The adaptive immune system can be a potent defense mechanism against cancer; however, it is often hampered by immune suppressive mechanisms in the tumor microenvironment. Coinhibitory molecules expressed by tumor cells, immune cells, and stromal cells in the tumor milieu can dominantly attenuate T-cell responses against cancer cells. Today, a variety of coinhibitory molecules, including cytotoxic T lymphocyte–associated antigen-4, programmed death-1, B and T lymphocyte attenuator, LAG3, T-cell immunoglobulin and mucin domain 3, and CD200 receptor, have been implicated in immune escape of cancer cells. Sustained signaling via these coinhibitory molecules results in functional exhaustion of T cells, during which the ability to proliferate, secrete cytokines, and mediate lysis of tumor cells is sequentially lost. In this review, we discuss the influence of coinhibitory pathways in suppressing autologous and allogeneic T cell–mediated immunity against hematologic malignancies. In addition, promising preclinical and clinical data of immunotherapeutic approaches interfering with negative cosignaling, either as monotherapy or in conjunction with vaccination strategies, are reviewed. Numerous studies indicate that coinhibitory signaling hampers the clinical benefit of current immunotherapies. Therefore, manipulation of coinhibitory networks is an attractive adjuvant immunotherapeutic intervention for hematologic cancers after standard treatment with chemotherapy and hematopoietic stem cell transplantation.

Despite the powerful aspects of immune reactions, most often tumor cells are able to evade immune recognition and destruction. Mechanisms exploited by tumor cells to escape T cell–mediated immunity include disruption of antigen presentation, down-regulation of HLA molecules, secretion of immune suppressive cytokines, as well as recruitment of regulatory T cells (TREG) and myeloid-derived suppressor cells.1  In the last decade, another powerful immune suppressive mechanism gained much attention: the repressive action of coinhibitory molecules.2  Activation of T cells is predominantly dependent on both costimulatory and coinhibitory members, including members of the B7/ CD28 family. The balance between positive and negative cosignals determines the functionality of T cells during immunity and tolerance. In addition to the native role of cosignaling, tumor cells can evade immune control by down-regulating costimulatory molecules, such as CD80 and CD86, and up-regulating various coinhibitory ligands, thereby limiting the therapeutic potential of current immunotherapy against cancer.

Standard treatment for hematologic cancers includes chemotherapy and radiotherapy, which reduce tumor burden and can induce long-term remission. Moreover, in the past years, new therapeutics, including imatinib, dasatinib, rituximab, bortezomib, and lenalidomide, have been developed that target tumor cells. However, drug resistance and relapse remain major problems. In addition, cellular immunotherapy is an attractive treatment option to cure hematologic malignancies. Such cell-based immunotherapies include allogeneic stem cell transplantation (alloSCT), T-cell and NK-cell adoptive transfer, and vaccination-based approaches using various antigen formulations or dendritic cells (DCs). AlloSCT can be regarded as the most powerful cell-based immunotherapy, because of the graft-versus-tumor (GVT) responses constituted by alloreactive T cells.3  These alloreactive T-cell responses eradicate the malignant cells on recognition of polymorphic HLA-presented peptides, known as minor histocompatibility antigens (MiHA). AlloSCT greatly enhanced the cure rate for aggressive hematologic cancers, although many patients fail to launch productive immune responses and develop relapses. Moreover, a major drawback of alloSCT is the occurrence of GVHD, a potentially life-threatening side effect predominantly caused by alloreactive T cells recognizing healthy tissues, notably the skin, liver, and gastrointestinal tract. Because hemato-restricted MiHA are solely expressed by the redundant patient hematopoietic system and the hematologic malignancy, they hold the key to separate GVT from GVHD.4  Studies by us and others demonstrated that the cellular immunotherapies described earlier in the “Introduction” are often hampered by the action of coinhibitory molecules that attenuate tumor-reactive T-cell responses, resulting in suboptimal clinical results. This review will address the role of coinhibitory molecules in immune evasion by hematologic malignancies and discuss options to circumvent T-cell inhibition without severe adverse effects. In addition, we address whether a differential effect of coinhibitory molecules exists in GVT and GVHD, creating an opportunity to limit GVHD toxicity without dampening antitumor immunity.

Today a variety of coinhibitory molecules have been implicated in immune escape of cancer. Here, we discuss the coinhibitory molecules involved in suppressing antitumor immunity against hematologic malignancies (summarized in Table 1).

Table 1

Major coinhibitory molecules and their corresponding binding partners involved in attenuating antitumor immunity

Receptor
Binding partners
NameExpression patternNameExpression on normal cellsExpression on malignant cells
CTLA-4 Activated T, TREG CD80/CD86 T, B, DCs, macrophages Down-regulated on AML, MM18,20  
PD-1 Activated T and B, NKT, monocytes, myeloid cells PD-L1 Activated T, B, DCs, macrophages, monocytes, nonlymphoid tissues AML, NHL, MM20,32  
  PD-L2 DCs, monocytes AML, NHL, MM20,32  
BTLA T, B, DCs, myeloid cells HVEM T, B, DCs, NK, myeloid cells and nonlymphoid tissues AML, CLL, NHL, MM67,68,75  
LAG-3 Activated T, TREG, B, pDCs, NK MHC-II Activated T, B, DCs, macrophages, monocytes, endothelium Down-regulated in tumors1  
TIM-3 Th1 CD4+ T, CD8+ T, DCs, NK, monocytes, epithelium Galectin-9 CD4 T cells, Treg, DCs, fibroblasts, granulocytes, endothelium AML, lymphoma93,94  
CD200R Activated T, B, NK, DCs, mast cells, myeloid cells, neutrophils CD200 Activated T, B, DCs, thymocytes, endothelium, nonlymphoid tissues AML, CLL, MM84,85,108  
Receptor
Binding partners
NameExpression patternNameExpression on normal cellsExpression on malignant cells
CTLA-4 Activated T, TREG CD80/CD86 T, B, DCs, macrophages Down-regulated on AML, MM18,20  
PD-1 Activated T and B, NKT, monocytes, myeloid cells PD-L1 Activated T, B, DCs, macrophages, monocytes, nonlymphoid tissues AML, NHL, MM20,32  
  PD-L2 DCs, monocytes AML, NHL, MM20,32  
BTLA T, B, DCs, myeloid cells HVEM T, B, DCs, NK, myeloid cells and nonlymphoid tissues AML, CLL, NHL, MM67,68,75  
LAG-3 Activated T, TREG, B, pDCs, NK MHC-II Activated T, B, DCs, macrophages, monocytes, endothelium Down-regulated in tumors1  
TIM-3 Th1 CD4+ T, CD8+ T, DCs, NK, monocytes, epithelium Galectin-9 CD4 T cells, Treg, DCs, fibroblasts, granulocytes, endothelium AML, lymphoma93,94  
CD200R Activated T, B, NK, DCs, mast cells, myeloid cells, neutrophils CD200 Activated T, B, DCs, thymocytes, endothelium, nonlymphoid tissues AML, CLL, MM84,85,108  

T indicates T cells; B, B cells; pDCs, plasmacytoid dendritic cells; NK(T), natural killer (T) cells; CLL, chronic lymphoid leukemia; and (B-)NHL, (B-cell) non-Hodgkin lymphoma.

CTLA-4

Expression and function of CTLA-4.

Cytotoxic T lymphocyte associated antigen-4 (CTLA-4; CD152) was the first coinhibitory molecule identified and is partly similar to the cosignaling molecule CD28.5  However, whereas CD28 is constitutively expressed on the membrane of naive T cells, CTLA-4 is primarily localized in intracellular compartments and rapidly translocates to the cell membrane on T-cell activation. The inhibitory function of CTLA-4 was revealed in knockout mice, which developed lethal lymphoproliferative disease with multiorgan T-cell infiltration.6  Like CD28, CTLA-4 has an extracellular domain containing the MYPPPY binding motif, enabling both receptors to interact with CD80 (B7-1) and CD86 (B7-2) expressed by antigen-presenting cells (APCs). However, the binding affinity of CTLA-4 for these ligands is 10- to 100-fold higher, thereby outcompeting CD28 and promoting immune inhibition.7 

As CTLA-4 is up-regulated on TCR ligation, it plays an important role in dampening effector T-cell activation and regulating immune homeostasis. In addition, CTLA-4 signaling in immunosuppressive TREG mediates the control of autoreactive T cells, as in vivo interference with CTLA-4 on these cells elicited pathologic autoimmunity.8  The effect of CTLA-4 interference could be the result of depletion and/or inhibition of TREG. Wing et al showed that TREG-specific CTLA-4 deficiency resulted in impaired suppressive TREG function because CTLA-4 enables the down-regulation of CD80/CD86 on APCs,9  which can be partly the result of endocytosis of CD80 and CD86 by TREG.10  This renders a less stimulatory APC, resulting in a lasting cell-extrinsic inhibitory effect. CTLA-4 signaling can attenuate adaptive immune responses in chronic viral infections and cancer. CTLA-4 as such is not a marker of exhausted cells, but elevated levels on viral antigen-specific T cells correlated with their dysfunction in patients with chronic viral infections, which could be restored by CTLA-4 blockade.11  In addition, in cancer, high expression of CTLA-4 was correlated to antigen-specific T cell dysfunction in metastatic melanoma.12  In various CD80 and CD86-positive solid tumor models, monotherapy with CTLA-4 blocking antibody resulted in elimination of established tumors and long-lasting antitumor immunity.13  Several clinical trials have been performed with anti–CTLA-4 antibodies, mostly with ipilimumab in melanoma. Interestingly, an increase in overall survival of melanoma patients has been observed.14  However, not all patients gain clinical benefit, and individual responses are hard to predict. Furthermore, the occurrence of adverse toxic effects remains a problem. Interestingly, in one trial, patients responding to ipilimumab were reported to have high titers of anti-MICA antibodies, probably because of enhanced CD4+ T-cell function resulting in increased antibody responses. These antibodies may revert the functional inhibition of NK and CD8+ T cells induced by tumor-secreted MICA. In 2011, the FDA and European Medicines Agency approved ipilimumab treatment for advanced melanoma, paving the way for further exploration of therapies targeting coinhibitory molecules in cancer.15  Although anti–CTLA-4 treatment works in vivo, either alone or in combination with vaccines, in vitro CTLA-4 blockade has not been very successful in reversing T-cell dysfunction. This might be the result of limitations of the in vitro models, as CTLA-4 blockade probably exerts its in vivo action via multiple immune mediators (eg, effector T cells, TREG, antibody responses).16 

CTLA-4 in hematologic malignancies.

Numerous experimental and clinical studies have demonstrated that coinhibitory molecules hamper T-cell immunity against hematologic cancers in both the autologous and allogeneic settings (Tables 2 and 3). For instance, a causal relationship between CTLA-4 and TREG was demonstrated in lymphoma patients.17  A large proportion of the lymphoma-infiltrating lymphocytes was identified as CTLA-4+ TREG, and TREG-mediated T-cell suppression could be abrogated by CTLA-4 blockade. In addition, CTLA-4:CD80/86 interactions also take place between T cells and tumor cells. In multiple myeloma (MM) patients, CD86 but not CD80 was expressed by tumor cells, whereas CTLA-4 was up-regulated on T cells, resulting in anergy of tumor-specific T cells.18  In concordance with these results, T cells from chronic lymphocytic leukemia patients responded to anti-CD3 activation by a decrease in CD28 and an increase in CTLA-4 expression, resulting in an inhibitory phenotype.19  Similar to MM, we and others showed that acute myeloid leukemia (AML) cells heterogeneously express CD86, but CD80 levels are generally low or absent.20,21  As CD80 and CD86 can mediate either T-cell stimulation via CD28 or T-cell inhibition via CTLA-4, their role in the induction of tumor-specific T-cell immunity was investigated in an AML model.22  Expression of CD86 on AML resulted in tumor rejection, whereas CD80+ AML tumors grew progressively. The latter observation was shown to be CTLA-4 dependent, as blockade with anti–CTLA-4 resulted in clearance of CD80+ AML cells.

Table 2

Outcome of interference with murine coinhibitory molecules

MoleculeTherapyTumorOutcomeReference
Autologous     
    CTLA-4 Anti–CTLA-4 + ova-DC vaccination Thymoma Improved tumor rejection, enhanced antigen-specific T-cell responses 23  
    CTLA-4 Anti–CTLA-4; CTLA-4 deletional knockout AML Increased survival and improved tumor rejection 22  
    CTLA-4 Anti–CTLA-4 AML Enhanced T-cell response, prolonged survival 54  
    PD-1 Anti–PD-L1; PD-1 knockout MM Delayed tumor growth; complete tumor rejection in PD-1 knockout 49  
    PD-1 HSCT + whole cell vaccination + anti–PD-L1 MM Increased survival 52  
    PD-1 Anti–PD-L1; PD-1 knockout AML Enhanced T-cell response, improved tumor rejection, increased survival 50  
    PD-1/TIM-3 Anti–PD-L1 and/or mTim-3 hFc AML Delayed tumor growth on monotherapy, improved tumor rejection on combined blockade 93  
    CD200 Anti-CD200 AML Increased survival 86  
    CD200 Anti-CD200 B-CLL Improved tumor rejection 87  
    CD200 Anti-CD200 B-cell lymphoma Delayed tumor growth 88  
Allogeneic     
    CTLA-4 Anti–CTLA-4 AML Enhanced T-cell response and GVHD early after BMT; enhanced tumor-specific T-cell response later after BMT with low GVHD 28  
    PD-1 Anti–PD-L1 Lymphoma Enhanced T-cell response 63  
    PD-1 Anti–PD-L1 None Enhanced alloreactive T-cell response, no GVHD 64  
    PD-1 Anti–PD-L1; PD-1 knockout CML Increased survival 65  
    BTLA BTLA agonist B-cell lymphoma Early after BMT: prevention of GVHD; later after BMT: no effect on GVHD; effective GVT response 78  
    BTLA BTLA agonist Mastocytoma/T-cell lymphoma Inhibition of alloreactive T-cell response, prevention from GVHD 79  
    HVEM Specific blockade of BTLA binding None Prevention from acute GVHD 80  
    PD-1H/ VISTA PD-1H/VISTA agonist None Prevention from acute GVHD 103  
MoleculeTherapyTumorOutcomeReference
Autologous     
    CTLA-4 Anti–CTLA-4 + ova-DC vaccination Thymoma Improved tumor rejection, enhanced antigen-specific T-cell responses 23  
    CTLA-4 Anti–CTLA-4; CTLA-4 deletional knockout AML Increased survival and improved tumor rejection 22  
    CTLA-4 Anti–CTLA-4 AML Enhanced T-cell response, prolonged survival 54  
    PD-1 Anti–PD-L1; PD-1 knockout MM Delayed tumor growth; complete tumor rejection in PD-1 knockout 49  
    PD-1 HSCT + whole cell vaccination + anti–PD-L1 MM Increased survival 52  
    PD-1 Anti–PD-L1; PD-1 knockout AML Enhanced T-cell response, improved tumor rejection, increased survival 50  
    PD-1/TIM-3 Anti–PD-L1 and/or mTim-3 hFc AML Delayed tumor growth on monotherapy, improved tumor rejection on combined blockade 93  
    CD200 Anti-CD200 AML Increased survival 86  
    CD200 Anti-CD200 B-CLL Improved tumor rejection 87  
    CD200 Anti-CD200 B-cell lymphoma Delayed tumor growth 88  
Allogeneic     
    CTLA-4 Anti–CTLA-4 AML Enhanced T-cell response and GVHD early after BMT; enhanced tumor-specific T-cell response later after BMT with low GVHD 28  
    PD-1 Anti–PD-L1 Lymphoma Enhanced T-cell response 63  
    PD-1 Anti–PD-L1 None Enhanced alloreactive T-cell response, no GVHD 64  
    PD-1 Anti–PD-L1; PD-1 knockout CML Increased survival 65  
    BTLA BTLA agonist B-cell lymphoma Early after BMT: prevention of GVHD; later after BMT: no effect on GVHD; effective GVT response 78  
    BTLA BTLA agonist Mastocytoma/T-cell lymphoma Inhibition of alloreactive T-cell response, prevention from GVHD 79  
    HVEM Specific blockade of BTLA binding None Prevention from acute GVHD 80  
    PD-1H/ VISTA PD-1H/VISTA agonist None Prevention from acute GVHD 103  

B-CLL indicates B-cell chronic lymphoid leukemia; and BMT, bone marrow transplantation.

Table 3

Outcome of interference with human coinhibitory molecules

MoleculeTherapyTumorOutcomeReference
CTLA-4 Ipilimumab, in vivo NHL Two of 4 tumor regression, no increase in vaccine-specific T-cell responses, reduction in TREG number early after treatment; toxicity: mainly grade 1 or 2, 1 times grade 3 24  
CTLA-4 Ipilimumab, in vivo Relapsed/refractory B-cell NHL Two of 18 clinical response, 5 of 16 enhanced T-cell response to recall Ag; toxicity: mainly grade 1 or 2, 6 of 18 grade 3 25  
CTLA-4 Ipilimumab after alloSCT, in vivo AML, CML, CLL, HL, NHL, MM Three of 29 clinical response; toxicity: no induction of GVHD, 4 of 29 organ-specific immune adverse events 29  
CTLA-4 Anti–CTLA-4, ex vivo HL Abrogated TREG suppression 17  
CTLA-4 Anti–CTLA-4, ex vivo CLL Enhanced tumor-specific T-cell response 109  
PD-1 BMS-936,558, ex vivo ALL, AML, CML, NHL, MM Enhanced alloreactive T-cell response 20  
PD-1 Anti–PD-L1, ex vivo NHL Enhanced T-cell response 52  
PD-1 Anti–PD-L1 and anti–PD-L2, ex vivo HL Restored T-cell response 53  
PD-1 Anti–PD-L1, ex vivo HCV lymphoma Abrogated TREG suppression, reduction in Treg number 55  
PD-1 CT-011, in vivo AML, CLL, HL, NHL, MDS, MM Six of 17 clinical response, 1 complete remission, no toxicity 57  
PD-1 CT-011 with or without lenalidomide, ex vivo MM Enhanced NK cytotoxicity, additive effect of lenalidomide 58  
PD-1 CT-011 + tumor/DC vaccination, ex vivo MM Reduction in TREG number, enhanced T-cell response 59  
PD-1 DC vaccination with PD-L silencing, ex vivo AML, CML Enhanced alloreactive T-cell response 105  
BTLA Anti-BTLA, ex vivo ALL, AML, CML, NHL, MM Enhanced alloreactive T-cell response 67  
CD200 Anti-CD200, ex vivo AML Enhanced NK cytotoxicity 84  
CD200 Anti-CD200, ex vivo CLL Enhanced antigen-specific T-cell responses, reduction in Treg number 85  
MoleculeTherapyTumorOutcomeReference
CTLA-4 Ipilimumab, in vivo NHL Two of 4 tumor regression, no increase in vaccine-specific T-cell responses, reduction in TREG number early after treatment; toxicity: mainly grade 1 or 2, 1 times grade 3 24  
CTLA-4 Ipilimumab, in vivo Relapsed/refractory B-cell NHL Two of 18 clinical response, 5 of 16 enhanced T-cell response to recall Ag; toxicity: mainly grade 1 or 2, 6 of 18 grade 3 25  
CTLA-4 Ipilimumab after alloSCT, in vivo AML, CML, CLL, HL, NHL, MM Three of 29 clinical response; toxicity: no induction of GVHD, 4 of 29 organ-specific immune adverse events 29  
CTLA-4 Anti–CTLA-4, ex vivo HL Abrogated TREG suppression 17  
CTLA-4 Anti–CTLA-4, ex vivo CLL Enhanced tumor-specific T-cell response 109  
PD-1 BMS-936,558, ex vivo ALL, AML, CML, NHL, MM Enhanced alloreactive T-cell response 20  
PD-1 Anti–PD-L1, ex vivo NHL Enhanced T-cell response 52  
PD-1 Anti–PD-L1 and anti–PD-L2, ex vivo HL Restored T-cell response 53  
PD-1 Anti–PD-L1, ex vivo HCV lymphoma Abrogated TREG suppression, reduction in Treg number 55  
PD-1 CT-011, in vivo AML, CLL, HL, NHL, MDS, MM Six of 17 clinical response, 1 complete remission, no toxicity 57  
PD-1 CT-011 with or without lenalidomide, ex vivo MM Enhanced NK cytotoxicity, additive effect of lenalidomide 58  
PD-1 CT-011 + tumor/DC vaccination, ex vivo MM Reduction in TREG number, enhanced T-cell response 59  
PD-1 DC vaccination with PD-L silencing, ex vivo AML, CML Enhanced alloreactive T-cell response 105  
BTLA Anti-BTLA, ex vivo ALL, AML, CML, NHL, MM Enhanced alloreactive T-cell response 67  
CD200 Anti-CD200, ex vivo AML Enhanced NK cytotoxicity 84  
CD200 Anti-CD200, ex vivo CLL Enhanced antigen-specific T-cell responses, reduction in Treg number 85  

ALL indicates acute lymphoid leukemia; CLL, chronic lymphoid leukemia; NHL, non-Hodgkin lymphoma; MDS, myelodysplastic syndrome; HCV, hepatitis C virus; and GI, gastrointestinal tract.

Because of their potent suppressive function, coinhibitory molecules became major targets of preclinical and clinical blocking studies. For example, in a murine thymoma model, CTLA-4 blockade after DC vaccination improved survival and resulted in a sustained increase in the number of antigen-specific T cells.23  In a phase 1 study that included 4 non-Hodgkin lymphoma patients, 2 subjects developed a clinical response on ipilimumab treatment.24  No enhanced T cell–mediated antitumor reactivity could be observed, although TREG levels decreased, suggesting that CTLA-4's effectiveness may be attributed to TREG depletion via antibody-dependent cell-mediated cytotoxicity. In a follow-up study with 18 non-Hodgkin lymphoma patients, ipilimumab administration resulted in clinical responses in 2 patients, and in several patients enhanced T-cell responses against KLH and tetanus toxoid were observed.25  Overall toxic effects were limited in these studies; and although durable responses were rare, the response rate resembled that of the first clinical trials in solid cancers. Because only small numbers of patients with hematologic malignancies have been treated so far, more research is warranted to draw conclusions.

Allogeneic T-cell function after alloSCT is also strongly influenced by coinhibitory molecules. The importance of CTLA-4 in modulating allogeneic immune responses has been confirmed by the association of certain CTLA-4 genotypes with the incidence of leukemia relapse and overall survival after alloSCT.26  Although not all functional consequences of reported polymorphisms have been elucidated, the CT60 single nucleotide polymorphism is postulated to influence the transcription of the sCTLA-4 variant, hampering normal CTLA-4 function.27  Interestingly, it was demonstrated that CTLA-4 blockade shortly after alloSCT increased GVHD in a CD28-dependent manner.28  However, when anti–CTLA-4 was administered at later time points after alloSCT, the GVT effect was boosted without signs of GVHD. Shortly after alloSCT, conditioning-related mucosal barrier injury, leading to a proinflammatory cytokine storm, tissue damage, and inflammation, may induce major T-cell activation in GVHD tissues. However, at later time points, these inflammatory events have diminished, and there is no general T-cell activation. In patients, ipilimumab administration at late time points after alloSCT has been explored in one phase 1 trial.29  After a single infusion of ipilimumab in 29 alloSCT patients with a recurrent or progressive hematologic malignancy, 3 clinical responses were observed. Importantly, no induction or exacerbation of clinical GVHD was reported, although, similar to other CTLA-4 blockade trials, 14% of the patients showed organ-specific immune adverse events. The lack of GVHD induction is probably attributed to the median interval of 1 year between last donor cell infusion and ipilimumab administration. This provides a window for antitumor immunotherapy in the posttransplantation setting and emphasizes the importance of appropriate timing.

PD-1

Expression and function of PD-1.

Programmed death-1 (PD-1; CD279) is another immunoreceptor belonging to the B7/CD28 family.30  In 1992, PD-1 was identified on hybridoma T cells undergoing apoptosis and was thought to be a programmed cell death-induced gene.31  Further characterization demonstrated that PD-1 is inducibly expressed on stimulated CD4+ T cells, CD8+ T cells, B cells, and monocytes.32  PD-1 binds 2 B7 family ligands, PD-L1 (B7-H1; CD274) and PD-L2 (B7-DC; CD273).33  Their interaction with PD-1 differs in affinity34  and type because of a conformational transition in PD-L1, but not PD-L2, on binding.35  Although PD-L2 expression is mainly restricted to APCs, such as DCs and macrophages, PD-L1 is expressed on many nonlymphoid tissues as well.36  Furthermore, multiple tumor types express PD-L1, and its expression is elevated after IFN-γ exposure.37  PD-L1 molecules on tumor cells can deliver negative signals toward PD-1–expressing tumor-reactive T cells, thereby inhibiting antitumor immunity.38  Indeed, PD-L1 expression has been associated with poor prognosis in solid tumors.37,39  Interestingly, PD-L1 is also able to bind CD80, mediating T-cell inhibition.40  In addition to downstream signaling of PD-L1,41  also engagement of PD-L2 resulted in T-cell inhibition, further illustrating the complexity of these interactions.42 

It has been well demonstrated that PD-1 plays a crucial role in T-cell regulation in various immune responses, such as peripheral tolerance, autoimmunity, infection, and antitumor immunity.36  Elevated PD-1 expression on viral antigen-specific CD8+ T cells in chronic viral infections was recognized as a hallmark for T-cell dysfunction on antigen restimulation.43  This phenomenon known as exhaustion is characterized by the sequential loss of the ability to proliferate, secrete cytokines, and lyse target cells. Especially in HIV infection, T-cell impairment could be relieved by PD-1 blockade both in vitro and in animal models.44,45  Exhausted T cells have increased expression of multiple coinhibitory receptors and a distinct gene signature, different from anergic cells, resulting in changes in TCR and cytokine signaling pathways.46  Indeed, an exhaustion-specific gene signature, recently defined by Quigley et al,47  demonstrated that PD-1 downstream signaling effects play an important role in the exhaustion of HIV-specific T cells. Furthermore, they showed that the transcription factor BATF (basic leucine zipper transcription factor, ATF-like) appears essential for downstream PD-1 signaling. In addition to these signaling effects, the PD-1 gene itself is subject to epigenetic regulation, as increased PD-1 expression on activated CD8+ T cells results from demethylation of the Pdcd1 locus.48  During conversion to functional memory T cells, remethylation of Pdcd1 occurs, whereas in exhausted T cells the Pdcd1 regulatory region remains demethylated.

PD-1 in hematologic malignancies.

In addition to CTLA-4, PD-1/PD-L interactions were shown to be of importance in hematologic malignancies. For instance, PD-L1 overexpression enhanced MM invasiveness and rendered tumor cells less susceptible to cytotoxic T lymphocytes (CTLs).49  This effect was alleviated in PD-1 knockout mice or by anti–PD-L1 antibody treatment, demonstrating the importance of the PD-1/PD-L pathway in this process. This role of PD-1 was also confirmed in an AML model, and interestingly, PD-L1 expression was elevated on tumor cells in vivo compared with in vitro.50  In another report, increased levels of PD-L1 on MM cells together with enhanced PD-1 expression on exhausted T cells was demonstrated.51  As expected, in mice, PD-L1 blockade improved survival after autologous SCT and whole cell vaccination from 0% to 40%. In humans, PD-L1 expression was observed on non-Hodgkin lymphoma tumor cells, and blockade greatly enhanced cytokine production of autologous tumor-reactive T cells.52  Furthermore, it was shown that tumor cells of Hodgkin lymphoma (HL) patients can express both PD-L1 and PD-L2, and PD-1 expression was elevated on HL-infiltrating T cells.53  In addition, in this case, blockade of PD-Ls mediated increased cytokine secretion by the infiltrated T cells. Furthermore, long-term persistent murine leukemia cells were shown to sequentially up-regulate PD-L1 and CD80, thereby conferring protection against immune destruction.54  On PD-L1 or CTLA-4 blockade, CTL-mediated lysis of these persistent AML cells was improved. Similar to the link of CTLA-4 and TREG, an elevated number of TREG exhibiting high PD-1 expression was described in HCV-associated lymphoma.55  In addition to PD-1 expression on CD8+ T cells and TREG, PD-L1 expression on APCs was important for tumor persistence of murine AML. Combining PD-L1 blockade with TREG depletion showed superior efficacy in clearance of AML because of alleviation of PD-1–dependent TREG-mediated suppression.56 

Although clinical PD-1 blockade has not been as extensively tested as ipilimumab for CTLA-4, multiple clinical grade antagonistic anti–PD-1 antibodies have been developed (ie, CT-011, BMS-936,558 and MK-3475; NCT01295827). Furthermore, 2 anti–PD-L1 antibodies, BMS-936,559 (NCT00729664) and MPDL2180A (NCT01375842), one anti–PD-L2 antibody (NCT00658892), and a PD-L2 fusion protein AMP-224 (NCT01352884) are being tested in phase 1 clinical trials. Three studies involving hematologic cancers were performed with CT-011. One phase 1 clinical trial was conducted in patients with various hematologic malignancies and showed a clinical response in 6 of 17 patients, with few adverse events.57  Although the CD4+ T-cell count was elevated in the treated patients, no additional evidence of T-cell activation was found. In a preclinical study, ex vivo treatment with CT-011 enhanced the functionality of NK cells against autologous primary MM cells.58  In addition, the drug lenalidomide down-regulated PD-L1, and an additive effect was shown by combining lenalidomide with CT-011, rendering this combination a promising therapy for MM patients. Another study examined whether PD-1 blockade improves the effectiveness of myeloma/DC vaccination therapy because it is known that both myeloma cells and myeloma/DC hybridomas highly express PD-L1.59  Indeed, ex vivo addition of CT-011 resulted in enhanced myeloma lysis by T cells as well as a reduction in the number of TREG. However, until now, the most promising effects have been obtained with the monoclonal human anti–PD-1 antibody BMS-936,558 (MDX-1106; ONO-4538). Administration to patients with solid tumors was well tolerated, and only one serious adverse event (inflammatory colitis) was reported.60  Follow-up reports presented at ASCO 2010 and GU ASCO 2011 showed that persistent clinical responses were observed in approximately 30% of patients with renal cell carcinoma, prostate cancer, melanoma, and lung cancer on treatment with repetitive doses of anti–PD-1 antibodies.61,62  The lack of strong toxic effects in this study holds promise that PD-1 blockade might have a more subtle effect than CTLA-4 blockade, thereby highlighting anti–PD-1 antibodies as interesting candidates for cancer therapy.

The role of PD-1 in alloSCT has been investigated both in mice and men. In 2 similar murine studies dissecting the role of alloantigens in GVT and GVHD reactivity, it was found that alloreactive T cells recognizing antigens on GVHD-prone tissues are driven into dysfunction and apoptosis.63  Furthermore, the interaction of nonhematopoietic cells with alloreactive T cells prevented the formation of proper alloreactive memory cells by exploiting the PD-1/PD-L pathway.64  This means that, in addition to the detrimental effect of GVHD as such, the beneficial GVT effect is hampered as alloreactive T cells become functionally impaired. Notably, PD-L1 blockade late after alloSCT may partly restore the GVT reactivity without inducing GVHD. Moreover, these results support the importance of targeting hematopoietic-restricted MiHA because these are solely expressed by hematopoietic tumor cells and residual healthy immune cells of the recipient, but not by GVHD-prone tissues. In one of the few studies investigating chronic myeloid leukemia (CML), using a retrovirus-induced CML model, it was demonstrated that tumor-specific T cells can become exhausted.65  In this model, consisting of PD-1+ tumor-specific T cells and PD-L1+ CML cells, exhaustion was overcome using either PD-1–deficient cells or anti–PD-L1 administration. We and others have investigated the role of PD-1 in GVT immunity in alloSCT patients. High PD-1 expression was observed on alloreactive CD8+ TEM cells that specifically recognize hematopoietic-restricted MiHA in myeloid leukemia patients.20  In agreement, Mumprecht et al showed that the total T-cell population from CML patients had elevated levels of PD-1.65  In addition, CD117+ progenitor AML cells displayed low levels of CD80 and CD86, whereas PD-L1 was highly expressed, especially under inflammatory conditions.20  Because these observations were made in alloSCT patients who relapsed after initial powerful MiHA-specific T-cell responses, we postulated that PD-1 expression is involved in T-cell exhaustion. By stimulation with MiHA-loaded DC ex vivo, we aimed at activating these PD-1+ MiHA-specific TEM cells; however, results were suboptimal, suggesting an impaired state. Importantly, on treatment with anti–PD-1 or anti–PD-L1 blocking antibodies, we were able to reinvigorate MiHA-specific TEM proliferation. Notably, the effect of PD-1 blockade on MiHA-specific TEM cells from relapsed patients compared with patients in long-term remission was significantly stronger, indicating the function of PD-1 in T-cell exhaustion and subsequent tumor immune evasion.

BTLA

Expression and function of BTLA.

B and T lymphocyte attenuator (BTLA), that is, CD272, was identified in 2003 as an inhibitory receptor with structural similarities to CTLA-4 and PD-1.66  BTLA is mainly expressed by immune cells, including T and B cells, DCs, and myeloid cells.67,68  In contrast to other B7/CD28 family members, BTLA binds a member of the tumor necrosis factor receptor superfamily, namely, herpesvirus entry mediator (HVEM).69  Although HVEM is part of an intricate signaling network as it has at least 4 additional binding partners that distinctively mediate T-cell responses (ie, CD160, LIGHT; for lymphotoxin-like, exhibits inducible expression, and competes with HSV glycoprotein D for HVEM, a receptor expressed by T lymphocytes), lymphotoxin-α (LT-α) and herpes simplex virus glycoprotein D.70  BTLA or CD160 signaling on HVEM binding results in T-cell inhibition.69,71  However, HVEM present on T cells acts as a costimulatory receptor,72  thereby constituting a bidirectional pathway. Interestingly, naive T cells express both HVEM and BTLA, and these molecules form a T cell–intrinsic heterodimer complex.73  Because of formation of this complex, HVEM is unavailable for extrinsic ligands, and no costimulatory signal is transduced. Studies in BTLA-deficient mice revealed a predisposition to experimentally induced autoimmune encephalomyelitis.66  Furthermore, these mice develop late-onset spontaneous autoimmune hepatitis-like disease and multiorgan lymphocyte infiltration,74  implying the involvement of BTLA in maintaining self-tolerance. In humans, persistent expression of BTLA was observed on EBV- and CMV-specific CD8+ T cells, negatively affecting T-cell function.75,76  Furthermore, in melanoma patients, high BTLA expression correlated with impaired tumor-specific T-cell function.12,75  These tumor-specific T-cell responses could be restored in vitro by interference with the BTLA-HVEM pathway in combination with vaccination therapy. In addition, coexpression of PD-1, BTLA, and T-cell immunoglobulin and mucin domain 3 (TIM-3) rendered melanoma-specific CD8+ T cells highly dysfunctional, which could be restored by combined blockade of all 3 coinhibitory molecules.77 

BTLA in hematologic malignancies.

A single administration of agonistic anti-BTLA antibody directly after alloSCT in mice completely prevented GVHD, whereas this treatment did not hamper GVT responses.78,79  Moreover, when using an antibody that specifically blocked the interaction of HVEM with BTLA, but not LIGHT, GVHD was attenuated.80  This suggests that in this model the costimulatory function of HVEM was dominant over its coinhibitory activity. Because both HVEM and BTLA can be expressed by T cells, bidirectional signaling can occur. Therefore, combining specific blocking antibodies with cell-specific knockout models for these molecules will be necessary to further unravel the intricate interactions between HVEM, BTLA, CD160, and LIGHT. In addition, we investigated the effect of a BTLA-blocking antibody on MiHA-specific T-cell function in alloSCT patients.67  As shown for PD-1, we observed that BTLA was also highly expressed on MiHA-specific TEM cells. Moreover, in 7 of 11 patients, BTLA blockade resulted in increased outgrowth of MiHA-specific TEM cells of transplanted patients. Interestingly, in 3 patients, BTLA blockade effects were more prominent than those of PD-1, indicating that BTLA has a nonredundant function to PD-1; therefore, it holds promise in post-SCT therapies.

New coinhibitory players

In addition to the previously discussed molecules, CD200 receptor (CD200R), TIM-3, LAG3, and PD-1H/VISTA were recently shown to contribute to T-cell inhibition and/or exhaustion in hematologic cancers.

CD200R is an inhibitory receptor previously thought to be most important on myeloid cells but is also expressed in the lymphoid lineage, such as NK, CD4+, and CD8+ T cells, especially on stimulation.81  Its ligand, CD200 (OX2), is a glycoprotein expressed on a broad number of cell types, including solid tumors and hematologic malignancies.82,83  In addition to the previously discussed coinhibitory molecules, CD200R inhibits both T- and NK-cell functionality.84,85  Furthermore, CD200/CD200R interactions are involved in tumor immune evasion, as CD200 expression on AML cells promoted tumor growth in mice.86  Interestingly, patients with CD200+ AML cells displayed a lower number of activated NK cells, and the effect on NK functionality was correlated to CD200 expression on the leukemia cells.84  In concordance with this, blockade of CD200 enhanced IFN-γ release and cytotoxicity by NK cells. Moreover, CD200 blockade restored T-cell proliferation and tumor control by immune cells for human CD200+ chronic lymphocytic leukemia both in vitro85  and in a humanized mouse model.87  However, in a follow-up report, treatment with anti-CD200 antibody caused loss of T cell–mediated tumor control because of clearance of the T cells.88  This was attributed to antibody-dependent cell-mediated cytotoxicity of CD200+ T cells caused by the IgG1 variant of anti-CD200, which was not observed for the IgG4 isotype. Therefore, blocking strategies should be carefully designed before proceeding to the clinic.

In addition, the cosignaling receptor TIM-3 is expressed on Th1 CD4+ and CD8+ T cells, and is involved in coinhibition. In mice, the interaction of TIM-3 with its ligand galectin-9 was demonstrated to be inhibitory in autoimmune diseases and malignancies.89  Furthermore, in HIV90  and melanoma patients,91  dysfunctional T cells have been shown to coexpress TIM-3. In this regard, interference with TIM-3 signaling is an interesting treatment option, and enhanced tumor vaccine efficacy has been observed by TIM-3 blockade.92  Interestingly, both TIM-3 and PD-1 were expressed on a subset of exhausted CD8+ T cells in a murine AML model and expression increased during tumor progression.93  Although either TIM-3 or PD-L1 blockade alone was not sufficient to improve survival, a combination of the 2 antibodies decreased tumor burden and enhanced survival. Furthermore, in human lymphoma, an interesting role for TIM-3 has been described on tumor endothelium.94  TIM-3 expressed on these endothelial cells mediated impaired CD4+ T-cell responses, and thereby promoted lymphoma onset, growth, and dissemination. In contrast, a stimulatory role for TIM-3 and galectin-9 has been reported in the interaction of CD8+ T cells and DCs.95  This discrepancy is reflected in research investigating its mechanism of action, where T-cell receptor stimulation is enhanced on TIM-3 signaling.96  This might be explained by the fact that T-cell exhaustion could be caused by prolonged TCR signaling, and TIM-3 accelerates this process. Another explanation is that, depending on which ligand binds to TIM-3, different modes of signaling are initiated. Therefore, TIM-3 may act as either a costimulatory or a coinhibitory factor, similar to BTLA.

Lymphocyte-activation gene 3 (LAG3; CD223) is a coinhibitory receptor highly similar to CD4 and binds HLA class II molecules.97,98  Importantly, LAG3 was implicated to inhibit T-cell function in HL patients.97,99  LAG3 seems to be nonredundant from PD-1, as both are expressed on distinct populations of CD8+ T cells.100  Recently, it was shown that in mice PD-1 and LAG3 act synergistically in the onset of autoimmune diseases and tumor escape.101,102  Furthermore in HL, both TREG and LAG3+ CD4+ T cells were shown to be involved in tumor immune evasion because the expression of FoxP3 and LAG3 coincided with the impairment of tumor-specific T-cell responses.99  Therefore, LAG3 is an interesting candidate to combine with therapies that use TREG depletion or PD-1 blockade.

Recently, another immunoregulatory molecule with similarities to PD-1, as well as to PD-L1, was simultaneously discovered by 2 groups: PD-1H (PD-1Homolog)103  or VISTA (V-domain Ig suppressor of T-cell activation).104  This molecule is broadly expressed on hematopoietic cells and is up-regulated on APCs and T cells on activation. Mice treated with a single dose of PD-1H/VISTA antibody did not develop GVHD after alloSCT; however, the mechanism of action was not elucidated.103  Another study identified PD-1H/VISTA as an inhibitory ligand on APCs and tumor cells.104  Here, PD-1H/VISTA Ig-fusion protein conveyed a lasting negative signal to T cells, and expression of the protein on APCs suppressed T-cell proliferation. Importantly, PD-1H/VISTA expression on tumor cells resulted in diminished antitumor immunity. The human ortholog was determined on the genomic level; and because of the important role of this immunoregulatory molecule in GVHD and tumor escape, PD-1H/VISTA is anticipated to be a potential therapeutic target.

Several therapeutic strategies to interfere with the function of coinhibitory molecules are being explored to enhance antitumor T-cell immunity. The challenge of interference with immune checkpoints is to boost antitumor reactivity while avoiding systemic toxicity. This could potentially be achieved by (1) combining the alleviation of coinhibition with other therapeutic options, (2) blocking coinhibitory molecules that are intrinsically skewed toward antitumor responses rather than GVHD or autoimmune effects, and (3) optimal dosage and timing of antibody administration. Appealing combinations are the simultaneous targeting of multiple coinhibitory receptors or incorporation in existing cellular therapies. For example, DC vaccination may be applied together with blocking antibodies or siRNA knockdown of coinhibitory molecules to boost antitumor immunity or by administration of agonistic antibodies against coinhibitory molecules implicated in GVHD, adverse effects may be reduced (Figure 1).

Figure 1

Therapeutic strategies for interfering with coinhibitory molecules. First, blocking antibodies can be used to abrogate binding between coinhibitory molecules with the tumor-reactive T cell and tumor cells, thereby enhancing T-cell responses. In addition, DC therapy can be combined with antagonistic antibodies to boost the tumor-specific effect of DC vaccination. Another method to circumvent coinhibitory signaling during DC vaccination is silencing of coinhibitory molecules. Delivery of siRNA can be achieved either by electroporation or via lipid nanoparticles. Finally, coinhibitory molecules, which are differentially more involved in GVHD, can be stimulated by agonistic antibodies. This will attenuate GVHD-specific T-cell responses, thereby preventing attack of GVHD-prone tissues.

Figure 1

Therapeutic strategies for interfering with coinhibitory molecules. First, blocking antibodies can be used to abrogate binding between coinhibitory molecules with the tumor-reactive T cell and tumor cells, thereby enhancing T-cell responses. In addition, DC therapy can be combined with antagonistic antibodies to boost the tumor-specific effect of DC vaccination. Another method to circumvent coinhibitory signaling during DC vaccination is silencing of coinhibitory molecules. Delivery of siRNA can be achieved either by electroporation or via lipid nanoparticles. Finally, coinhibitory molecules, which are differentially more involved in GVHD, can be stimulated by agonistic antibodies. This will attenuate GVHD-specific T-cell responses, thereby preventing attack of GVHD-prone tissues.

Close modal

Although anti–CTLA-4 and anti–PD-1 monotherapy have shown promising results, combination therapy with other treatment modalities, such as immunomodulatory anticancer agents, vaccines, or TREG depletion, is potentially necessary to effectively cure hematologic cancers. At the moment, several clinical trials are underway that target coinhibitory receptors in hematologic cancers. The effectiveness of CTLA-4 blockade by ipilimumab is investigated in lymphoma patients (NCT00047164), and the anti–PD-1 monoclonal antibody CT-011 is combined with 3 different therapies. In lymphoma patients, CT-011 is administered after autologous SCT (NCT00532259) and combined with rituximab (NCT00904722). Furthermore, the combination of CT-011 with a DC/AML fusion vaccine is being investigated as a therapy for AML patients (NCT01096602). Recently, we explored another treatment option in which an antigen-specific stimulation is combined with interference of coinhibition. We demonstrated that stimulation with PD-L1/L2 silenced MiHA-loaded DC boosted the expansion of MiHA-specific T cells ex vivo.105  After these promising results, we will start a clinical trial shortly combining DLI with vaccination of PD-L1/L2 silenced donor DCs loaded with hematopoietic-restricted MiHA (CCMO-trial no. NL37318). We think that these clinical studies provide a platform for incorporating blockade of coinhibitory molecules in adjuvant therapy of hematologic malignancies, with numerous options for combination therapies. Importantly, the risk of breaking tolerance systemically by blockade of one coinhibitory molecule could be prevented by using lower levels of multiple blocking antibodies targeting different inhibitory molecules simultaneously because together these may boost immune responses in a nonredundant manner. This is stressed by the fact that exhausted T cells are known to display multiple different coinhibitory receptors.106  By analyzing the downstream pathways of different coinhibitory receptors, one could limit these options and exclude combinations of receptors that have redundant effects, and focus on synergistic combinations. Notably, a clinical trial in solid tumors has started which combines blocking antibodies against PD-1 en CTLA-4 (NCT01024231), harnessing the power of these 2 nonredundant immune checkpoints.

The crux in alloSCT is the separation of GVT and GVHD reactivity. Although CTLA-4 and PD-1 clearly contribute to T-cell exhaustion, their activation might have too broad consequences early after alloSCT, and interfering with their signaling might deteriorate GVHD. Interestingly, anti-BTLA was recently reported to impair GVHD while allowing GVT reactions.78,107  Whether or not this important distinction in alloreactive responses also exists in humans needs to be evaluated, but it renders BTLA an important candidate for posttransplantation immunotherapy. Furthermore, the timing of coreceptor blockade seems to be essential to boost GVT without causing GVHD.28,64  Early after alloSCT, the patient is in a highly activated immunologic state because of chemotherapy-induced tissue damage and subsequent inflammation, especially in GVHD target tissues, such as skin and gut. To release immune checkpoints at this time would be dangerous because T cells would home to these inflamed alloantigen-expressing GVHD sites and destroy healthy cells. However, a delayed treatment window after alloSCT is possible, when there is no systemic “inflammatory” state. However, at the tumor site where the inflammation is sustained, antitumor MiHA-specific T cells may be specifically boosted, resulting in renewed GVT effects.

Altogether, coinhibitory molecules play a pivotal role in natural and immunotherapy-induced T cell–mediated immunity against hematologic cancers. With increasing knowledge of a growing number of coinhibitory molecules, novel mono- and combinatorial treatment options become available. In the end, this could lead to optimized immunotherapy against hematologic cancers, with limited risk of adverse events, such as the induction of autoimmune diseases and GVHD.

This work was supported by the Dutch Cancer Society (grants KWF 2008-4018 and KWF 2012-5041).

Contribution: W.J.N., W.H., R.v.d.V., and H.D. wrote the manuscript.

Conflict-of-interest disclosure: The authors declare no competing financial interests.

Correspondence: Harry Dolstra, Laboratory of Hematology, Department of Laboratory Medicine, Radboud University Nijmegen Medical Centre, Geert Grooteplein 8, PO Box 9101, 6500 HB Nijmegen, The Netherlands; e-mail: h.dolstra@labgk.umcn.nl.

1
Croci
 
DO
Zacarias Fluck
 
MF
Rico
 
MJ
, et al. 
Dynamic cross-talk between tumor and immune cells in orchestrating the immunosuppressive network at the tumor microenvironment.
Cancer Immunol Immunother
2007
, vol. 
56
 
11
(pg. 
1687
-
1700
)
2
Chen
 
L
Coinhibitory molecules of the B7-CD28 family in the control of T-cell immunity.
Nat Rev Immunol
2004
, vol. 
4
 
5
(pg. 
336
-
347
)
3
Jenq
 
RR
van den Brink
 
MR
Allogeneic haematopoietic stem cell transplantation: individualized stem cell and immune therapy of cancer.
Nat Rev Cancer
2010
, vol. 
10
 
3
(pg. 
213
-
221
)
4
Feng
 
X
Hui
 
KM
Younes
 
HM
Brickner
 
AG
Targeting minor histocompatibility antigens in graft versus tumor or graft versus leukemia responses.
Trends Immunol
2008
, vol. 
29
 
12
(pg. 
624
-
632
)
5
Brunet
 
JF
Denizot
 
F
Luciani
 
MF
, et al. 
A new member of the immunoglobulin superfamily: CTLA-4.
Nature
1987
, vol. 
328
 
6127
(pg. 
267
-
270
)
6
Tivol
 
EA
Borriello
 
F
Schweitzer
 
AN
, et al. 
Loss of CTLA-4 leads to massive lymphoproliferation and fatal multiorgan tissue destruction, revealing a critical negative regulatory role of CTLA-4.
Immunity
1995
, vol. 
3
 
5
(pg. 
541
-
547
)
7
Peach
 
RJ
Bajorath
 
J
Brady
 
W
, et al. 
Complementarity determining region 1 (CDR1)- and CDR3-analogous regions in CTLA-4 and CD28 determine the binding to B7-1.
J Exp Med
1994
, vol. 
180
 
6
(pg. 
2049
-
2058
)
8
Takahashi
 
T
Tagami
 
T
Yamazaki
 
S
, et al. 
Immunologic self-tolerance maintained by CD25(+)CD4(+) regulatory T cells constitutively expressing cytotoxic T lymphocyte-associated antigen 4.
J Exp Med
2000
, vol. 
192
 
2
(pg. 
303
-
310
)
9
Wing
 
K
Onishi
 
Y
Prieto-Martin
 
P
, et al. 
CTLA-4 control over Foxp3+ regulatory T-cell function.
Science
2008
, vol. 
322
 
5899
(pg. 
271
-
275
)
10
Qureshi
 
OS
Zheng
 
Y
Nakamura
 
K
, et al. 
Trans-endocytosis of CD80 and CD86: a molecular basis for the cell-extrinsic function of CTLA-4.
Science
2011
, vol. 
332
 
6029
(pg. 
600
-
603
)
11
Kaufmann
 
DE
Kavanagh
 
DG
Pereyra
 
F
, et al. 
Upregulation of CTLA-4 by HIV-specific CD4+ T cells correlates with disease progression and defines a reversible immune dysfunction.
Nat Immunol
2007
, vol. 
8
 
11
(pg. 
1246
-
1254
)
12
Baitsch
 
L
Baumgaertner
 
P
Devevre
 
E
, et al. 
Exhaustion of tumor-specific CD8 T cells in metastases from melanoma patients.
J Clin Invest
2011
, vol. 
121
 
6
(pg. 
2350
-
2360
)
13
Leach
 
DR
Krummel
 
MF
Allison
 
JP
Enhancement of antitumor immunity by CTLA-4 blockade.
Science
1996
, vol. 
271
 
5256
(pg. 
1734
-
1736
)
14
Callahan
 
MK
Wolchok
 
JD
Allison
 
JP
Anti–CTLA-4 antibody therapy: immune monitoring during clinical development of a novel immunotherapy.
Semin Oncol
2010
, vol. 
37
 
5
(pg. 
473
-
484
)
15
Lipson
 
EJ
Drake
 
CG
Ipilimumab: an anti–CTLA-4 antibody for metastatic melanoma.
Clin Cancer Res
2011
, vol. 
17
 
22
(pg. 
6958
-
6962
)
16
Fong
 
L
Small
 
EJ
Anti-cytotoxic T-lymphocyte antigen-4 antibody: the first in an emerging class of immunomodulatory antibodies for cancer treatment.
J Clin Oncol
2008
, vol. 
26
 
32
(pg. 
5275
-
5283
)
17
Marshall
 
NA
Christie
 
LE
Munro
 
LR
, et al. 
Immunosuppressive regulatory T cells are abundant in the reactive lymphocytes of Hodgkin lymphoma.
Blood
2004
, vol. 
103
 
5
(pg. 
1755
-
1762
)
18
Brown
 
RD
Pope
 
B
Yuen
 
E
Gibson
 
J
Joshua
 
DE
The expression of T cell related costimulatory molecules in multiple myeloma.
Leuk Lymphoma
1998
, vol. 
31
 
3
(pg. 
379
-
384
)
19
Frydecka
 
I
Kosmaczewska
 
A
Bocko
 
D
, et al. 
Alterations of the expression of T-cell-related costimulatory CD28 and downregulatory CD152 (CTLA-4) molecules in patients with B-cell chronic lymphocytic leukaemia.
Br J Cancer
2004
, vol. 
90
 
10
(pg. 
2042
-
2048
)
20
Norde
 
WJ
Maas
 
F
Hobo
 
W
, et al. 
PD-1/PD-L1 interactions contribute to functional T-cell impairment in patients who relapse with cancer after allogeneic stem cell transplantation.
Cancer Res
2011
, vol. 
71
 
15
(pg. 
5111
-
5122
)
21
Costello
 
RT
Mallet
 
F
Sainty
 
D
, et al. 
Regulation of CD80/B7-1 and CD86/B7-2 molecule expression in human primary acute myeloid leukemia and their role in allogenic immune recognition.
Eur J Immunol
1998
, vol. 
28
 
1
(pg. 
90
-
103
)
22
LaBelle
 
JL
Hanke
 
CA
Blazar
 
BR
Truitt
 
RL
Negative effect of CTLA-4 on induction of T-cell immunity in vivo to B7-1+, but not B7-2+, murine myelogenous leukemia.
Blood
2002
, vol. 
99
 
6
(pg. 
2146
-
2153
)
23
Met
 
O
Wang
 
M
Pedersen
 
AE
, et al. 
The effect of a therapeutic dendritic cell-based cancer vaccination depends on the blockage of CTLA-4 signaling.
Cancer Lett
2006
, vol. 
231
 
2
(pg. 
247
-
256
)
24
O'Mahony
 
D
Morris
 
JC
Quinn
 
C
, et al. 
A pilot study of CTLA-4 blockade after cancer vaccine failure in patients with advanced malignancy.
Clin Cancer Res
2007
, vol. 
13
 
3
(pg. 
958
-
964
)
25
Ansell
 
SM
Hurvitz
 
SA
Koenig
 
PA
, et al. 
Phase I study of ipilimumab, an anti-CTLA-4 monoclonal antibody, in patients with relapsed and refractory B-cell non-Hodgkin lymphoma.
Clin Cancer Res
2009
, vol. 
15
 
20
(pg. 
6446
-
6453
)
26
Pérez-Garcia
 
A
Brunet
 
S
Berlanga
 
JJ
, et al. 
CTLA-4 genotype and relapse incidence in patients with acute myeloid leukemia in first complete remission after induction chemotherapy.
Leukemia
2009
, vol. 
23
 
3
(pg. 
486
-
491
)
27
Daroszewski
 
J
Pawlak
 
E
Karabon
 
L
, et al. 
Soluble CTLA-4 receptor an immunological marker of Graves' disease and severity of ophthalmopathy is associated with CTLA-4 Jo31 and CT60 gene polymorphisms.
Eur J Endocrinol
2009
, vol. 
161
 
5
(pg. 
787
-
793
)
28
Blazar
 
BR
Taylor
 
PA
Panoskaltsis-Mortari
 
A
Sharpe
 
AH
Vallera
 
DA
Opposing roles of CD28:B7 and CTLA-4:B7 pathways in regulating in vivo alloresponses in murine recipients of MHC disparate T cells.
J Immunol
1999
, vol. 
162
 
11
(pg. 
6368
-
6377
)
29
Bashey
 
A
Medina
 
B
Corringham
 
S
, et al. 
CTLA4 blockade with ipilimumab to treat relapse of malignancy after allogeneic hematopoietic cell transplantation.
Blood
2009
, vol. 
113
 
7
(pg. 
1581
-
1588
)
30
Freeman
 
GJ
Long
 
AJ
Iwai
 
Y
, et al. 
Engagement of the PD-1 immunoinhibitory receptor by a novel B7 family member leads to negative regulation of lymphocyte activation.
J Exp Med
2000
, vol. 
192
 
7
(pg. 
1027
-
1034
)
31
Ishida
 
Y
Agata
 
Y
Shibahara
 
K
Honjo
 
T
Induced expression of PD-1, a novel member of the immunoglobulin gene superfamily, upon programmed cell death.
EMBO J
1992
, vol. 
11
 
11
(pg. 
3887
-
3895
)
32
Okazaki
 
T
Honjo
 
T
The PD-1-PD-L pathway in immunological tolerance.
Trends Immunol
2006
, vol. 
27
 
4
(pg. 
195
-
201
)
33
Freeman
 
GJ
Structures of PD-1 with its ligands: sideways and dancing cheek to cheek.
Proc Natl Acad Sci U S A
2008
, vol. 
105
 
30
(pg. 
10275
-
10276
)
34
Youngnak
 
P
Kozono
 
Y
Kozono
 
H
, et al. 
Differential binding properties of B7-H1 and B7-DC to programmed death-1.
Biochem Biophys Res Commun
2003
, vol. 
307
 
3
(pg. 
672
-
677
)
35
Ghiotto
 
M
Gauthier
 
L
Serriari
 
N
, et al. 
PD-L1 and PD-L2 differ in their molecular mechanisms of interaction with PD-1.
Int Immunol
2010
, vol. 
22
 
8
(pg. 
651
-
660
)
36
Keir
 
ME
Butte
 
MJ
Freeman
 
GJ
Sharpe
 
AH
PD-1 and its ligands in tolerance and immunity.
Annu Rev Immunol
2008
, vol. 
26
 (pg. 
677
-
704
)
37
Gao
 
Q
Wang
 
XY
Qiu
 
SJ
, et al. 
Overexpression of PD-L1 significantly associates with tumor aggressiveness and postoperative recurrence in human hepatocellular carcinoma.
Clin Cancer Res
2009
, vol. 
15
 
3
(pg. 
971
-
979
)
38
Zhang
 
C
Wu
 
S
Xue
 
X
, et al. 
Anti–tumor immunotherapy by blockade of the PD-1/PD-L1 pathway with recombinant human PD-1-IgV.
Cytotherapy
2008
, vol. 
10
 
7
(pg. 
711
-
719
)
39
Hino
 
R
Kabashima
 
K
Kato
 
Y
, et al. 
Tumor cell expression of programmed cell death-1 ligand 1 is a prognostic factor for malignant melanoma.
Cancer
2010
, vol. 
116
 
7
(pg. 
1757
-
1766
)
40
Park
 
JJ
Omiya
 
R
Matsumura
 
Y
, et al. 
B7-H1/CD80 interaction is required for the induction and maintenance of peripheral T-cell tolerance.
Blood
2010
, vol. 
116
 
8
(pg. 
1291
-
1298
)
41
Ghebeh
 
H
Lehe
 
C
Barhoush
 
E
, et al. 
Doxorubicin downregulates cell surface B7-H1 expression and upregulates its nuclear expression in breast cancer cells: role of B7-H1 as an anti-apoptotic molecule.
Breast Cancer Res
2010
, vol. 
12
 
4
pg. 
R48
 
42
Messal
 
N
Serriari
 
NE
Pastor
 
S
Nunes
 
JA
Olive
 
D
PD-L2 is expressed on activated human T cells and regulates their function.
Mol Immunol
2011
, vol. 
48
 
15
(pg. 
2214
-
2219
)
43
Velu
 
V
Kannanganat
 
S
Ibegbu
 
C
, et al. 
Elevated expression levels of inhibitory receptor programmed death 1 on simian immunodeficiency virus-specific CD8 T cells during chronic infection but not after vaccination.
J Virol
2007
, vol. 
81
 
11
(pg. 
5819
-
5828
)
44
Velu
 
V
Titanji
 
K
Zhu
 
B
, et al. 
Enhancing SIV-specific immunity in vivo by PD-1 blockade.
Nature
2009
, vol. 
458
 
7235
(pg. 
206
-
210
)
45
Kaufmann
 
DE
Walker
 
BD
PD-1 and CTLA-4 inhibitory cosignaling pathways in HIV infection and the potential for therapeutic intervention.
J Immunol
2009
, vol. 
182
 
10
(pg. 
5891
-
5897
)
46
Wherry
 
EJ
Ha
 
SJ
Kaech
 
SM
, et al. 
Molecular signature of CD8+ T-cell exhaustion during chronic viral infection.
Immunity
2007
, vol. 
27
 
4
(pg. 
670
-
684
)
47
Quigley
 
M
Pereyra
 
F
Nilsson
 
B
, et al. 
Transcriptional analysis of HIV-specific CD8+ T cells shows that PD-1 inhibits T-cell function by upregulating BATF.
Nat Med
2010
, vol. 
16
 
10
(pg. 
1147
-
1151
)
48
Youngblood
 
B
Oestreich
 
KJ
Ha
 
SJ
, et al. 
Chronic virus infection enforces demethylation of the locus that encodes PD-1 in antigen-specific CD8(+) T cells.
Immunity
2011
, vol. 
35
 
3
(pg. 
400
-
412
)
49
Iwai
 
Y
Ishida
 
M
Tanaka
 
Y
, et al. 
Involvement of PD-L1 on tumor cells in the escape from host immune system and tumor immunotherapy by PD-L1 blockade.
Proc Natl Acad Sci U S A
2002
, vol. 
99
 
19
(pg. 
12293
-
12297
)
50
Zhang
 
L
Gajewski
 
TF
Kline
 
J
PD-1/PD-L1 interactions inhibit antitumor immune responses in a murine acute myeloid leukemia model.
Blood
2009
, vol. 
114
 
8
(pg. 
1545
-
1552
)
51
Hallett
 
WH
Jing
 
W
Drobyski
 
WR
Johnson
 
BD
Immunosuppressive effects of multiple myeloma are overcome by PD-L1 blockade.
Biol Blood Marrow Transplant
2011
, vol. 
17
 
8
(pg. 
1133
-
1145
)
52
Andorsky
 
DJ
Yamada
 
RE
Said
 
J
, et al. 
Programmed death ligand 1 is expressed by non-Hodgkin lymphomas and inhibits the activity of tumor-associated T cells.
Clin Cancer Res
2011
, vol. 
17
 
13
(pg. 
4232
-
4244
)
53
Yamamoto
 
R
Nishikori
 
M
Kitawaki
 
T
, et al. 
PD-1-PD-1 ligand interaction contributes to immunosuppressive microenvironment of Hodgkin lymphoma.
Blood
2008
, vol. 
111
 
6
(pg. 
3220
-
3224
)
54
Saudemont
 
A
Quesnel
 
B
In a model of tumor dormancy, long-term persistent leukemic cells have increased B7-H1 and B7.1 expression and resist CTL-mediated lysis.
Blood
2004
, vol. 
104
 
7
(pg. 
2124
-
2133
)
55
Ni
 
L
Ma
 
CJ
Zhang
 
Y
, et al. 
PD-1 modulates regulatory T cells and suppresses T-cell responses in HCV-associated lymphoma.
Immunol Cell Biol
2011
, vol. 
89
 
4
(pg. 
535
-
539
)
56
Zhou
 
Q
Munger
 
ME
Highfill
 
SL
, et al. 
Program death-1 signaling and regulatory T cells collaborate to resist the function of adoptively transferred cytotoxic T lymphocytes in advanced acute myeloid leukemia.
Blood
2010
, vol. 
116
 
14
(pg. 
2484
-
2493
)
57
Berger
 
R
Rotem-Yehudar
 
R
Slama
 
G
, et al. 
Phase I safety and pharmacokinetic study of CT-011, a humanized antibody interacting with PD-1, in patients with advanced hematologic malignancies.
Clin Cancer Res
2008
, vol. 
14
 
10
(pg. 
3044
-
3051
)
58
Benson
 
DM
Bakan
 
CE
Mishra
 
A
, et al. 
The PD-1/PD-L1 axis modulates the natural killer cell versus multiple myeloma effect: a therapeutic target for CT-011, a novel monoclonal anti–PD-1 antibody.
Blood
2010
, vol. 
116
 
13
(pg. 
2286
-
2294
)
59
Rosenblatt
 
J
Glotzbecker
 
B
Mills
 
H
, et al. 
PD-1 blockade by CT-011, anti-PD-1 antibody, enhances ex vivo T-cell responses to autologous dendritic cell/myeloma fusion vaccine.
J Immunother
2011
, vol. 
34
 
5
(pg. 
409
-
418
)
60
Brahmer
 
JR
Drake
 
CG
Wollner
 
I
, et al. 
Phase I study of single-agent anti-programmed death-1 (MDX-1106) in refractory solid tumors: safety, clinical activity, pharmacodynamics, and immunologic correlates.
J Clin Oncol
2010
, vol. 
28
 
19
(pg. 
3167
-
3175
)
61
Sznol
 
M
Powderly
 
JD
Smith
 
DC
, et al. 
Safety and antitumor activity of biweekly MDX-1106 (anti-PD-1, BMS-936558/ONO-4538) in patients with advanced refractory malignancies [abstract].
J Clin Oncol
2010
, vol. 
28
 pg. 
15S
  
Abstract 2506
62
McDermott
 
DF
Drake
 
CG
Sznol
 
M
, et al. 
A phase I study to evaluate safety and antitumor activity of biweekly BMS-936558 (anti-PD-1, MDX-1106/ONO-4538) in patients with RCC and other advanced refractory malignancies [abstract].
J Clin Oncol
2011
, vol. 
29
 pg. 
S7
  
Abstract 331
63
Asakura
 
S
Hashimoto
 
D
Takashima
 
S
, et al. 
Alloantigen expression on non-hematopoietic cells reduces graft-versus-leukemia effects in mice.
J Clin Invest
2010
, vol. 
120
 
7
(pg. 
2370
-
2378
)
64
Flutter
 
B
Edwards
 
N
Fallah-Arani
 
F
, et al. 
Nonhematopoietic antigen blocks memory programming of alloreactive CD8+ T cells and drives their eventual exhaustion in mouse models of bone marrow transplantation.
J Clin Invest
2010
, vol. 
120
 
11
(pg. 
3855
-
3868
)
65
Mumprecht
 
S
Schurch
 
C
Schwaller
 
J
Solenthaler
 
M
Ochsenbein
 
AF
Programmed death 1 signaling on chronic myeloid leukemia-specific T cells results in T-cell exhaustion and disease progression.
Blood
2009
, vol. 
114
 
8
(pg. 
1528
-
1536
)
66
Watanabe
 
N
Gavrieli
 
M
Sedy
 
JR
, et al. 
BTLA is a lymphocyte inhibitory receptor with similarities to CTLA-4 and PD-1.
Nat Immunol
2003
, vol. 
4
 
7
(pg. 
670
-
679
)
67
Hobo
 
W
Norde
 
WJ
Schaap
 
N
, et al. 
B and T lymphocyte attenuator mediates inhibition of tumor-reactive CD8+ T cells in patients after allogeneic stem cell transplantation.
J Immunol
2012
, vol. 
189
 
1
(pg. 
39
-
49
)
68
Murphy
 
TL
Murphy
 
KM
Slow down and survive: enigmatic immunoregulation by BTLA and HVEM.
Annu Rev Immunol
2010
, vol. 
28
 (pg. 
389
-
411
)
69
Sedy
 
JR
Gavrieli
 
M
Potter
 
KG
, et al. 
B and T lymphocyte attenuator regulates T-cell activation through interaction with herpesvirus entry mediator.
Nat Immunol
2005
, vol. 
6
 
1
(pg. 
90
-
98
)
70
del Rio
 
ML
Lucas
 
CL
Buhler
 
L
Rayat
 
G
Rodriguez-Barbosa
 
JI
HVEM/LIGHT/BTLA/CD160 cosignaling pathways as targets for immune regulation.
J Leukoc Biol
2010
, vol. 
87
 
2
(pg. 
223
-
235
)
71
Cai
 
G
Anumanthan
 
A
Brown
 
JA
, et al. 
CD160 inhibits activation of human CD4+ T cells through interaction with herpesvirus entry mediator.
Nat Immunol
2008
, vol. 
9
 
2
(pg. 
176
-
185
)
72
Tamada
 
K
Shimozaki
 
K
Chapoval
 
AI
, et al. 
LIGHT, a TNF-like molecule, costimulates T-cell proliferation and is required for dendritic cell-mediated allogeneic T-cell response.
J Immunol
2000
, vol. 
164
 
8
(pg. 
4105
-
4110
)
73
Cheung
 
TC
Oborne
 
LM
Steinberg
 
MW
, et al. 
T cell intrinsic heterodimeric complexes between HVEM and BTLA determine receptivity to the surrounding microenvironment.
J Immunol
2009
, vol. 
183
 
11
(pg. 
7286
-
7296
)
74
Oya
 
Y
Watanabe
 
N
Owada
 
T
, et al. 
Development of autoimmune hepatitis-like disease and production of autoantibodies to nuclear antigens in mice lacking B and T lymphocyte attenuator.
Arthritis Rheum
2008
, vol. 
58
 
8
(pg. 
2498
-
2510
)
75
Derré
 
L
Rivals
 
JP
Jandus
 
C
, et al. 
BTLA mediates inhibition of human tumor-specific CD8+ T cells that can be partially reversed by vaccination.
J Clin Invest
2010
, vol. 
120
 
1
(pg. 
157
-
167
)
76
Serriari
 
NE
Gondois-Rey
 
F
Guillaume
 
Y
, et al. 
B and T lymphocyte attenuator is highly expressed on CMV-specific T cells during infection and regulates their function.
J Immunol
2010
, vol. 
185
 
6
(pg. 
3140
-
3148
)
77
Fourcade
 
J
Sun
 
Z
Pagliano
 
O
, et al. 
CD8(+) T cells specific for tumor antigens can be rendered dysfunctional by the tumor microenvironment through upregulation of the inhibitory receptors BTLA and PD-1.
Cancer Res
2012
, vol. 
72
 
4
(pg. 
887
-
896
)
78
Albring
 
JC
Sandau
 
MM
Rapaport
 
AS
, et al. 
Targeting of B and T lymphocyte associated (BTLA) prevents graft-versus-host disease without global immunosuppression.
J Exp Med
2010
, vol. 
207
 
12
(pg. 
2551
-
2559
)
79
Sakoda
 
Y
Park
 
JJ
Zhao
 
Y
, et al. 
Dichotomous regulation of GVHD through bidirectional functions of the BTLA-HVEM pathway.
Blood
2011
, vol. 
117
 
8
(pg. 
2506
-
2514
)
80
Del Rio
 
ML
Jones
 
ND
Buhler
 
L
, et al. 
Selective blockade of herpesvirus entry mediator-B and T lymphocyte attenuator pathway ameliorates acute graft-versus-host reaction.
J Immunol
2012
, vol. 
188
 
10
(pg. 
4885
-
4896
)
81
Rijkers
 
ES
De Ruiter
 
T
Baridi
 
A
, et al. 
The inhibitory CD200R is differentially expressed on human and mouse T and B lymphocytes.
Mol Immunol
2008
, vol. 
45
 
4
(pg. 
1126
-
1135
)
82
Olteanu
 
H
Harrington
 
AM
Hari
 
P
Kroft
 
SH
CD200 expression in plasma cell myeloma.
Br J Haematol
2011
, vol. 
153
 
3
(pg. 
408
-
411
)
83
Stumpfova
 
M
Ratner
 
D
Desciak
 
EB
Eliezri
 
YD
Owens
 
DM
The immunosuppressive surface ligand CD200 augments the metastatic capacity of squamous cell carcinoma.
Cancer Res
2010
, vol. 
70
 
7
(pg. 
2962
-
2972
)
84
Coles
 
SJ
Wang
 
EC
Man
 
S
, et al. 
CD200 expression suppresses natural killer cell function and directly inhibits patient anti-tumor response in acute myeloid leukemia.
Leukemia
2011
, vol. 
25
 
5
(pg. 
792
-
799
)
85
Pallasch
 
CP
Ulbrich
 
S
Brinker
 
R
, et al. 
Disruption of T cell suppression in chronic lymphocytic leukemia by CD200 blockade.
Leuk Res
2009
, vol. 
33
 
3
(pg. 
460
-
464
)
86
Gorczynski
 
RM
Chen
 
Z
Hu
 
J
Kai
 
Y
Lei
 
J
Evidence of a role for CD200 in regulation of immune rejection of leukaemic tumour cells in C57BL/6 mice.
Clin Exp Immunol
2001
, vol. 
126
 
2
(pg. 
220
-
229
)
87
Kretz-Rommel
 
A
Qin
 
F
Dakappagari
 
N
, et al. 
CD200 expression on tumor cells suppresses antitumor immunity: new approaches to cancer immunotherapy.
J Immunol
2007
, vol. 
178
 
9
(pg. 
5595
-
5605
)
88
Kretz-Rommel
 
A
Qin
 
F
Dakappagari
 
N
, et al. 
Blockade of CD200 in the presence or absence of antibody effector function: implications for anti-CD200 therapy.
J Immunol
2008
, vol. 
180
 
2
(pg. 
699
-
705
)
89
Sakuishi
 
K
Jayaraman
 
P
Behar
 
SM
Anderson
 
AC
Kuchroo
 
VK
Emerging Tim-3 functions in antimicrobial and tumor immunity.
Trends Immunol
2011
, vol. 
32
 
8
(pg. 
345
-
349
)
90
Jones
 
RB
Ndhlovu
 
LC
Barbour
 
JD
, et al. 
Tim-3 expression defines a novel population of dysfunctional T cells with highly elevated frequencies in progressive HIV-1 infection.
J Exp Med
2008
, vol. 
205
 
12
(pg. 
2763
-
2779
)
91
Fourcade
 
J
Sun
 
Z
Benallaoua
 
M
, et al. 
Upregulation of Tim-3 and PD-1 expression is associated with tumor antigen-specific CD8+ T cell dysfunction in melanoma patients.
J Exp Med
2010
, vol. 
207
 
10
(pg. 
2175
-
2186
)
92
Lee
 
MJ
Woo
 
MY
Heo
 
YM
, et al. 
The inhibition of the T-cell immunoglobulin and mucin domain 3 (Tim3) pathway enhances the efficacy of tumor vaccine.
Biochem Biophys Res Commun
2010
, vol. 
402
 
1
(pg. 
88
-
93
)
93
Zhou
 
Q
Munger
 
ME
Veenstra
 
RG
, et al. 
Coexpression of Tim-3 and PD-1 identifies a CD8+ T-cell exhaustion phenotype in mice with disseminated acute myelogenous leukemia.
Blood
2011
, vol. 
117
 
17
(pg. 
4501
-
4510
)
94
Huang
 
X
Bai
 
X
Cao
 
Y
, et al. 
Lymphoma endothelium preferentially expresses Tim-3 and facilitates the progression of lymphoma by mediating immune evasion.
J Exp Med
2010
, vol. 
207
 
3
(pg. 
505
-
520
)
95
Nagahara
 
K
Arikawa
 
T
Oomizu
 
S
, et al. 
Galectin-9 increases Tim-3+ dendritic cells and CD8+ T cells and enhances antitumor immunity via galectin-9-Tim-3 interactions.
J Immunol
2008
, vol. 
181
 
11
(pg. 
7660
-
7669
)
96
Lee
 
J
Su
 
EW
Zhu
 
C
, et al. 
Phosphotyrosine-dependent coupling of Tim-3 to T-cell receptor signaling pathways.
Mol Cell Biol
2011
, vol. 
31
 
19
(pg. 
3963
-
3974
)
97
Baixeras
 
E
Huard
 
B
Miossec
 
C
, et al. 
Characterization of the lymphocyte activation gene 3-encoded protein. A new ligand for human leukocyte antigen class II antigens.
J Exp Med
1992
, vol. 
176
 
2
(pg. 
327
-
337
)
98
Triebel
 
F
Jitsukawa
 
S
Baixeras
 
E
, et al. 
LAG-3, a novel lymphocyte activation gene closely related to CD4.
J Exp Med
1990
, vol. 
171
 
5
(pg. 
1393
-
1405
)
99
Gandhi
 
MK
Lambley
 
E
Duraiswamy
 
J
, et al. 
Expression of LAG-3 by tumor-infiltrating lymphocytes is coincident with the suppression of latent membrane antigen-specific CD8+ T-cell function in Hodgkin lymphoma patients.
Blood
2006
, vol. 
108
 
7
(pg. 
2280
-
2289
)
100
Grosso
 
JF
Goldberg
 
MV
Getnet
 
D
, et al. 
Functionally distinct LAG-3 and PD-1 subsets on activated and chronically stimulated CD8 T cells.
J Immunol
2009
, vol. 
182
 
11
(pg. 
6659
-
6669
)
101
Okazaki
 
T
Okazaki
 
IM
Wang
 
J
, et al. 
PD-1 and LAG-3 inhibitory co-receptors act synergistically to prevent autoimmunity in mice.
J Exp Med
2011
, vol. 
208
 
2
(pg. 
395
-
407
)
102
Woo
 
SR
Turnis
 
ME
Goldberg
 
MV
, et al. 
Immune inhibitory molecules LAG-3 and PD-1 synergistically regulate T-cell function to promote tumoral immune escape.
Cancer Res
2012
, vol. 
72
 
4
(pg. 
917
-
927
)
103
Flies
 
DB
Wang
 
S
Xu
 
H
Chen
 
L
Cutting edge: a monoclonal antibody specific for the programmed death-1 homolog prevents graft-versus-host disease in mouse models.
J Immunol
2011
, vol. 
187
 
4
(pg. 
1537
-
1541
)
104
Wang
 
L
Rubinstein
 
R
Lines
 
JL
, et al. 
VISTA, a novel mouse Ig superfamily ligand that negatively regulates T-cell responses.
J Exp Med
2011
, vol. 
208
 
3
(pg. 
577
-
592
)
105
Hobo
 
W
Maas
 
F
Adisty
 
N
, et al. 
siRNA silencing of PD-L1 and PD-L2 on dendritic cells augments expansion and function of minor histocompatibility antigen-specific CD8+ T cells.
Blood
2010
, vol. 
116
 
22
(pg. 
4501
-
4511
)
106
Blackburn
 
SD
Shin
 
H
Haining
 
WN
, et al. 
Coregulation of CD8+ T-cell exhaustion by multiple inhibitory receptors during chronic viral infection.
Nat Immunol
2009
, vol. 
10
 
1
(pg. 
29
-
37
)
107
del Rio
 
ML
Kurtz
 
J
Perez-Martinez
 
C
, et al. 
B- and T-lymphocyte attenuator targeting protects against the acute phase of graft versus host reaction by inhibiting donor antihost cytotoxicity.
Transplantation
2011
, vol. 
92
 
10
(pg. 
1085
-
1093
)
108
Moreaux
 
J
Hose
 
D
Reme
 
T
, et al. 
CD200 is a new prognostic factor in multiple myeloma.
Blood
2006
, vol. 
108
 
13
(pg. 
4194
-
4197
)
109
Motta
 
M
Rassenti
 
L
Shelvin
 
BJ
, et al. 
Increased expression of CD152 (CTLA-4) by normal T lymphocytes in untreated patients with B-cell chronic lymphocytic leukemia.
Leukemia
2005
, vol. 
19
 
10
(pg. 
1788
-
1793
)

Author notes

*

W.J.N. and W.H. contributed equally to this study.

Sign in via your Institution