-Diaz A, Shin DS, Escuin-Ordinas H, Hugo W, Hu-Lieskovan S, Torrejon DY, Abril-Rodriguez G, Sandoval S, Barthly et al.: Mutations associated with acquired resistance to PD-1 blockade in melanoma. N Engl J Med 2016, 375:819-829. This report shows that mutations accumulate in relapsed metastatic lesions after treatment with immune checkpoint monoclonal antibodies, including mutations in b2m and IFN activation pathways. www.sciencedirect.com
44. Garrido F, Cabrera T, Aptsiauri N: ‘Hard’ and ‘soft’ lesions underlying the HLA Class I alterations in cancer cells: implications for immunotherapy. Int J Cancer 2010, 127:249-256. This paper summarizes the accumulating clinical and experimental evidences indicating that the nature of the preexisting tumor MHC-I alterations determines the metastatic potential of a malignancy and the final outcome of cancer immunotherapy. Reversible tumor MHC-I alterations (‘soft’ lesions) can be recovered leading to the activation of T cellmediated responses and tumor regression. However, MHC-I structural defects (‘hard’ lesions) such as mutations or LOH in chromosome 6 and 15 including the HLA and b2-microglobuline genes respectively cannot, ending in the relapse of metastatic lesions. 45. Angell TE, Lechner MG, Jang JK, LoPresti JS, Epstein AL: MHC class I loss is a frequent mechanism of immune escape in papillary thyroid cancer that is reversed by interferon and selumetinib treatment in vitro. Clin Cancer Res 2014, 20:6034-6044. 46. Romero I, Martinez M, Garrido C, Collado A, Algarra I, Garrido F, Garcia-Lora AM: The tumour suppressor Fhit positively regulates MHC class I expression on cancer cells. J. Pathol 2012, 227:367-379. 47. Yoshihama S, Roszik J, Downs I, Meissner TB, Vijayan S, Chapuy B, Sidiq T, Shipp MA, Lizee GA, Kobayashi KS: NLRC5/ MHC class I transactivator is a target for immune evasion in cancer. Proc Natl Acad Sci U S A 2016, 113:5999-6004 http://dx. doi.org/10.1073/pnas.1602069113. This paper identify a MHC class I transactivator that regulate the expression of MHC class I, LMP2/LMP7, TAP1 and b2 microglobuline genes. NLRC5 constitutes a new prognostic biomarker and potential therapeutic target of cancers. 48. Gutierrez J, Lopez-Nevot MA, Cabrera T, Oliva R, Esquivias J, Ruiz-Cabello, Garrido F: Class I and II HLA antigen distribution in normal mucosa, adenoma and colon carcinoma: relation with malignancy and invasiveness. Exp Clin Immunogenet 1987, 4:144-152. 49. Carretero R, Cabrera T, Saenz-Lopez P, Maleno I, Aptsiauri N, Cozar JM, Garrido F: Bacillus Calmette-Guerin immunotherapy of bladder cancer induces selection of human leucocyte antigen class I-deficient tumor cells. Int J Cancer 2001, 129:839-846. 50. Garcia-Lora A, Algarra I, Gaforio JJ, Ruiz-Cabello F, Garrido F: Immunoselection by T lymphocytes generates repeated MHC class I-deficient metastatic tumor variants. Int J Cancer 2001, 91:109-119. 51. Garcia-Lora A, Martinez M, Algarra I, Gaforio JJ, Garrido F: MHC class I-deficient metastatic tumor variants immunoselected by T lymphocytes originate from the coordinated downregulation of APM components. Int J Cancer 2003, 106:521-527. 52. Algarra I, Gaforio JJ, Garrido A, Mialdea MJ, Perez M, Garrido F: Heterogeneity of MHC-class-I antigens in clones of methylcholanthrene-induced tumors. Implications for local growth and metastasis. Int J Cancer Suppl 1991, 6:73-81. 53. del Campo AB, Kyte JA, Carretero J, Zinchencko S, Me´ndez R, Gonza´lez-Aseguinolaza G, Ruiz-Cabello F, Aamdal S, Gaudernack G, Garrido F, Aptsiauri N: Immune escape of cancer cells with b2-microglobulin loss over the course of metastatic melanoma. Int J Cancer 2014, 134:102-113. 54. Sucker A, Zhao F, Real B, Heeke C, Bielefeld N, Ma(en S, Horn S, Moll I, Maltaner R, Horn PA et al.: Genetic evolution of T-cell resistance in the course of melanoma progression. Clin Cancer Res 2014, 20:6593-6604. 55. Bartholomew JS, Glenville S, Sarkar S, Burt DJ, Stanley MA, RuizCabello F, Chengang J, Garrido F, Stern PL: Integration of highrisk human papillomavirus DNA is linked to the downregulation of class I human leukocyte antigens by steroid hormones in cervical tumor cells. Cancer Res 1997, 57:937-942. 56. Vertuani S, Triulzi C, Roos AK, Charo J, Norell H, Lemonnier F, Pisa P, Seliger B, Kiessling R: HER-2/neu mediated downregulation of MHC class I antigens processing prevents CTLmediated tumor recognition upon DNA vaccination in Current Opinion in Immunology 2018, 51:123–132
132 Tumour immunology
HLA-A2 transgenic mice. Cancer Immunol Immunother 2009, 58:653-654. 57. Sa´enz-Lo´pez P, Gouttefangeas C, Hennenlotter J, Concha A, Maleno I, Ruiz-Cabello F, Co´zar JM, Tallada M, Stenzl A, Rammensee HG, Garrido F, Cabrera T: Higher HLA class I expression in renal cell carcinoma than in autologous normal tissue. Tissue Antigens 2010, 75:110-118. 58. Galon J, Pages F, Marincola FM, Angell HK, Thurin M, Lugli A et al.: Cancer classification using the Immunoscore: a worldwide task force. J Transl Med 2012, 10:205. 59. Hams E, Aviello G, Fallon PG: The schistosoma granuloma: friend or foe? Front Immunol 2013, 4:89 http://dx.doi.org/ 10.3389/fimmu.2013.00089 eCollection 2013. 60. Ramakrishnan L: Revisiting the role of granuloma in tuberculosis. Nat Rev Immunol 2012, 12:352-366. 61. Ryschich E, Notzel T, Hinz U, Autschbach F, Ferguson J, Simon I, Weitz J, Frohlich B, Klar E, Buchler MW, Schmidt J: Control of Tcell-mediated immune response by HLA class I in human pancreatic carcinoma. Clin Cancer Res 2005, 11(Pt 1):498-504. 62. Kikuchi E, Yamazaki K, Torigoe T, Cho Y, Miyamoto M, Otzumi S, Hommura F, Dosaka-Akita H, Nishimura M: HLA class I antigen expression is associated with a favourable prognosis in early stage non-small cell lung cancer. Cancer Sci 2007, 98:1424-1430. 63. Rosenberg SA, Lotze MT, Muul LM: A progress report on the treatment of 157 patients with advance cancer using lymphokine-activated killer cells and interleukin-2 or highdose interleukin-2 alone. N Engl J Med 1987, 316:889-897. 64. Andersen R, Donia M, Ellebaek E, Borch TH, Kongsted P, Iversen TZ, Holmich LR, Hendel HW, Met O, Hald AM et al.: Longlasting complete responses in patients with metastatic melanoma after adoptive cell therapy with tumor infiltrating lymphocytes and an attenuated IL-2 regimen. Clin Cancer Res 2016, 22:3734-3745 http://dx.doi.org/10.1158/1078-0432.CCR15-1879. 65. Honeychurch J, Cheadle EJ, Dovedi SJ, Illidge TM: Immunoregulatory antibodies for the treatment of cancer. Expert Opin Biol Ther 2015, 15:787-801.
Current Opinion in Immunology 2018, 51:123–132
66. Wang E, Worschech A, Marincola FM: The immunological constant of rejection. Trends Immunol 2008, 29:256-262. 67. Garrido C, Romero I, Berruguilla E, Cancela B, Algarra I, Collado A, Garcia-Lora AM, Garrido F: Immunotherapy eradicates metastases with reversible defects in MHC class I expression. Cancer Immunol Immunother 2011, 60:1257-1268. 68. Garrido G, Rabasa A, Garrido C, Chao L, Garrido F, GarciaLora AM, Sanchez-Ramirez B: Upregulation of HLA class I expression on tumor cells by the anti-EGFR antibody nimotuzumab. Front Pharmacol 2017 http://dx.doi.org/10.3389/ fphar.2017.00595. 69. del Campo A, Aptsiauri N, Mendez R, Zinchenko S, Vales A, Paschen A, Ward S, Ruiz-Cabello F, Gonzalez-Aseguinolaza G, Garrido F: Efficient recovery of HLA class I expression in human tumor cells after b2-microglobulin gene transfer using adenoviral vector: implications for cancer immunotherapy. Scand J Immunol 2009, 70:125-135. 70. del Campo Ana B, Carretero J, Aptsiauri N, Garrido F: Targeting tumor HLA class I expression to increase tumor immunogenicity. Tissue Antigens 2012, 79:147-154. 71. del Campo AB, Carretero J, Munoz JA, Zinchenko S, RuizCabello F, Gonzalez-Aseguinolaza G, Garrido F, Aptsiauri N: Adenovirus expressing b2-microglobulin recovers HLA class I expression and antitumor immunity by increasing T-cell recognition. Cancer Gene Ther 2014, 21:317-332. 72. Morales A, Eidinger D, Bruce AW: Intracavitary Bacillus Calmette Guerin in the treatment of superficial bladder tumors. J Urol 1976, 116:180-183. 73. Garrido F, Aptsiauri N, Doorduijn EM, Garcia Lora A, van hall T: The urgent need to recover MHC class I in cancers for effective immunotherapy. Curr Opin Immunol 2016, 39:44-51. 74. Thor Straten P, Garrido F: Targetless T cells in cancer immunotherapy. J Immunother Cancer 2016, 3:23. 75. Cabrera T, Lopez-Nevot MA, Gaforio JJ, Ruiz-Cabello F, Garrido F: Analysis of HLA expression in human tumor tissues. Cancer Immunol Immunother 2003, 52:1-9. 76. Gorer P: The genetic and antigenic basis of tumor transplantation. J Pathol Bacteriol 1937, 44:691-697 http://dx. doi.org/10.1002/path 1700440313.
www.sciencedirect.com