Published OnlineFirst May 14, 2020; DOI: 10.1158/1078-0432.CCR-19-2095

CLINICAL CANCER RESEARCH | REVIEW

The NKG2A–HLA-E Axis as a Novel Checkpoint in the Tumor Microenvironment Linda Borst, Sjoerd H. van der Burg, and Thorbald van Hall

ABSTRACT ◥ The success of checkpoint blockade therapy revolutionized binds a limited peptide repertoire and its expression is commonly cancer treatment. However, we need to increase the fraction of detected in human cancer. NKG2A blockade as a standalone responding patients and overcome acquired resistance to these therapy appears poorly effective in mouse tumor models, how- therapies. Recently, the inhibitory receptor NKG2A received ever, in the presence of activated T cells, for example, induced by attention as a new kid on the block of immune checkpoints. PD-1/PD- blockade or cancer vaccines, exerts strongly This receptor is selectively expressed on cytotoxic lymphocytes, enhanced efficacy. Clinical trials demonstrated safety of the þ including natural killer cells and CD8 T cells, and NKG2A T humanized NKG2A-blocking antibody, monalizumab, and first cells are preferentially residing in tissues, like the tumor micro- results of phase II trials demonstrate encouraging durable environment. Its ligand, histocompatibility leucocyte antigen E response rates. Further development of this axis is clearly (HLA-E), is a conserved nonclassical HLA class I molecule that warranted.

Introduction The Similarities and Differences within The unprecedented clinical impact of immune checkpoint block- the NKG2A Family ade therapy induced a new era of cancer treatment (1). After Activation of T cells is the result of T-cell receptor (TCR) ligation introduction in clinical practice of antibodies to CTLA-4 and the with antigen–HLA complexes in combination with costimulatory PD-1/PD-L1 axis, additional checkpoints like TIM-3, TIGIT, and signals, like CD28 and CD27, and cytokines. To avoid unrestricted VISTA and stimulation of activating receptors like CD27, CD40, immune responses that might result in pathology toward host and 4-1BB are evaluated for their antitumor-inducing immunity. tissues, T cells are also equipped with multiple inhibitory receptors, Recently, the immune inhibitory receptor NKG2A draw the atten- which are expressed in a programmed way (11). As revealed, CTLA- – tion as the new kid on the block of checkpoints (2 5). NKG2A 4 and PD-1 inhibitory receptors are also considered as markers of forms heterodimers with the CD94 chain (6) and recognizes the recent T-cell activation. The spaciotemporal dynamics are different nonclassical HLA class I molecule HLA-E. These recent studies for each inhibitory receptor, resulting in distinct profiles of immune demonstrated high expression of NKG2A on natural killer (NK) pathology revealed in knockout mice. The CTLA-4–knockout cells and cytotoxic CD8 T cells in the tumor microenvironment as a mouse displays spontaneous and overt lymphoproliferation, where- result of PD-1 blockade therapy as well as after immune activation as PD-1 and NKG2A knockouts show much milder phenotypes by cancer vaccines (7, 8). First clinical trials in gynecologic cancers (12–15). The cytoplasmic tail of the NKG2A receptor contains two and head and neck carcinoma with the NKG2A-blocking antibody immunoreceptor tyrosine-based inhibition motifs (ITIM) capable monalizumab shows safety and promising clinical responses in of recruiting both SHP-1 (16) and SHP-2 phosphatases, but not the refractory disease, including regressions of target lesions (8, 9). inositol phosphatase SHIP (Fig. 1;refs.17–19). Both ITIMs Interestingly, levels of histocompatibility leucocyte antigen are required to mediate the maximal inhibitory signal, but the E (HLA-E), which is the sole ligand of the CD94/NKG2A recep- membrane-distal ITIM is of primary importance rather than the tor (10), are frequently upregulated in many cancers, suggesting membrane-proximal ITIM (20). The partner CD94 has only seven that this axis functions as an acquired resistance mechanism after cytoplasmic amino acids, thus lacks ITIMs and has no role in immune activation in the tumor microenvironment. Here, we downstream signaling. Interestingly, CD94 can also form a hetero- provide an overarching view on the current understanding of dimer with a very close family member of NKG2A (encoded by the NKG2A and HLA-E in cancer and touch upon gaps in our Klrc1 ), which is named NKG2C (encoded by the Klrc2 gene). knowledge. NKG2C is an activating receptor transducing a stimulating signal via association with DAP12, bearing an immunoreceptor tyrosine- based activation motif (21). The NKG2C protein carries some Department of Medical Oncology, Oncode Institute, Leiden University Medical amino acid differences compared with NKG2A, resulting in a 6-fold Center, Leiden, the Netherlands. lower affinity for the shared ligand HLA-E (22, 23). In NK cells, the Corresponding Authors: Thorbald van Hall, Leiden University Medical Center, two family members are usually not expressed together in the same Albinusdreef 2, Leiden 2333 ZA, the Netherlands. Phone: 3107-1526-6945; Fax: cell and a switch to NKG2C marks more mature NK cells, also 31 71 5264036; E-mail: [email protected]; and Linda Borst, E-mail: “ ” [email protected] referred to as adaptive NK cells (24, 25). Another activating family member, NKG2D (encoded by the Klrk1 gene) is more distantly Clin Cancer Res 2020;26:1–8 related and does not partner with CD94, but rather forms homo- doi: 10.1158/1078-0432.CCR-19-2095 dimers, and is engaged by stress-induced self-, for example, 2020 American Association for Cancer Research. MICA and ULBP members (26).

AACRJournals.org | OF1

Downloaded from clincancerres.aacrjournals.org on September 26, 2021. © 2020 American Association for Cancer Research. Published OnlineFirst May 14, 2020; DOI: 10.1158/1078-0432.CCR-19-2095

Borst et al.

Expression and Function of NKG2A in Box 1: The basics of HLA-E Cytotoxic Lymphocytes In contrast to classical MHC class I molecules, HLA-E is Approximately half of peripheral NK cells display the NKG2A high virtually nonpolymorphic with only two functional alleles present receptor and these cells are mostly present in the CD56 fraction, in the human population: the HLA-E01:01 and the HLA-E01:03 which contains the more immature cells. Intratumoral NK cells have variants. These two alleles only differ in a single amino acid at somewhat higher frequencies of NKG2A (7). Interestingly, NKG2A position 107, being arginine (01:01) or glycine (01:03). Position 107 expression on CD8 T cells seems to be highly regulated, as peripheral is located outside the peptide-binding groove of the heavy cells hardly express the receptor, but a substantial fraction of intra- chain (53). The crystal structure of the peptide-binding groove, tumoral T cells, especially those in immune reactive milieu, display single alanine substitutions, and peptide elution, have demonstrat- NKG2A (7, 27). These CD8 T cells often display a late effector memory ed that HLA-E has an optimal structure to bind peptides with two phenotype and lack expression of typical central memory markers primary anchor residues at positions 2 and 9, and secondary anchor CCR7, CD27, and CD28 or late effector marker KLRG1. Interestingly, residues at position 7 and possibly 3 (54–59). Surprisingly, a very we and others found NKG2A expression on CD8 T cells harboring a limited repertoire of peptides was found under homeostatic con- tissue-resident signature, marked by specific integrins like CD103 – ditions and the most prominent are the signal peptides of classical (7, 28 30). Although induction of NKG2A is initiated by TCR MHC class I molecules. The majority of HLA class I alleles share a b triggering, the presence of tissue cytokines like IL15 and TGF might consensus sequence in their signal peptides with the amino acid – b enhance its expression (31 33). TGF is often overtly present in the motif of VMAPRTLLL (Table 1). Positions 7 and 8 vary between tumor microenvironment (34). However, the functional relationship leucine and valine, without affecting binding affinity. Some HLA b between the tissue residence program, NKG2A, and TGF is unclear at alleles, however, contain a substitution at residues p2, 3, or 6, which the moment. impair binding (23). In addition, the immunotolerance molecule Aside from its expression regulation, the NKG2A downstream HLA-G, which is expressed in immune-privileged sites and fre- effects are also not completely unraveled. The inhibitory signals quently in cancers, encodes a unique leader peptide that binds with induced by NKG2A receptor engagement results in decreased capacity fi – high af nity to HLA-E (60, 61). Interestingly, the leader peptide of NK cells and CD8 T cells to lyse target cells (33, 35 39). NKG2A motif is extremely conserved among mammalian species, implying triggering inhibits cytotoxic effector functions on NK cells by disrupt- an important role in immune defense (62) and is even copied by the ing the actin network at the immunologic synapse of activating human cytomegalovirus gpUL40 protein to sustain HLA-E surface receptor NKG2D (40). For CD8 T cells such mode of action is still display on infected cells while shutting down classical HLA class I to be elucidated, although NKG2D or the immunologic synapse of the – fi antigen presentation of host cells (63 65). Importantly, the CD94/ TCR could be lucid candidates (41). In this context, the recent ndings NKG2A receptor exhibits clear preference for peptide-containing on the operational mechanism of PD-1, which was suggested to HLA-E, especially for this consensus leader peptide (56). Under dephosphorylate T-cell costimulatory receptor CD28, illustrate our stress, HLA-E can present a peptide of multidrug resistance– limited understanding at molecular level of these important check- associated protein 7 (ALALVRMLI; ref. 66) and under high cell points (42, 43). CTLA-4 executes its inhibitory effects by competing densities the leader peptide QMRPVSRVL of hSP60 stabilizes with CD28 for the same ligands on antigen presenting cells, thereby HLA-E (67). In cancers, classical HLA class I molecules are limiting T-cell costimulation (44). Such molecular details are lacking frequently lost to prevent T-cell–mediated recognition (68, 69), for NKG2A, let alone our insight on direct downstream target but surprisingly, expression of the HLA-E molecule is often affected by NKG2A triggering. enhanced (70–78). Finally, HLA-E surface display is determined by expression levels of classical alleles through the availability HLA-E as a Molecule of Immune of their leader peptide and the binding affinity of this peptide to Tolerance HLA-E, as peptides with a threonine at position 2 poorly bind (see also Table 1; refs. 79, 80). The ligand of the heterodimeric receptor CD94/NKG2A is the human HLA-E and its mouse ortholog Qa-1b. These nonclassical MHC class Ib molecules are surprisingly conserved in the popu- lation and present signal peptides of classical MHC class I mole- cules (Box 1; ref. 45). Interestingly, whereas most tissues express low basal cell surface levels, HLA-E is highly expressed in immune- HLA-E Expression in Cancer privileged sites of the body, including trophoblast cells of the Whereas classical HLA alleles are frequently lost in human cancer to placenta and ductal epithelial cells in the testis and epididymis, prevent T-cell recognition (68, 69), we and others reported high levels suggesting that HLA-E has a role to counter potential attack by CD8 of HLA-E in several cancer types, including gynecologic cancers (up to T cells and NK cells against the partly HLA-mismatched fetus and 90% of tumor samples; refs. 70–72) and up to 50% in breast cancer, haploid reproductive cells (Fig. 2;ref.46).Interestingly,CD94/ non–small cell lung carcinoma (NSCLC), liver, pancreas, kidney, NKG2Aisratherpeptide-specific and preferentially interacts with melanoma, prostate, head and neck, stomach, rectal, and colorectal HLA-E when canonical peptides are bound, but not when these are cancer (73–78). Figure 2 displays some examples of tissue sections of absent (22, 47). These canonical peptides are derived from the human cancers and their healthy counterparts. The surface expression leaders of HLA class I molecules, including the immune-tolerant of HLA-E is correlated with functional antigen-processing compo- molecule HLA-G (48). In addition, the expression of HLA-E or its nents and infiltration of CD8 T cells, however, expression can also be mouse ortholog Qa-1b provides protection of tissue integrity during observed in tumors with downregulated classical HLA class I expres- viral clearance and limits excessive activation and apoptosis of CD8 sion. In a cohort of patients diagnosed with high-grade serous ovarian T cells (15, 49–52). carcinoma, HLA-E expression correlated with a significantly worse

OF2 Clin Cancer Res; 26(21) November 1, 2020 CLINICAL CANCER RESEARCH

Downloaded from clincancerres.aacrjournals.org on September 26, 2021. © 2020 American Association for Cancer Research. Published OnlineFirst May 14, 2020; DOI: 10.1158/1078-0432.CCR-19-2095

NKG2A as a Novel Checkpoint in the Tumor Microenvironment

Table 1. Sequences and origins of HLA-E–binding peptides.

HLA-derived leader peptides HLA locus Allele

Substitution at residues P7 and P8 MAVMAPRTLLLLLSGALALTQTWAa HLA-A A01, A03, A11, A29, A30, A31, A32, A33, A36, A74 HLA-C C02, C15 VMAPRTLVL HLA-A A02, A23, A24, A25, A26, A34, A43, A66, A68, A69 VMAPRTVLL HLA-B B07, B08, B14, B38, B39, B42, B48, B67, B73, B81 VMAPRTLIL HLA-C C01, C03, C04, C05, C06, C08, C12, C14, C16 VMAPRTVFL HLA-G G01 Substitution at residues P2, P3, and P6 VMPPRTLLL HLA-A A80 VTAPRTVLL HLA-B B15, B35, B40, B41, B45, B46, B49, B50, B52, B53, B57, B58 VTAPRTLLL HLA-B B13, B18, B27, B37, B44, B47, B54, B55, B56, B59, B82, B83 VMAPRALLL HLA-C C07, C18 VMAPQALLL HLA-C C17 MVDGTLLLb HLA-E E01 MAPRSLLLb HLA-F F01

Note: IPD-IMGT/HLA Database, http://www.ebi.ac.uk/ipd/imgt/hla/. aConsensus HLA class I-derived signal sequence. bLeader peptides from HLA-E and HLA-F are shorter and are therefore unable to serve as HLA-E binders.

survival (81). A poor relapse-free survival rate was also observed in cervical, and ovarian carcinomas. However, this predictive value is breast cancer, but only when classical class I expression was lost (73). abrogated when tumor display high HLA-E levels, suggesting that CD8 T-cell infiltration and retained expression of classical HLA class I HLA-E mediates resistance against CD8 T-cell attack (70, 71, 74). In strongly associate with a better prognosis for patients with NSCLC, primary colorectal cancers, patients with high HLA-E expression

Tumor cell (Polymorphic) (Monomorphic) Classical Nonclassical HLA class I HLA class I molecule molecule HLA-E

HLA class I epitope Signal peptide β α

TCR CD8 NKG2A CD94 CD94 NKG2C (KLRC1) (KLRD1) (KLRD1) (KLRC2) CD3

ITIM DAP12 ITAM ITAM P SHP1 P P ITIM P P P

CD8 T lymphocyte Cytotoxic CD8 T and NK lymphocyte

Figure 1. NKG2 family and their ligands. The polymorphic classical HLA class I molecules present antigenic peptide epitopes to the TCR in complex with CD3 and coreceptor CD8 at the surface of CD8 T cells. In contrast, HLA-E is a monomorphic nonclassical HLA class I molecule that presents a limited set of conserved signal peptides. These peptides are derived from leader sequences of classical HLA class I molecules. NKG2A and NKG2C form heterodimer receptors with CD94 and both target the same p/ HLA-E complex, but ligation induces an inhibitory signal for NKG2A and an activation signal for NKG2C. The NKG2 family of mouse and man also comprises the activating family member NKG2E (not shown here), but its function remains elusive. The more distantly related activating homodimer NKG2D receptor binds MICA/B and ULBP1-6, “empty” molecules that fold like HLA class I heavy chains but do not contain b2-microglobulin (not shown here). The NKG2 family is expressed by both cytotoxic CD8 T cells and NK lymphocytes. Adapted from an image created with BioRender.com.

AACRJournals.org Clin Cancer Res; 26(21) November 1, 2020 OF3

Downloaded from clincancerres.aacrjournals.org on September 26, 2021. © 2020 American Association for Cancer Research. Published OnlineFirst May 14, 2020; DOI: 10.1158/1078-0432.CCR-19-2095

Borst et al.

Healthy Cancer also signify the negative effect of HLA-E in these cohorts as observed in others. Finally, it should be noted that HLA-E–directed antibodies fail to distinguish between peptide- filled molecules and empty conformers (see Box 2), complicating the Colon interpretation of these tissue slide results, as especially HLA-E filled with leader peptides constitute ligands for the CD94/NKG2A receptor (56).

Kidney Box 2: Antibodies against HLA-E Generation of specific antibodies to HLA-E was challenging due Liver to its close homology with classical HLA class I molecules. Three antibodies stand out as most specific: (i) clone 3D12 is frequently used in flow cytometry and was generated against soluble HLA-E with the HLA-A2 leader peptide (83) and is capable to stain the Lung native conformation of the molecule and also binds to b2m-free HLA-E conformations (84). (ii) Clone MEM-E/02 recognizes a linear epitope on HLA-E and is mostly used in formalin-fixed, paraffin-embedded tissue slide staining (84, 85). (iii) Clone TFL- Cervix 033 binds HLA-E in a highly specific manner and is reactive to the a1 and a2 helices of HLA-E, which are the same sequences recognized by the CD94/NKG2A inhibitory receptors (86). A Healthy debate on the exact specificity and potential cross-reactivity with classical HLA molecules of these antibodies is ongoing (83–85, 87, 88), and stirred by recent findings that HLA-E and Epididymis its mouse ortholog Qa-1b poorly associate with b2m and accu- mulates as an b2m-free and peptide-empty form at the cell surface (84, 89–91). As currently available antibodies fail to distinguish between peptide-loaded or open HLA-E conformers and the biological relevance of open conformations remains to be deter- Placenta mined, novel antibodies to peptide-containing forms of HLA-E and preferably even peptide-specific antibodies are urgently required to promote the development of this field. Figure 2. HLA-E expression in healthy and cancer tissue. IHC staining of healthy and cancer tissues for HLA-E, as deposited in the Human Protein Atlas (www. proteinatlas.org). Tissue slides were stained either with MEM-E/02 antibody Induction of HLA-E or rabbit polyclonal HPA031454 (85, 86). Of note, HLA-E antibodies also binds free heavy chains. The fact that HLA-E is generally upregulated in cancers compared with healthy tissue touches upon responsible gene transcription mechanisms. IFNg is proposed as an important cytokine and is locally showed a significantly decreased disease-free survival for Dukes C produced by tumor-infiltrating T cells and NK cells. At the transcrip- patients (76). In another retrospective cohort of 234 colorectal patients, tion level, this is substantiated through binding of a STAT1-containing the expression of HLA-E, b2m, CD94, CD8, and NKp46 was examined complex to distinct IFNg-responsive region (IRR) present upstream of by IHC on tissue microarray. HLA-E/b2m was overexpressed in the HLA-E gene (92). In addition, the upstream UIRR region mediates microsatellite instable tumors and to a lesser extend in microsatellite a 3- to 8-fold increase in HLA-E transcription in response to IFNg stable ones (45% vs. 19%, respectively) and corresponded with worse upon binding of GATA-1 (93). In vitro experiments indeed showed survival (78). Unfortunately, NKG2A expression cannot be quantified that IFNg significantly increases the surface expression of HLA-E and in paraffin-embedded tissues because of lack of specific antibody. In an the shedding of soluble HLA-E by melanoma cells, in a metallopro- independent study of colon cancers, the researchers concluded that the teinase-dependent fashion (94). This also corresponds to studies total absence of HLA class I, including HLA-E and HLA-G, related to a showing enhanced HLA-E expression on tumors upon increased better overall and disease-free survival (77). This was attributed to NK- cytotoxic lymphocyte infiltration (71) and enhanced Qa-1b expression cell–mediated killing in the portal vein of MHC class I–negative cancer in mice in response to immune therapy leading to increased T-cell cells, which give rise to liver metastases. infiltration (7). Nevertheless, there are also tumors and tumor cell lines It should be stated that there are also several studies of patient that constitutively upregulate HLA-E molecules, soluble and at the cell cohorts in which the researchers did not observe a relevant corre- surface, irrespective of IFNg treatment (94). Importantly, HLA-E lation with HLA-E expression (75, 82). However, as previously surface expression is posttranslationally regulated by availability of demonstrated, the role of HLA-E expression seems to be dependent the conserved leader peptides, the peptide transporter TAP, and on immune contexture, like lymphocyte infiltration and presence of proteolytic enzymes. Moreover, HLA-E can present alternative classical HLA class I molecules. Stratification on these factors might peptides aside from the HLA class I leaders and can be recognized

OF4 Clin Cancer Res; 26(21) November 1, 2020 CLINICAL CANCER RESEARCH

Downloaded from clincancerres.aacrjournals.org on September 26, 2021. © 2020 American Association for Cancer Research. Published OnlineFirst May 14, 2020; DOI: 10.1158/1078-0432.CCR-19-2095

NKG2A as a Novel Checkpoint in the Tumor Microenvironment

by ab TCRs (95, 96). Clearly, novel research tools are required to of other forms of immunotherapy, like PD-L1 blockade or cancer distinguish HLA-E complexed with the monomorphic leader peptides vaccines (7, 8, 101). Cancer vaccines induced strong tumor-directed from those complexed with alternative peptides, which might actually T-cell responses and infiltration of CD8 T cells in the tumors, be present in tumors (58). leading to local IFNg release and increase of the mouse HLA-E þ ortholog (7). In addition, frequencies of NKG2A CD8 T cells were increased in tumor-infiltrating lymphocyte (TIL). The association – þ Targeting the NKG2A HLA-E Axis of immune reactivity and NKG2A T-cell frequency was corrob- Similarities between the PD1–PD-L1 axis and the NKG2A– orated in human TIL of oropharyngeal carcinomas (7). Together, HLA-E axis are obvious, as they both involve lymphocytes and these observations designated a role for NKG2A as an acquired represent inhibitory immune receptors, and their ligands are resistance mechanism (102). This concept is further substantiated expressed on cancer cells and inducible by the proinflammatory by preclinical in vitro data demonstrating a requirement of immune cytokine IFNg. Of note, HLA-E transcripts in The Cancer activation signals to reveal beneficial effects of NKG2A block- Genome Atlas (TCGA) database exceed those of PD-L1, suggest- ade (8, 27, 38). Design of clinical trials should therefore be based ing a high and general overexpression (Fig. 3). These two pairs on combination therapy leading to inflammatory responses in reflect feedback signals to dampen overt T-cell–mediated tissue patients with cancer. Induced expression of HLA-E or enhanced þ damage at affected lesion sites, which might result in resistance and frequencies of NKG2A immune cells in the tumor might serve as immune escape of cancer (97, 98). In contrast to PD-1, NKG2A is predictive biomarkers. selectively expressed on lymphocytes with cytolytic function, including NK cells, NKT cells, and a subset of CD8 T cells. Thereby, the NKG2A–HLA-E axis is suggested to predominantly act at the NKG2A in Clinical Trials terminal tumor–attack stage and not to be involved in priming or Monalizumab (IPH2201, Innate Pharma/AstraZeneca) is a human- regulation of immune responses. Interruption of this axis by ized IgG4 antibody blocking the interaction of human NKG2A with blocking antibodies can be envisaged at both sides. However, HLA-E and showed a therapeutic effect in immunodeficient mice NKG2A is the preferred target instead of HLA-E, because blockade harboring human leukemia (103). The NKG2A-blocking antibody of HLA-E would also prevent interaction with the activating monalizumab was furthermore administered in a dose ranging phase II NKG2C receptor. A NKG2C-expressing subset of NK cells, also clinical trial in patients with gynecologic malignancies (9). Adminis- called “adaptive NK cells,” displays an altered receptor profile and is tration of 10 mg/kg i.v. every 2 weeks was well-tolerated and even associated with chronic viruses, like cytomegalovirus (CMV; short-term disease stabilizations were observed (9). In another recent refs. 99, 100). Its role in immunity to CMV remains elusive, but clinical trial, monalizumab was combined with the EGFR-blocking anti-HLA-E antibodies would also block activation of such NK cells antibody cetuximab in previously treated squamous cell carcinoma of through NKG2C. the head and neck. This combination showed a promising 31% Disappointingly, mouse cancer models demonstrated limited objective response rate (8). Cetuximab is able to activate NK cells via success of NKG2A blockade therapy when provided as a standalone Fcg receptors, leading to antibody-mediated cellular cytotoxicity. blocking antibody, however, it greatly improved antitumor efficacy Addition of monalizumab enhanced NK-cell effector functions (8). Currently, several trials in which monalizumab is tested are enrolling patients. Different combinations with anti-EGFR, anti-PD-L1, tyrosine kinase inhibitors, and chemotherapy are tested in several 2,500 cancer indications, including patients with resectable NSCLC HLA-E (NCT03794544), patients with PD-1 therapy-resistant NSCLC 2,000 PD-L1 (NCT03833440), and patients with advanced nonresectable stage III NSCLC (NCT03822351), as well as patients with advanced squamous cell carcinoma of the head and neck (NCT02643550), refractory 1,500 chronic lymphocytic leukemia (NCT02557516), or other hematologic malignancies after stem cell transplantation (NCT02921685), and also FPKM 1,000 a basket trial with advanced solid malignancies (NCT02671435). This latter trial reported a manageable safety profile and durable partial 500 responses in patients with microsatellite stable (MSS) colorectal carcinoma treated with combinations of FOLFOX chemotherapy and blocking antibodies to VEGF, PD-L1, and NKG2A (104). On the basis 0 of these promising results, a larger phase II trial will start in the near Liver Lung Renal Breast future for patients with high-risk MSS colorectal carcinoma who Cervical Ovarian Colorectal received radical surgery, testing different adjuvant therapies based on Head and neck standard-of-care chemotherapy FOLFOX in combination with dur- valumab (anti-PD-L1) and monalizumab (anti-NKG2A; Columbia 2; NCT04145193). Figure 3. HLA-E and PD-L1 transcription levels in various cancer types. RNA-sequencing data for HLA-E and PD-L1 is reported as median FPKM (number fragments per Conclusions kilobase of exon per million reads), generated by TCGA. Normal distribution across the dataset is visualized with box plots, shown as median and 25th and The current challenge of checkpoint blockade therapy is to 75th percentiles. Points are outliers when they are above the 97.5 or below the increase the fraction of responding patients and, second, to over- 2.5 percentile. come acquired resistance. NKG2Ablockadedisplaysunique

AACRJournals.org Clin Cancer Res; 26(21) November 1, 2020 OF5

Downloaded from clincancerres.aacrjournals.org on September 26, 2021. © 2020 American Association for Cancer Research. Published OnlineFirst May 14, 2020; DOI: 10.1158/1078-0432.CCR-19-2095

Borst et al.

characteristics compared with other immune checkpoints, as this Disclosure of Potential Conflicts of Interest target is selectively expressed within tumor lesions on cytotoxic S.H. van der Burg reports receiving other commercial research support from lymphocytes and is not involved in priming or regulation of Innate Pharma. T. van Hall reports receiving commercial research grants from þ fl immunity against cancer. NKG2A T cells are mostly residing in Innate Pharma. No potential con icts of interest were disclosed by the other author. the tumor microenvironment where they are effector cells attacking the transformed cells. Moreover, we and others showed increased Acknowledgments frequencies of these cytotoxic lymphocytes upon administration of This study was financially supported by the Dutch Cancer Society (2014-7146 therapeutic cancer vaccines. These observations plea for combina- to T. van Hall and S.H. van der Burg) and by a research grant from Innate Pharma tion therapies of the NKG2A-blocking antibody monalizumab with (to T. van Hall and S.H. van der Burg). such vaccines. In the meanwhile, fundamental investigation on NKG2A and its ligand HLA-E should provide mode-of-action of Received January 27, 2020; revised March 31, 2020; accepted May 12, 2020; this novel immune checkpoint. published first May 14, 2020.

References 1. Tumeh PC, Harview CL, Yearley JH, Shintaku IP, Taylor EJ, Robert L, et al. PD- 18. Le Drean E, Vely F, Olcese L, Cambiaggi A, Guia S, Krystal G, et al. Inhibition of 1 blockade induces responses by inhibiting adaptive immune resistance. Nature antigen-induced T cell response and antibody-induced NK cell cytotoxicity by 2014;515:568–71. NKG2A: association of NKG2A with SHP-1 and SHP-2 protein-tyrosine 2. Creelan BC, Antonia SJ. The NKG2A immune checkpoint - a new direction in phosphatases. Eur J Immunol 1998;28:264–76. cancer immunotherapy. Nat Rev Clin Oncol 2019;16:277–8. 19. Viant C, Fenis A, Chicanne G, Payrastre B, Ugolini S, Vivier E. SHP-1-mediated 3. Zaghi E, Calvi M, Marcenaro E, Mavilio D, Di Vito C. Targeting NKG2A to inhibitory signals promote responsiveness and anti-tumour functions of nat- elucidate ontogenesis and to develop novel immune- ural killer cells. Nat Commun 2014;5:5108. therapeutic strategies in cancer therapy. J Leukoc Biol 2019;105:1243–51. 20. Kabat J, Borrego F, Brooks A, Coligan JE. Role that each NKG2A immunor- 4. Mingari MC, Pietra G, Moretta L. Immune checkpoint inhibitors: Anti- eceptor tyrosine-based inhibitory motif plays in mediating the human CD94/ NKG2A antibodies on board. Trends Immunol 2019;40:83–5. NKG2A inhibitory signal. J Immunol 2002;169:1948–58. 5. Haanen JB, Cerundolo V. NKG2A, a new kid on the immune checkpoint block. 21. Lanier LL, Corliss B, Wu J, Phillips JH. Association of DAP12 with activating Cell 2018;175:1720–2. CD94/NKG2C NK cell receptors. Immunity 1998;8:693–701. 6. Carretero M, Cantoni C, Bellon T, Bottino C, Biassoni R, Rodríguez A, et al. The 22. Kaiser BK, Barahmand-pour F, Paulsene W, Medley S, Geraghty DE, Strong CD94 and NKG2-A C-type covalently assemble to form a natural RK. Interactions between NKG2x immunoreceptors and HLA-E ligands killer cell inhibitory receptor for HLA class I molecules. Eur J Immunol 1997;27: display overlapping affinities and thermodynamics. J Immunol 2005;174: 563–7. 2878–84. 7. van Montfoort N, Borst L, Korrer MJ, Sluijter M, Marijt KA, Santegoets SJ, et al. 23. Vales-Gomez M, Reyburn HT, Erskine RA, Lopez-Botet M, Strominger JL. NKG2A blockade potentiates CD8 T cell immunity induced by cancer vaccines. Kinetics and peptide dependency of the binding of the inhibitory NK receptor Cell 2018;175:1744–55. CD94/NKG2-A and the activating receptor CD94/NKG2-C to HLA-E. EMBO J 8. Andre P, Denis C, Soulas C, Bourbon-Caillet C, Lopez J, Arnoux T, et al. Anti- 1999;18:4250–60. NKG2A mAb is a checkpoint inhibitor that promotes anti-tumor immunity by 24. Rolle A, Meyer M, Calderazzo S, Jager D, Momburg F. Distinct HLA-E peptide unleashing both T and NK cells. Cell 2018;175:1731–43. complexes modify antibody-driven effector functions of adaptive NK cells. 9. Tinker AV, Hirte HW, Provencher D, Butler M, Ritter H, Tu D, et al. Dose- Cell Rep 2018;24:1967–76. ranging and cohort-expansion study of monalizumab (IPH2201) in patients 25. Tesi B, Schlums H, Cichocki F, Bryceson YT. Epigenetic regulation of adaptive with advanced gynecologic malignancies: a trial of the Canadian Cancer Trials NK cell diversification. Trends Immunol 2016;37:451–61. Group (CCTG): IND221. Clin Cancer Res 2019;25:6052–60. 26. Zingoni A, Molfetta R, Fionda C, Soriani A, Paolini R, Cippitelli M, et al. 10. Lee N, Llano M, Carretero M, Ishitani A, Navarro F, Lopez-Botet M, et al. HLA- NKG2D and its ligands: “one for all, all for one”. Front Immunol 2018;9: E is a major ligand for the natural killer inhibitory receptor CD94/NKG2A. 476. Proc Natl Acad Sci U S A 1998;95:5199–204. 27. Abd Hamid M, Wang RZ, Yao X, Fan P, Li X, Chang XM, et al. Enriched HLA-E 11. Chen L, Flies DB. Molecular mechanisms of T cell co-stimulation and co- and CD94/NKG2A interaction limits antitumor CD8(þ) tumor-infiltrating t inhibition. Nat Rev Immunol 2013;13:227–42. lymphocyte responses. Cancer Immunol Res 2019;7:1293–306. 12. Waterhouse P, Penninger JM, Timms E, Wakeham A, Shahinian A, Lee KP, 28. Piet B, de Bree GJ, Smids-Dierdorp BS, van der Loos CM, Remmerswaal EBM, et al. Lymphoproliferative disorders with early lethality in mice deficient in von der Thusen JH, et al. CD8(þ) T cells with an intraepithelial phenotype Ctla-4. Science 1995;270:985–8. upregulate cytotoxic function upon influenza infection in human lung. J Clin 13. Tivol EA, Borriello F, Schweitzer AN, Lynch WP, Bluestone JA, Sharpe AH. Invest 2011;121:2254–63. Loss of CTLA-4 leads to massive lymphoproliferation and fatal multiorgan 29. Hombrink P, Helbig C, Backer RA, Piet B, Oja AE, Stark R, et al. Programs for tissue destruction, revealing a critical negative regulatory role of CTLA-4. the persistence, vigilance and control of human CD8(þ) lung-resident memory Immunity 1995;3:541–7. T cells. Nat Immunol 2016;17:1467–78. 14. Nishimura H, Nose M, Hiai H, Minato N, Honjo T. Development of lupus-like 30. Savas P, Virassamy B, Ye C, Salim A, Mintoff CP, Caramia F, et al. Single-cell autoimmune diseases by disruption of the PD-1 gene encoding an ITIM motif- profiling of breast cancer T cells reveals a tissue-resident memory subset carrying immunoreceptor. Immunity 1999;11:141–51. associated with improved prognosis. Nat Med 2018;24:986–93. 15. Rapaport AS, Schriewer J, Gilfillan S, Hembrador E, Crump R, Plougastel 31.JabriB,SelbyJM,NegulescuH,LeeL,RobertsAI,BeavisA,etal.TCR BF, et al. The inhibitory receptor NKG2A sustains virus-specificCD8(þ) specificity dictates CD94/NKG2A expression by human CTL. Immunity T cells in response to a lethal poxvirus infection. Immunity 2015;43: 2002;17: 1112–24. 487–99. 16. Carretero M, Palmieri G, Llano M, Tullio V, Santoni A, Geraghty DE, et al. 32. Gunturi A, Berg RE, Crossley E, Murray S, Forman J. The role of TCR Specific engagement of the CD94/NKG2-A killer inhibitory receptor by the stimulation and TGF-beta in controlling the expression of CD94/NKG2A HLA-E class Ib molecule induces SHP-1 phosphatase recruitment to tyrosine- receptors on CD8 T cells. Eur J Immunol 2005;35:766–75. phosphorylated NKG2-A: evidence for receptor function in heterologous 33. Bertone S, Schiavetti F, Bellomo R, Vitale C, Ponte M, Moretta L, et al. transfectants. Eur J Immunol 1998;28:1280–91. Transforming growth factor-b-induced expression of CD94/NKG2A inhibi- 17. Brooks AG, Posch PE, Scorzelli CJ, Borrego F, Coligan JE. NKG2A complexed tory receptors in human T lymphocytes. Eur J Immunol 1999;29:23–9. with CD94 defines a novel inhibitory natural killer cell receptor. J Exp Med 34. Huang JJ, Blobe GC. Dichotomous roles of TGF-b in human cancer. 1997;185:795–800. Biochem Soc Trans 2016;44:1441–54.

OF6 Clin Cancer Res; 26(21) November 1, 2020 CLINICAL CANCER RESEARCH

Downloaded from clincancerres.aacrjournals.org on September 26, 2021. © 2020 American Association for Cancer Research. Published OnlineFirst May 14, 2020; DOI: 10.1158/1078-0432.CCR-19-2095

NKG2A as a Novel Checkpoint in the Tumor Microenvironment

35. Sheu BC, Chiou SH, Lin HH, Chow SN, Huang SC, Ho HN, et al. Up-regulation with primary anchor residues at positions 2 and 9. Eur J Immunol 1997;27: of inhibitory natural killer receptors CD94/NKG2A with suppressed intracel- 1164–9. lular perforin expression of tumor-infiltrating CD8(þ) T lymphocytes in 60. Llano M, Lee N, Navarro F, Garcia P, Albar JP, Geraghty DE, et al. HLA-E- human cervical carcinoma. Cancer Res 2005;65:2921–9. bound peptides influence recognition by inhibitory and triggering CD94/ 36. Kamiya T, Seow SV, Wong D, Robinson M, Campana D. Blocking expression of NKG2 receptors: preferential response to an HLA-G-derived nonamer. inhibitory receptor NKG2A overcomes tumor resistance to NK cells. J Clin Eur J Immunol 1998;28:2854–63. Invest 2019;129:2094–106. 61. Perez-Villar JJ, Melero I, Navarro F, Carretero M, Bellon T, Llano M, et al. The 37. Zhang C, Wang XM, Li SR, Twelkmeyer T, Wang WH, Zhang SY, et al. NKG2A CD94/NKG2-A inhibitory receptor complex is involved in natural killer is a NK cell exhaustion checkpoint for HCV persistence. Nat Commun 2019;10: cell-mediated recognition of cells expressing HLA-G1. J Immunol 1997;158: 1507. 5736–43. 38. Courau T, Bonnereau J, Chicoteau J, Bottois H, Remark R, Assante Miranda L, 62. Kurepa Z, Hasemann CA, Forman J. Qa-1b binds conserved class I leader et al. Cocultures of human colorectal tumor spheroids with immune cells reveal peptides derived from several mammalian species. J Exp Med 1998;188:973–8. the therapeutic potential of MICA/B and NKG2A targeting for cancer treat- 63. Tomasec P, Braud VM, Rickards C, Powell MB, McSharry BP, Gadola S, et al. ment. J Immunother Cancer 2019;7:74. Surface expression of HLA-E, an inhibitor of natural killer cells, enhanced by 39. Pereira BI, Devine OP, Vukmanovic-Stejic M, Chambers ES, Subramanian P, human cytomegalovirus gpUL40. Science 2000;287:1031. Patel N, et al. Senescent cells evade immune clearance via HLA-E-mediated NK 64. Wang EC, McSharry B, Retiere C, Tomasec P, Williams S, Borysiewicz LK, et al. and CD8(þ) T cell inhibition. Nat Commun 2019;10:2387. UL40-mediated NK evasion during productive infection with human cyto- 40. Masilamani M, Nguyen C, Kabat J, Borrego F, Coligan JE. CD94/NKG2A megalovirus. Proc Natl Acad Sci U S A 2002;99:7570–5. inhibits NK cell activation by disrupting the actin network at the immuno- 65. Ulbrecht M, Martinozzi S, Grzeschik M, Hengel H, Ellwart JW, Pla M, et al. logical synapse. J Immunol 2006;177:3590–6. Cutting edge: the human cytomegalovirus UL40 gene product contains a 41. Roy NH, Burkhardt JK. The actin cytoskeleton: a mechanical intermediate ligand for HLA-E and prevents NK cell-mediated lysis. J Immunol 2000;164: for signal integration at the immunological synapse. Front Cell Dev Biol 2018;6: 5019–22. 116. 66. Wooden SL, Kalb SR, Cotter RJ, Soloski MJ. Cutting edge: HLA-E binds a 42. Kamphorst AO, Wieland A, Nasti T, Yang S, Zhang R, Barber DL, et al. Rescue peptide derived from the ATP-binding cassette transporter multidrug resis- of exhausted CD8 T cells by PD-1–targeted therapies is CD28-dependent. tance-associated protein 7 and inhibits NK cell-mediated lysis. J Immunol 2005; Science 2017;355:1423–7. 175:1383–7. 43. Hui E, Cheung J, Zhu J, Su X, Taylor MJ, Wallweber HA, et al. T cell 67. Micha€elsson J, Teixeira De Matos C, Achour A, Lanier LL, K€arre K, Soderstr€ om€ costimulatory receptor CD28 is a primary target for PD-1-mediated inhibition. K. A signal peptide derived from hsp60 binds HLA-E and interferes with CD94/ Science 2017;355:1428–33. NKG2A recognition. J Exp Med 2002;196:1403–14. 44. Lee KM, Chuang E, Griffin M, Khattri R, Hong DK, Zhang W, et al. Molecular 68. Rosenthal R, Cadieux EL, Salgado R, Bakir MA, Moore DA, Hiley CT, et al. basis of T cell inactivation by CTLA-4. Science 1998;282:2263–6. Neoantigen-directed immune escape in lung cancer evolution. Nature 2019; 45. Braud VM, Allan DS, O'Callaghan CA, Soderstrom K, D'Andrea A, Ogg GS, 567:479–85. et al. HLA-E binds to natural killer cell receptors CD94/NKG2A, B and C. 69. McGranahan N, Rosenthal R, Hiley CT, Rowan AJ, Watkins TBK, Wilson GA, Nature 1998;391:795–9. et al. Allele-specific HLA loss and immune escape in lung cancer evolution. Cell 46. King A, Allan DSJ, Bowen M, Powis SJ, Joseph S, Verma S, et al. HLA-E is 2017;171:1259–71. expressed on trophoblast and interacts with CD94 / NKG2 receptors on 70. Gooden M, Lampen M, Jordanova ES, Leffers N, Trimbos JB, van der Burg decidual NK cells. Eur J Immunol 2000;30:1623–31. SH, et al. HLA-E expression by gynecological cancers restrains tumor- 47. Kaiser BK, Pizarro JC, Kerns J, Strong RK. Structural basis for NKG2A/CD94 infiltrating CD8(þ) T lymphocytes. Proc Natl Acad Sci U S A 2011;108: recognition of HLA-E. Proc Natl Acad Sci U S A 2008;105:6696–701. 10656–61. 48. Ferreira LMR, Meissner TB, Tilburgs T, Strominger JL. HLA-G: at the interface 71. van Esch EM, Tummers B, Baartmans V, Osse EM, Ter Haar N, Trietsch MD, of maternal–fetal tolerance. Trends Immunol 2017;38:272–86. et al. Alterations in classical and nonclassical HLA expression in recurrent and 49. Zhou J, Matsuoka M, Cantor H, Homer R, Enelow RI. Cutting edge: engage- progressive HPV-induced usual vulvar intraepithelial neoplasia and implica- ment of NKG2A on CD8þ effector T cells limits immunopathology in tions for immunotherapy. Int J Cancer 2014;135:830–42. influenza pneumonia. J Immunol 2008;180:25–9. 72. Ferns DM, Heeren AM, Samuels S, Bleeker MCG, de Gruijl TD, Kenter GG, 50. Ely KH, Matsuoka M, DeBerge MP, Ruby JA, Liu J, Schneider MJ, et al. Tissue- et al. Classical and non-classical HLA class I aberrations in primary cervical protective effects of NKG2A in immune-mediated clearance of virus infection. squamous- and adenocarcinomas and paired lymph node metastases. PLoS One 2014;9:e108385. J Immunother Cancer 2016;4:78. 51. Moser JM, Gibbs J, Jensen PE, Lukacher AE. CD94-NKG2A receptors regulate 73. de Kruijf EM, Sajet A, van Nes JGH, Natanov R, Putter H, Smit VTHBM, et al. antiviral CD8(þ) T cell responses. Nat Immunol 2002;3:189–95. HLA-E and HLA-G expression in classical HLA class i-negative tumors is of 52. Suvas S, Azkur AK, Rouse BT. Qa-1b and CD94-NKG2a interaction regulate prognostic value for clinical outcome of early breast cancer patients. J Immunol cytolytic activity of herpes simplex virus-specific memory CD8þ T cells in the 2010;185:7452–9. latently infected trigeminal ganglia. J Immunol 2006;176:1703–11. 74. Talebian Yazdi M, van Riet S, van Schadewijk A, Fiocco M, van Hall T, Taube C, 53. Grimsley C, Ober C. Population genetic studies of HLA-E: evidence for et al. The positive prognostic effect of stromal CD8þ tumor-infiltrating T cells selection. Hum Immunol 1997;52:33–40. is restrained by the expression of HLA-E in non-small cell lung carcinoma. 54. O'Callaghan CA, Tormo J, Willcox BE, Braud VM, Jakobsen BK, Stuart DI, et al. Oncotarget 2016;7:3477–88. Structural features impose tight peptide binding specificity in the nonclassical 75. Seliger B, Jasinski-Bergner S, Quandt D, Stoehr C, Bukur J, Wach S, et al. HLA-E MHC molecule HLA-E. Mol Cell 1998;1:531–41. expression and its clinical relevance in human renal cell carcinoma. Oncotarget 55. Zeng L, Sullivan LC, Vivian JP, Walpole NG, Harpur CM, Rossjohn J, et al. A 2016;7:67360–72. structural basis for antigen presentation by the MHC class Ib molecule, Qa-1b. 76. Levy EM, Bianchini M, Von Euw EM, Barrio MM, Bravo AI, Furman D, et al. J Immunol 2012;188:302–10. Human leukocyte antigen-E protein is overexpressed in primary human 56. Kraft JR, Vance RE, Pohl J, Martin AM, Raulet DH, Jensen PE. Analysis of Qa-1 colorectal cancer. Int J Oncol 2008;32:633–41. (b) peptide binding specificity and the capacity of CD94/NKG2A to discrim- 77. Zeestraten EC, Reimers MS, Saadatmand S, Goossens-Beumer IJ, Dekker JW, inate between Qa-1-peptide complexes. J Exp Med 2000;192:613–24. Liefers GJ, et al. Combined analysis of HLA class I, HLA-E and HLA-G predicts 57. Oliveira CC, van Veelen PA, Querido B, de Ru A, Sluijter M, Laban S, et al. The prognosis in colon cancer patients. Br J Cancer 2014;110:459–68. nonpolymorphic MHC Qa-1b mediates CD8þ T cell surveillance of antigen- 78. Eugene J, Jouand N, Ducoin K, Dansette D, Oger R, Deleine C, et al. The processing defects. J Exp Med 2010;207:207–21. inhibitory receptor CD94/NKG2A on CD8(þ) tumor-infiltrating lymphocytes 58. Lampen MH, Hassan C, Sluijter M, Geluk A, Dijkman K, Tjon JM, et al. in colorectal cancer: a promising new druggable immune checkpoint in the Alternative peptide repertoire of HLA-E reveals a binding motif that is context of HLAE/beta2m overexpression. Mod Pathol 2020;33:468–82. strikingly similar to HLA-A2. Mol Immunol 2013;53:126–31. 79. Ramsuran V, Naranbhai V, Horowitz A, Qi Y, Martin MP, Yuki Y, et al. 59. Braud V, Jones EY, McMichael A. The human major histocompatibility Elevated HLA-A expression impairs HIV control through inhibition of complex class Ib molecule HLA-E binds signal sequence-derived peptides NKG2A-expressing cells. Science 2018;359:86–90.

AACRJournals.org Clin Cancer Res; 26(21) November 1, 2020 OF7

Downloaded from clincancerres.aacrjournals.org on September 26, 2021. © 2020 American Association for Cancer Research. Published OnlineFirst May 14, 2020; DOI: 10.1158/1078-0432.CCR-19-2095

Borst et al.

80. Yunis EJ, Romero V, Diaz-Giffero F, Zuniga J, Koka P. Natural killer cell tolerability and conformationally malleable peptide binding. Nat Commun receptor NKG2A/HLA-E interaction dependent differential thymopoiesis of 2018;9:3137. hematopoietic progenitor cells influences the outcome of HIV infection. J Stem 92. Gustafson KS, Ginder GD. Interferon-gamma induction of the human leuko- Cells 2007;2:237–48. cyte antigen-E gene is mediated through binding of a complex containing 81. Andersson E, Poschke I, Villabona L, Carlson JW, Lundqvist A, Kiessling R, STAT1alpha to a distinct interferon-gamma-responsive element. J Biol Chem et al. Non-classical HLA-class I expression in serous ovarian carcinoma: 1996;271:20035–46. correlation with the HLA-genotype, tumor infiltrating immune cells and 93. Barrett DM, Gustafson KS, Wang J, Wang SZ, Ginder GD. A GATA factor prognosis. Oncoimmunology 2016;5:e1052213. mediates cell type-restricted induction of HLA-E gene transcription by gamma 82. Nasman A, Andersson E, Nordfors C, Grun N, Johansson H, Munck-Wikland interferon. Mol Cell Biol 2004;24:6194–204. E, et al. MHC class I expression in HPV positive and negative tonsillar 94. Derre L, Corvaisier M, Charreau B, Moreau A, Godefroy E, Moreau-Aubry A, squamous cell carcinoma in correlation to clinical outcome. Int J Cancer et al. Expression and release of HLA-E by melanoma cells and melanocytes: 2013;132:72–81. potential impact on the response of cytotoxic effector cells. J Immunol 2006; 83. Lee N, Goodlett DR, Ishitani A, Marquardt H, Geraghty DE. HLA-E 177:3100–7. surface expression depends on binding of TAP-dependent peptides 95. Ottenhoff THM, Joosten SA. Mobilizing unconventional T cells. Science 2019; derived from certain HLA class I signal sequences. J Immunol 1998;160: 366:302–3. 4951–60. 96. Hansen SG, Wu HL, Burwitz BJ, Hughes CM, Hammond KB, Ventura AB, et al. 84. Tremante E, Lo Monaco E, Ingegnere T, Sampaoli C, Fraioli R, Giacomini P. Broadly targeted CD8(þ) T cell responses restricted by major histocompat- Monoclonal antibodies to HLA-E bind epitopes carried by unfolded b2 m-free ibility complex E. Science 2016;351:714–20. heavy chains. Eur J Immunol 2015;45:2356–64. 97. Malmberg KJ, Levitsky V, Norell H, de Matos CT, Carlsten M, Schedvins K, 85. Menier C, Saez B, Horejsi V, Martinozzi S, Krawice-Radanne I, Bruel S, et al. et al. IFN-gamma protects short-term ovarian carcinoma cell lines from CTL Characterization of monoclonal antibodies recognizing HLA-G or HLA-E: new lysis via a CD94/NKG2A-dependent mechanism. J Clin Invest 2002;110: tools to analyze the expression of nonclassical HLA class I molecules. 1515–23. Hum Immunol 2003;64:315–26. 98. Blank C, Gajewski TF, Mackensen A. Interaction of PD-L1 on tumor cells with 86. Ravindranath MH, Filippone EJ, Devarajan A, Asgharzadeh S. Enhancing PD-1 on tumor-specific T cells as a mechanism of immune evasion: implications þ natural killer and CD8 t cell-mediated anticancer cytotoxicity and prolifer- for tumor immunotherapy. Cancer Immunol Immunother 2005;54:307–14. þ ation of CD8 t cells with HLA-E monospecific monoclonal antibodies. 99. Hammer Q, Ruckert T, Borst EM, Dunst J, Haubner A, Durek P, et al. Peptide- Monoclon Antib Immunodiagn Immunother 2019;38:38–59. specific recognition of human cytomegalovirus strains controls adaptive nat- 87. Ravindranath MH, Taniguchi M, Chen CW, Ozawa M, Kaneku H, El-Awar N, ural killer cells. Nat Immunol 2018;19:453–63. et al. HLA-E monoclonal antibodies recognize shared peptide sequences on 100. Lopez-Botet M, Muntasell A, Vilches C. The CD94/NKG2Cþ NK-cell subset classical HLA class Ia: relevance to human natural HLA antibodies. on the edge of innate and adaptive immunity to human cytomegalovirus Mol Immunol 2010;47:1121–31. infection. Semin Immunol 2014;26:145–51. 88. Ravindranath MH, Tho P, El-Awar N, Kaneku H, Terasaki PI. Anti-HLA-E 101. Manguso RT, Pope HW, Zimmer MD, Brown FD, Yates KB, Miller BC, et al. mAb 3D12 mimics MEM-E/02 in binding to HLA-B and HLA-C alleles: web- In vivo CRISPR screening identifies Ptpn2 as a cancer immunotherapy target. tools validate the immunogenic epitopes of HLA-E recognized by the anti- Nature 2017;547:413–8. bodies. Mol Immunol 2011;48:423–30. 102. Sharma P, Hu-Lieskovan S, Wargo JA, Ribas A. Primary, adaptive, and acquired 89. Ulbrecht M, Kellermann J, Johnson JP, Weiss EH. Impaired intracellular resistance to cancer immunotherapy. Cell 2017;168:707–23. transport and cell surface expression of nonpolymorphic HLA-E: evidence 103. Ruggeri L, Urbani E, Andre P, Mancusi A, Tosti A, Topini F, et al. Effects of anti- for inefficient peptide binding. J Exp Med 1992;176:1083–90. NKG2A antibody administration on leukemia and normal hematopoietic cells. 90. Kambayashi T, Kraft-Leavy JR, Dauner JG, Sullivan BA, Laur O, Jensen PE. The Haematologica 2016;101:626–33. nonclassical MHC class I molecule Qa-1 forms unstable peptide complexes. 104. Cho M, Bendell JC, Han SW, Naidoo J, Lieu C, Carneiro BA, et al. Durvalumab J Immunol 2004;172:1661–9. plus monalizumab, mFOLFOX6, and bevacizumab in patients (pts) with met- 91. Walters LC, Harlos K, Brackenridge S, Rozbesky D, Barrett JR, Jain V, et al. astatic microsatellite-stable colorectal cancer (MSS-CRC). Ann Oncol 2019;30, Pathogen-derived HLA-E bound epitopes reveal broad primary anchor pocket suppl 5, V490–1.

OF8 Clin Cancer Res; 26(21) November 1, 2020 CLINICAL CANCER RESEARCH

Downloaded from clincancerres.aacrjournals.org on September 26, 2021. © 2020 American Association for Cancer Research. Published OnlineFirst May 14, 2020; DOI: 10.1158/1078-0432.CCR-19-2095

The NKG2A−HLA-E Axis as a Novel Checkpoint in the Tumor Microenvironment

Linda Borst, Sjoerd H. van der Burg and Thorbald van Hall

Clin Cancer Res Published OnlineFirst May 14, 2020.

Updated version Access the most recent version of this article at: doi:10.1158/1078-0432.CCR-19-2095

E-mail alerts Sign up to receive free email-alerts related to this article or journal.

Reprints and To order reprints of this article or to subscribe to the journal, contact the AACR Publications Subscriptions Department at [email protected].

Permissions To request permission to re-use all or part of this article, use this link http://clincancerres.aacrjournals.org/content/early/2020/09/15/1078-0432.CCR-19-2095. Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC) Rightslink site.

Downloaded from clincancerres.aacrjournals.org on September 26, 2021. © 2020 American Association for Cancer Research.