Skip to main content

Targeting tumor-necrosis factor receptor pathways for tumor immunotherapy

Abstract

With the success of ipilimumab and promise of programmed death-1 pathway-targeted agents, the field of tumor immunotherapy is expanding rapidly. Newer targets for clinical development include select members of the tumor necrosis factor receptor (TNFR) family. Agonist antibodies to these co-stimulatory molecules target both T and B cells, modulating T-cell activation and enhancing immune responses. In vitro and in vivo preclinical data have provided the basis for continued development of 4-1BB, OX40, glucocorticoid-induced TNFR-related gene, herpes virus entry mediator, and CD27 as potential therapies for patients with cancer. In this review, we summarize the immune response to tumors, consider preclinical and early clinical data on select TNFR family members, discuss potential translational challenges and suggest possible combination therapies with the aim of inducing durable antitumor responses.

Introduction

After nearly a century of skepticism regarding the efficacy of immunotherapy in cancer, a resurgence has begun that is driven primarily by the success of ipilimumab [1]. The immune-mediated mechanism of action and resultant antitumor activity of ipilimumab lend support to the notion that tumors are under immune surveillance. Moreover, the concept of “immuno-editing” suggests that the pressure exerted on tumors by the immune system shapes or “edits” tumor cells, allowing their escape from immune elimination [2]. Briefly, the immune system is capable of eliminating malignant cells during initial transformation. As tumors grow, an “equilibrium” is reached where immune tumor growth is matched by immune-mediated tumor destruction. Eventually, malignant cells either accumulate mutations, making them non-immunogenic, or immunosuppressive pathways become activated, allowing the tumor to escape immune recognition [25]. It now appears that targeting immunomodulatory mechanisms can tip the balance from escape back toward elimination. In addition to ipilimumab and other co-inhibitory checkpoints (i.e., programmed death-1 [PD-1]), research into the stimulation of T cell responses via agonist therapies has opened another therapeutic possibility. This review will focus on the rationale for targeting co-stimulatory pathways in T cells, summarize agents in development, and offer possible treatment strategies using these agents in combination with other immunotherapies.

Review

The host response to tumors has been well described [69]. However, tumors have developed ways to escape this response via a number of mechanisms; these have been extensively reviewed previously [68, 10]. The accumulation of suppressive cells and an inhibitory cytokine milieu in and around the tumor can form an immunosuppressive environment that prevents successful T cell-mediated destruction of malignant cells [11]. The goal of many immunotherapies is to help the immune system overcome the mechanisms that tumors employ to evade destruction.

Targeting immune “checkpoint” pathways

Upon initial activation by antigen-presenting cells (APCs), tumor-specific T cells receive essential co-stimulation through binding of CD28 to CD80/86 on activated APCs. However, the CD28-CD80/86 pathway is antagonized by cytotoxic T lymphocyte antigen-4 (CTLA-4; CD152), one of the first co-inhibitory checkpoints that self-limit responses. CTLA-4 competes with CD28 for CD80/86 binding and has a much greater affinity than CD28 for CD80; CTLA-4 signaling decreases the magnitude of early T-cell activation, expansion, and function [1214]. Ipilimumab blocks CTLA-4 antagonism of CD28 and was the first agent to demonstrate an overall survival benefit for patients with metastatic melanoma [1, 15]. The ability of CTLA-4 blockade to enhance tumor immunity may be because most tumor antigens are modified self-antigens or mutated unique antigens against which T cell cytotoxic function is believed to be relatively poor. Therefore, by releasing this first brake on immune activation, ipilimumab allows the generation of a more effective immune response [16].

Subsequent to CTLA-4, PD-1 was also shown to regulate the immune response. PD-1 interacts with B7-homolog 1 (B7-H1 or programmed death receptor-1 ligand1 [PD-L1]) and B7-DC (PD-L2) on APCs and tumor cells. Ligation of PD-1 by either of its ligands downregulates T-cell receptor (TCR) signaling and abrogates stimulatory cytokine production [17, 18]. Moreover, PD-1 upregulation on effector T cells identifies T cells with an exhausted phenotype that cannot maintain polyfunctional cytokine production [19]. Although the evaluation of PD-1/PD-L1–targeted therapies is at an early stage, they are showing promising results. Evaluation of the anti–PD-1 antibody nivolumab in more than 300 patients in a phase I trial, showed antitumor activity in patients with non-small cell lung cancer (NSCLC), renal cell carcinoma (RCC), and melanoma, and an encouraging safety profile [20, 21]. Combination of nivolumab with ipilimumab has also demonstrated a potentially synergistic effect in a recent phase I combination study for patients with advanced melanoma [22]. Phase III trials in these tumors are ongoing. The anti–PD-1 antibody CT-011 has shown activity in a small phase I trial of patients with various hematological malignancies [23]. Data with other anti–PD-1 and anti–PD-L1 antibodies and fusion proteins are becoming available [2427].

TNFR superfamily costimulatory molecules

In addition to blocking co-inhibitory pathways, activating co-stimulatory pathways to potentiate antitumor immune responses is a promising approach. Members of the tumor necrosis factor receptor superfamily (TNFRsf) include several co-stimulatory proteins with key roles in B and T cell development, survival, immune activation, and antitumor immune responses [28]. These co-stimulatory TNFRsf members lack death domains, enabling them to enhance activation and proinflammatory cascades [29], making them attractive therapeutic targets (Table 1). Recent clinical success with checkpoint inhibitors has provided the rationale for investigating agonism of 4-1BB (CD137), OX40, glucocorticoid-induced TNFR-related gene (GITR), herpes virus entry mediator (HVEM), and CD27 in order to extend clinical benefit to more patients.

Table 1 TNFR-targeted agents in development

4-1BB (CD137)

4-1BB is a molecule with profound effects on T-cell proliferation and CD8+ T-cell function [3032]. It is primarily present on activated but not resting T cells, activated natural killer (NK) cells, and natural killer T (NKT) cells. It is expressed constitutively on certain populations of dendritic cells (DCs) and regulatory T cells (Tregs) [3336]; 4-1BB expression is upregulated on monocytes upon activation [35]. Stimulation of 4-1BB by either its natural ligand, 4-1BBL, or by agonist antibodies enhances the activation of various immune cells, including T cells, DC (upregulation of B7 molecules and immunostimulatory cytokine production), monocytes, and neutrophils (proinflammatory cytokine secretion), and induces a spectrum of effects on B and NK cells [35, 36]. 4-1BB signaling can co-stimulate T cells in a CD28-independent manner [37], protect tumor-infiltrating lymphocytes (TILs) from activation-induced cell death (AICD) via upregulation of antiapoptotic pathways [38, 39], and enhance cytotoxic T lymphocyte (CTL) survival and cytolytic activity [40].

Preclinical data show that targeting 4-1BB with an agonist antibody can promote tumor control in numerous preclinical models, and is often associated with increased CTL effector capability [36, 4143]. The effects of pairing agonist anti–4-1BB antibodies with other therapies have also been investigated. Anti-4-1BB antibody administered with adoptive T-cell therapy resulted in survival and resistance to rechallenge in 80% of mice bearing established thymomas [44]. Combining anti–4-1BB with an agonist anti-CD40 plus a blocking antibody against DR5, a receptor for TRAIL (a strategy termed “trimab”), had activity against multiple transplanted tumors in mice [45]. Adding anti–CTLA-4 further increased the potency of this approach, resulting in ~80% tumor rejection of established mammary 4 T1 tumors in mice [46]. Combining CTLA-4 blockade and 4-1BB co-stimulation with a granulocyte-macrophage colony-stimulating factor (GM-CSF)–secreting melanoma vaccine greatly improved tumor eradication and promoted survival compared with vaccine plus either agent [47]. Combination treatment increased proliferation and tumor infiltration by both CD4+ and CD8+ T cells, and intratumor inflammatory cytokine production. Likewise, a 4-1BBL–expressing RM-1 cell vaccine used with CTLA-4 blockade in mice bearing RM-1 prostate tumors improved survival compared with mice treated with monotherapy [48]. These data suggest that concurrent manipulation of the CTLA-4 and 4-1BB signaling pathways merits clinical evaluation.

Other combinations have also been investigated. Kohrt and colleagues showed human trastuzumab-activated NK cells had improved cytotoxicity against breast cancer cells when stimulated with an agonist 4-1BB antibody [49]. Agonistic anti–4-1BB antibodies also had antitumor activity when given together with radiation therapy in murine breast and lung cancer models and when combined with anti–PD-1 plus radiotherapy [50, 51]. Finally, targeting 4-1BB with intratumoral interferon-α therapy or DC-based vaccination also produced significant antitumor responses and/or improved survival [52, 53].

Against this background, agonist 4-1BB therapy with urelumab (BMS-663513) was investigated in patients with melanoma, RCC, and ovarian cancer. Although some antitumor activity was observed during a phase I trial, safety concerns halted development [54, 55]. Development has recently restarted using urelumab at a lower dose in combination with other agents (NCT01471210 and NCT01775631, clincaltrials.gov).

A different approach to utilizing the immune-potentiating properties of 4-1BB has been explored in the context of chimeric antigen receptor (CAR)–transduced T cells. Expressing an antibody with specificity for the B cell antigen CD19 linked to the intracellular signaling domains of 4-1BB and CD3-ζ enhanced the survival and effector functions provided by transduced T cells directed against tumor cells. T cells with this modified CAR (termed “CART19” cells) infused into a patient with refractory chronic lymphocytic leukemia (CLL) induced complete disease remission that was ongoing for more than 10 months [56]. Clinical studies of CART19 cells in patients with various B cell leukemias and lymphomas are ongoing (clinicaltrials.gov).

OX40 (CD134)

OX40 (TNR4) [57] is similar to 4-1BB and GITR, and engagement promotes T-cell activation, survival, proliferation, and cytokine production [5862]. OX40 is primarily expressed on activated CD4+ T cells 24–48 hours after activation, and on CD8+ T cells, neutrophils, dendritic cells, and Tregs [60, 63]. The natural ligand of OX40, OX40 ligand (OX40L), is most common on APCs, and is also found on activated T cells. OX40 expression is induced by TCR stimulation, but co-stimulation through other molecules, such as CD28, or exposure to certain cytokines can further upregulate expression [64]. While OX40 has a key role in the development and function of Tregs, engagement of OX40 with an agonist antibody can also deactivate the suppressive function of Tregs [63, 6569].

OX40 agonism has been investigated in multiple tumor models. As a monotherapy, it delayed tumor growth in vivo and promoted the rejection of various tumors [60, 62, 70]. OX40-dependent antitumor immunity required both CD4+ and CD8+ T cells, and a significant proportion of treated mice remained tumor-free and resistant to rechallenge, supporting the notion that OX40 engagement promotes memory [71]. Combination approaches aimed at improving the efficacy of OX40 engagement have been explored. Combinations of anti-OX40 with fractionated radiotherapy, interleukin (IL)-12 and anti–4-1BB, anti–CTLA-4 and CpG oligonucleotides, anti-CD25 and anti–CTLA-4 with adoptive cell transfer, transforming growth factor (TGF)-β inhibition, or IL-2 improved antitumor responses, tumor rejection, long-term survival, and/or resistance to tumor rechallenge in mice bearing various cancers [64, 7175]. We have shown that combining anti-OX40 with cyclophosphamide led to the regression of the poorly immunogenic B16 murine melanoma and was associated with fewer intratumoral Tregs, leading to a favorable effector/Treg cell ratio within the tumor microenvironment [76]. Adoptive transfer of melanoma specific CD4+ T cells together with OX40 and cyclophosphamide eliminated even more advanced melanomas. This combination triggered cytolytic activity in the transferred CD4+ T cells, a phenomenon largely responsible for the potency of this combination [76].

In the first-in-human clinical trial, 30 patients with solid tumors received a murine anti-human OX40 monoclonal antibody at doses from 0.1 through 2 mg/kg; the maximum tolerated dose was not identified, and some patients had an objective response after one dose [77]. Humanized OX40 antibodies and a fully human OX40L:IgG fusion protein have been developed [60, 78, 79]. Phase II clinical trials evaluating OX40 agonists with stereotactic radiation and/or cyclophosphamide in patients with multiple tumor types are ongoing (NCT01642290, NCT01303705).

GITR (CD357)

GITR is expressed at low levels on resting CD4+ and CD8+ T cells, is upregulated after 24–72 hours of stimulation, and remains expressed for several days [8082]. In contrast to effector T cells, Tregs constitutively express GITR. GITR has also been observed on DC, monocytes, and NK cells. Similar to OX40 and 4-1BB, ligation provides co-stimulatory signals that enhance T-cell proliferation and effector function, and protect T cells from AICD [81, 82]. GITR ligand (GITRL) is highly expressed on activated APCs and endothelial cells (ECs). Interestingly, while the role of GITRL on APCs appears primarily to provide costimulation, it was recently shown that GITRL/GITR interactions on EC may be important for triggering leukocyte adhesion and transmigration [83]. Although human and mouse GITR have similar characteristics, unlike most TNFR family members, murine GITR exists as a dimer instead of a trimer [84, 85]. How this difference relates to the biological functions of mouse and human GITR is unknown.

Reports showing GITR ligation can break self-tolerance and abrogate T-cell suppression by Tregs identify it as a target for cancer immunotherapy [86, 87]. Our laboratory was the first to demonstrate that an agonist anti-GITR antibody, the rat monoclonal DTA-1, could protect mice from B16 tumor rechallenge and that treatment induced rejection of small, established B16 tumors [88, 89]. Stimulation of GITR also cured established sarcomas, induced long-lasting memory, and had activity in other mouse models [82, 90]. GITRL manipulation has also been explored. Mice bearing GITRL-expressing tumors had improved CTL effector function and peritumoral injection of a GITRL vaccine slowed the growth of established tumors [91].

Multiple mechanisms appear to contribute to the antitumor effects of GITR modulation. We have demonstrated that DTA-1 administration impaired intratumoral Treg expression of FoxP3, resulting in a loss of Treg lineage stability and abrogation of intratumor Treg suppressive function [92]. This led to a higher effector T cell (Teff):Treg ratio and improved antitumor CTL function [88]. Additionally, Côté and colleagues found that DTA-1 treatment enhanced the development of high-avidity CTL responses to tumor-associated antigens in a mouse melanoma model [93]. Recent reports show that DTA-1 can cause depletion of intratumoral Tregs through interactions between the antibody and activating FcγRs on myeloid cells [94]. Because of differences in tumor models and underlying treatment conditions, it is unclear if one of these mechanisms is dominant or if all play a role in GITR agonist immunotherapy. The exact contribution of each will require further investigation.

GITR agonism (either via DTA-1 or GITRL fusion protein) has been successfully combined with other immunotherapies, including DNA- or DC-based vaccines [93, 9597]. Response to DTA-1 in mice bearing fibrosarcoma or colorectal carcinoma improved in combination with adoptive T-cell transfer or an antagonistic anti–CTLA-4 antibody [90, 98, 99]. Pruitt and colleagues saw improvements in antitumor immunity when a melanoma DC vaccine was combined with DCs expressing anti–CTLA-4 and anti-GITR mRNA [100].

While most reports show GITR-GITRL interactions are co-stimulatory to T cells, data regarding the immune-stimulating potential of GITR on non–T-cell subsets, particularly NK cells, are conflicting. Human leukemia cell expression of GITR-L interferes with NK cell-mediated immunity through reverse signaling, which causes tumor production of suppressive cytokines, downregulation of co-stimulatory molecules, and evasion from immune detection [101, 102]. Nonetheless, in collaboration with Ludwig Cancer Research, Cancer Research Institute, and GITR, Inc. (Cambridge, MA, USA), we initiated a phase I trial (NCT01239134) of the humanized anti-GITR antibody TRX518 [103]. A trial examining the effects of GITRL-expressing DCs (with or without DC expressing anti–CTLA-4) plus a DC tumor vaccine in patients with melanoma is also ongoing (NCT01216436).

HVEM (CD270)

HVEM, another member of the TNFR superfamily [104], has multiple binding partners and various downstream effects that can positively and negatively modulate T-cell activation [105]. HVEM expression kinetics are the opposite of 4-1BB, OX40, and GITR: HVEM is highly expressed on resting T cells but downregulated upon activation. When cells return to less-activated states, HVEM expression is restored. There is significant HVEM expression on naive and memory, but not activated, B cells. Other immune cells, including Tregs, NK cells, monocytes, and immature DCs, also express HVEM. There are at least four natural ligands for HVEM: the inhibitory Ig-related molecules CD160 and B and T lymphocyte attenuator (BTLA; capable of bidirectional signaling) and the stimulatory TNF-related cytokines LIGHT and lymphotoxin-alpha [106109]. LIGHT is expressed upon T and B cell activation and on monocytes and immature DCs. BTLA is found on various lymphoid and myeloid cells, with particularly high expression on B cells and plasmacytoid DCs. CD160, by contrast, is observed only on subsets of T and NK cells [105]. While binding of LIGHT or lymphotoxin-α to HVEM triggers T and B cell stimulation and APC maturation and differentiation, CD160 or BTLA binding to HVEM on T or B cells has the reverse effect, inhibiting T and B cell activation.

Studies have explored the potential of the HVEM pathway as a therapeutic target. Murine P815 mastocytoma cells engineered to express the single-chain variable fragment (scFv) of an anti-HVEM agonistic monoclonal antibody stimulated proliferation and cytokine production in co-cultured T cells [110]. Expression of anti-HVEM scFv in vivo resulted in T-cell–dependent tumor rejection (in 80% of treated mice) and a lasting memory response. Combination of the anti-HVEM scFv-expressing tumor vaccine with anti–4-1BB monoclonal antibody therapy synergized to induce regression of more established tumors, leading to survival and memory responses not seen with either agent alone [111]. In melanoma patients, tumor-specific T cells were found to express BTLA concurrent with HVEM [108, 109]. Fourcade and colleagues tested the significance of this expression, showing that BTLA blockade enhanced the proliferation and antitumor activity of melanoma-specific CD8+ T cells [109]. Approaches to mono- or combination therapy are presented in the review by Pasero and colleagues [105]. However, manipulating the HVEM signaling pathway(s) provides a significant challenge, requiring agents that precisely induce HVEM-related immunostimulatory effects or block HVEM-related immunosuppressive effects.

CD27

CD27 has a key role in the generation of immunological memory via effects on T-cell expansion and survival, and B cell development [112114]. CD27 is constitutively expressed on conventional T cells (including populations of Tregs) and, like other members of the TNFRsf, is upregulated upon activation [115]. Expression of CD27 on human Tregs correlates with FoxP3 expression and suppressive functionality [116, 117]. CD27 is also expressed on subsets of B cells, most strikingly on plasma cells, for which it is a broadly accepted marker [118]. CD70, the natural ligand for CD27, is transiently expressed on activated APCs and T cells [119]. Ligation of CD27 by CD70 has stimulatory effects on T-cell proliferation, expansion, and survival dependent upon IL-2 autocrine signaling [120].

Triggering CD27 signaling in CAR-transduced T cells can enhance effector function, survival, and improve antitumor activity in a xenograft mouse model of ovarian cancer [121]. Constitutive CD70 expression on tumors or DCs improved antitumor immunity in murine lymphoma models, enhancing NK-mediated rejection of class-1 deficient tumor cells via perforin- and IFN-γ-dependent mechanisms [122]. CD70 expression also leads to generation of memory T-cell response to secondary tumor challenge. Conversely, intact CD27/CD70 signaling has been associated with decreased antitumor immune responses and an increased intratumoral Tregs (potentially due to improved Treg resistance to apoptosis and increased IL-2 secretion by effector T cells). Compared with wild-type mice, tumors in CD27-deficient mice had decreased vascularization and slower growth [123]. Furthermore, wild-type mice treated with an anti-CD27 blocking antibody had fewer Tregs and slower tumor growth than untreated mice. In this setting, Tregs were critical mediators of CD27-dependent effects on tumor growth. Schurch and colleagues similarly demonstrated that CD27 triggering by CD70 ligation in chronic myelogenous leukemia stem cells accelerated disease progression and was associated with activation of the Wnt pathway [124]. Such data complicate the consideration of CD27 as a target for tumor immunotherapy. The effects of CD27 pathway triggering may depend upon the environment in which the stimulus is delivered—within the tumor microenvironment or in peripheral lymphoid tissues—as well as length of exposure to stimulating agents.

A fully human monoclonal CD27 agonist antibody, CDX-1127 (Celldex Therapeutics, Inc. Needham, MA, USA), is being evaluated in a phase I clinical trial in select hematological and solid tumors [125]. Preclinical experiments with a mouse homologue showed delayed human hematological tumor growth in xenograft mouse models and murine tumor growth in immunocompetent mice [126]. Again, similar to HVEM, continued clinical development of CD27-related agents will require teasing apart the multiple inhibitory and co-stimulatory mechanisms that target multiple cell populations, and precise triggering of certain molecules in select environments to avoid exaggerating tumor-induced immunosuppression. Since Tregs appear to be a critical target of CD27 agonism, perhaps prior Treg depletion may be appropriate.

Conclusions

The success of cancer immunotherapy with agonist antibodies may vary according to tumor type, since the expression of the target molecules can differ across tumors (for example, the expression of HVEM in certain leukemias and lymphomas). Although a multifactorial approach to cancer therapy is attractive [127], the potential risk of autoimmune reactions and bystander tissue damage in the setting of immunostimulation should not be underestimated. The well-documented phase I study of the anti-CD28 monoclonal antibody TGN1412, in which six healthy volunteers suffered massive cytokine release and major organ damage, emphasized the need for extreme caution in trial design and execution when investigating immune activators [128]. It should be noted, however, that CD28 is constitutively expressed on T cells, whereas most of the TNFRsf members are upregulated upon activation. Therefore, targeting TNFRsf members might have more restricted downstream effects, including a more defined toxicity profile.

Targeting the co-stimulatory TNFRsf shows promise for the treatment of cancer. Although there have been life-threatening side effects with some agonist antibodies, there is obvious clinical activity, emphasizing the need for careful titration and dosing. From this perspective, it may not be necessary to “press on the gas” constantly if agonist antibodies are administered with a checkpoint blocker (i.e., anti-PD-1, anti-CTLA-4) to “remove the brake”.

The mechanisms of immune evasion are not the same for each patient’s tumor (even of the same tumor type) and as such, monotherapies may not achieve sufficient potency. Combinatorial approaches, which include targeting TNFRsf, show promise in preclinical studies. Thus, combination with chemotherapy or irradiation, which can induce immunogenic cell death and stimulate antitumor T cells, is a rational. Additionally, combining anti-TNFRsf antibodies with small molecule inhibitors should be considered. Although clinical studies of TNFRsf agonists are at an early stage, careful development may see additional therapies brought to patients who most need them most.

Abbreviations

AICD:

Activation-induced cell death

APCs:

Antigen-presenting cells

B7-H1:

B7-homolog 1

BTLA:

B and T lymphocyte attenuator

CAR:

Chimeric antigen receptor

CD137:

4-1BB

CLL:

Chronic lymphocytic leukemia

CTL:

Cytotoxic T lymphocyte

CTLA-4:

Cytotoxic T lymphocyte antigen-4

DCs:

Dendritic cells

ECs:

Endothelial cells

GITR:

Glucocorticoid-induced TNFR-related gene

GITRL:

GITR ligand

GM-CSF:

Granulocyte-macrophage colony-stimulating factor

HVEM:

Herpes virus entry mediator

IL:

Interleukin

MDSC:

Myeloid-derived suppressor cells

NK:

Natural killer

NKT:

Natural killer T cells

NSCLC:

Non-small cell lung cancer

OX40L:

OX40 ligand

PD-1:

Programmed death receptor-1

PD-L1/2:

Programmed death receptor-1 ligand1/2

RCC:

Renal cell carcinoma

scFv:

Single-chain variable fragment

TCR:

T-cell receptor

Teff:

Effector T cell

TILs:

Tumor-infiltrating lymphocytes

TNF:

Tumor-necrosis factor

TNFR:

Tumor-necrosis factor receptor

TNFRsf:

Tumor necrosis factor receptor superfamily

Treg:

Regulatory T cell.

References

  1. Hodi FS, O’Day SJ, McDermott DF, Weber RW, Sosman JA, Haanen JB, Gonzalez R, Robert C, Schadendorf D, Hassel JC, Akerley W, van den Eertwegh AJ, Lutzky J, Lorigan P, Vaubel JM, Linette GP, Hogg D, Ottensmeier CH, Lebbé C, Peschel C, Quirt I, Clark JI, Wolchok JD, Weber JS, Tian J, Yellin MJ, Nichol GM, Hoos A, Urba WJ: Improved survival with ipilimumab in patients with metastatic melanoma. N Engl J Med. 2010, 363: 711-723. 10.1056/NEJMoa1003466.

    CAS  PubMed  PubMed Central  Google Scholar 

  2. DuPage M, Mazumdar C, Schmidt LM, Cheung AF, Jacks T: Expression of tumour-specific antigens underlies cancer immunoediting. Nature. 2012, 482: 405-409. 10.1038/nature10803.

    CAS  PubMed  PubMed Central  Google Scholar 

  3. Schreiber RD, Old LJ, Smyth MJ: Cancer immunoediting: integrating immunity’s roles in cancer suppression and promotion. Science. 2011, 331: 1565-1570. 10.1126/science.1203486.

    CAS  PubMed  Google Scholar 

  4. Dunn GP, Bruce AT, Ikeda H, Old LJ, Schreiber RD: Cancer immunoediting: from immunosurveillance to tumor escape. Nat Immunol. 2002, 3: 991-998. 10.1038/ni1102-991.

    CAS  PubMed  Google Scholar 

  5. Matsushita H, Vesely MD, Koboldt DC, Rickert CG, Uppaluri R, Magrini VJ, Arthur CD, White JM, Chen YS, Shea LK, Hundal J, Wendl MC, Demeter R, Wylie T, Allison JP, Smyth MJ, Old LJ, Mardis ER, Schreiber RD: Cancer exome analysis reveals a T-cell-dependent mechanism of cancer immunoediting. Nature. 2012, 482: 400-404. 10.1038/nature10755.

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Draghiciu O, Nijman HW, Daemen T: From tumor immunosuppression to eradication: targeting homing and activity of immune effector cells to tumors. Clin Dev Immunol. 2011, 2011: 439053-

    PubMed  PubMed Central  Google Scholar 

  7. Mellman I, Coukos G, Dranoff G: Cancer immunotherapy comes of age. Nature. 2011, 480: 480-489. 10.1038/nature10673.

    CAS  PubMed  PubMed Central  Google Scholar 

  8. Töpfer K, Kempe S, Müller N, Schmitz M, Bachmann M, Cartellieri M, Schackert G, Temme A: Tumor evasion from T cell surveillance. J Biomed Biotechnol. 2011, 2011: 918471-

    PubMed  PubMed Central  Google Scholar 

  9. Fuertes MB, Kacha AK, Kline J, Woo SR, Kranz DM, Murphy KM, Gajewski TF: Host type I IFN signals are required for antitumor CD8+ T cell responses through CD8{alpha} + dendritic cells. J Exp Med. 2011, 208: 2005-2016. 10.1084/jem.20101159.

    CAS  PubMed  PubMed Central  Google Scholar 

  10. Zou W: Immunosuppressive networks in the tumour environment and their therapeutic relevance. Nat Rev Cancer. 2005, 5: 263-274. 10.1038/nrc1586.

    CAS  PubMed  Google Scholar 

  11. Schaer DA, Li Y, Merghoub T, Rizzuto GA, Shemesh A, Cohen AD, Li Y, Avogadri F, Toledo-Crow R, Houghton AN, Wolchok JD: Detection of intra-tumor self antigen recognition during melanoma tumor progression in mice using advanced multimode confocal/two photon microscope. PLoS One. 2011, 6: e21214-10.1371/journal.pone.0021214.

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Linsley PS, Greene JL, Brady W, Bajorath J, Ledbetter JA, Peach R: Human B7-1 (CD80) and B7-2 (CD86) bind with similar avidities but distinct kinetics to CD28 and CTLA-4 receptors. Immunity. 1994, 1: 793-801. 10.1016/S1074-7613(94)80021-9.

    CAS  PubMed  Google Scholar 

  13. Teft WA, Kirchhof MG, Madrenas J: A molecular perspective of CTLA-4 function. Annu Rev Immunol. 2006, 24: 65-97. 10.1146/annurev.immunol.24.021605.090535.

    CAS  PubMed  Google Scholar 

  14. Krummel MF, Allison JP: CD28 and CTLA-4 have opposing effects on the response of T cells to stimulation. J Exp Med. 1995, 182: 459-465. 10.1084/jem.182.2.459.

    CAS  PubMed  Google Scholar 

  15. Korman A, Yellin M, Keler T: Tumor immunotherapy: preclinical and clinical activity of anti-CTLA4 antibodies. Curr Opin Investig Drugs. 2005, 6: 582-591.

    CAS  PubMed  Google Scholar 

  16. Yuan J, Adamow M, Ginsberg BA, Rasalan TS, Ritter E, Gallardo HF, Xu Y, Pogoriler E, Terzulli SL, Kuk D, Panageas KS, Ritter G, Sznol M, Halaban R, Jungbluth AA, Allison JP, Old LJ, Wolchok JD, Gnjatic S: Integrated NY-ESO-1 antibody and CD8+ T-cell responses correlate with clinical benefit in advanced melanoma patients treated with ipilimumab. Proc Natl Acad Sci U S A. 2011, 108: 16723-16728. 10.1073/pnas.1110814108.

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Freeman GJ, Long AJ, Iwai Y, Bourque K, Chernova T, Nishimura H, Fitz LJ, Malenkovich N, Okazaki T, Byrne MC, Horton HF, Fouser L, Carter L, Ling V, Bowman MR, Carreno BM, Collins M, Wood CR, Honjo T: Engagement of the PD-1 immunoinhibitory receptor by a novel B7 family member leads to negative regulation of lymphocyte activation. J Exp Med. 2000, 192: 1027-1034. 10.1084/jem.192.7.1027.

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Chemnitz JM, Parry RV, Nichols KE, June CH, Riley JL: SHP-1 and SHP-2 associate with immunoreceptor tyrosine-based switch motif of programmed death 1 upon primary human T cell stimulation, but only receptor ligation prevents T cell activation. J Immunol. 2004, 173: 945-954. 10.4049/jimmunol.173.2.945.

    CAS  PubMed  Google Scholar 

  19. Barber DL, Wherry EJ, Masopust D, Zhu B, Allison JP, Sharpe AH, Freeman GJ, Ahmed R: Restoring function in exhausted CD8 T cells during chronic viral infection. Nature. 2006, 439: 682-687. 10.1038/nature04444.

    CAS  PubMed  Google Scholar 

  20. Topalian SL, Hodi FS, Brahmer JR, Gettinger SN, Smith DC, McDermott DF, Powderly JD, Carvajal RD, Sosman JA, Atkins MB, Leming PD, Spigel DR, Antonia SJ, Horn L, Drake CG, Pardoll DM, Chen L, Sharfman WH, Anders RA, Taube JM, McMiller TL, Xu H, Korman AJ, Jure-Kunkel M, Agrawal S, McDonald D, Kollia GD, Gupta A, Wigginton JM, Sznol M: Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N Engl J Med. 2012, 366: 2443-2454. 10.1056/NEJMoa1200690.

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Topalian SL, Brahmer JR, Hodi FS, McDermott DF, Smith DC, Gettinger S, Taube JM, Gupta A, Wigginton JM, Sznol M: Anti-programmed death-1 (PD-1) (BMS-936558/MDX-1106/ONO-4538) in patients with advanced solid tumors: clinical activity, safety, and molecular markers [Abstract]. Annals Oncol. 2012, 23 (suppl 9): xi157-

    Google Scholar 

  22. Wolchok JD, Kluger H, Callahan MK, Postow MA, Rizvi NA, Lesokhin AM, Segal NH, Ariyan CE, Gordon RA, Reed K, Burke MM, Caldwell A, Kronenberg SA, Agunwamba BU, Zhang X, Lowy I, Inzunza HD, Feely W, Horak CE, Hong Q, Korman AJ, Wigginton JM, Gupta A, Sznol M: Nivolumab plus ipilimumab in advanced melanoma. N Engl J Med. 2013, 369: 122-133. 10.1056/NEJMoa1302369.

    CAS  PubMed  Google Scholar 

  23. Berger R, Rotem-Yehudar R, Slama G, Landes S, Kneller A, Leiba M, Koren-Michowitz M, Shimoni A, Nagler A: 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, 14: 3044-3051. 10.1158/1078-0432.CCR-07-4079.

    CAS  PubMed  Google Scholar 

  24. Patnaik A, Kang SP, Tolcher AW, Rasco DW, Papadopoulos KP, Beeram M, Drengler R, Chen C, Smith L, Perez C, Gergich K, Lehnert M: Phase I study of MK-3475 (anti-PD-1 monoclonal antibody) in patients with advanced solid tumors [Abstract]. J Clin Oncol. 2012, 30 (Suppl abstr 2512):

  25. MedImmune joins forces with leading cancer organizations to advance novel immunotherapy research [press release].https://www.medimmune.com/media/press-releases/2012/10/09/medimmune-joins-forces-with-leading-cancer-organizations-to-advance-novel-immunotherapy-research,

  26. Brahmer JR, Tykodi SS, Chow LQ, Hwu WJ, Topalian SL, Hwu P, Drake CG, Camacho LH, Kauh J, Odunsi K, Pitot HC, Hamid O, Bhatia S, Martins R, Eaton K, Chen S, Salay TM, Alaparthy S, Grosso JF, Korman AJ, Parker SM, Agrawal S, Goldberg SM, Pardoll DM, Gupta A, Wigginton JM: Safety and activity of anti-PD-L1 antibody in patients with advanced cancer. N Engl J Med. 2012, 366: 2455-2465. 10.1056/NEJMoa1200694.

    CAS  PubMed  PubMed Central  Google Scholar 

  27. GlaxoSmithKline and Amplimmune form global strategic collaboration [press release].http://www.prnewswire.com/news-releases/glaxosmithkline-and-amplimmune-form-global-strategic-collaboration-99938599.html,

  28. Watts TH: TNF/TNFR family members in costimulation of T cell responses. Annu Rev Immunol. 2005, 23: 23-68. 10.1146/annurev.immunol.23.021704.115839.

    CAS  PubMed  Google Scholar 

  29. So T, Lee SW, Croft M: Tumor necrosis factor/tumor necrosis factor receptor family members that positively regulate immunity. Int J Hematol. 2006, 83: 1-11. 10.1532/IJH97.05120.

    CAS  PubMed  Google Scholar 

  30. Kwon BS, Weissman SM: cDNA sequences of two inducible T-cell genes. Proc Natl Acad Sci U S A. 1989, 86: 1963-1967. 10.1073/pnas.86.6.1963.

    CAS  PubMed  PubMed Central  Google Scholar 

  31. Pollok KE, Kim YJ, Zhou Z, Hurtado J, Kim KK, Pickard RT, Kwon BS: Inducible T cell antigen 4-1BB. Analysis of expression and function. J Immunol. 1993, 150: 771-781.

    CAS  PubMed  Google Scholar 

  32. Shuford WW, Klussman K, Tritchler DD, Loo DT, Chalupny J, Siadak AW, Brown TJ, Emswiler J, Raecho H, Larsen CP, Pearson TC, Ledbetter JA, Aruffo A, Mittler RS: 4-1BB costimulatory signals preferentially induce CD8+ T cell proliferation and lead to the amplification in vivo of cytotoxic T cell responses. J Exp Med. 1997, 186: 47-55. 10.1084/jem.186.1.47.

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Wilcox RA, Chapoval AI, Gorski KS, Otsuji M, Shin T, Flies DB, Tamada K, Mittler RS, Tsuchiya H, Pardoll DM, Chen L: Cutting edge: expression of functional CD137 receptor by dendritic cells. J Immunol. 2002, 168: 4262-4267. 10.4049/jimmunol.168.9.4262.

    CAS  PubMed  Google Scholar 

  34. McHugh RS, Whitters MJ, Piccirillo CA, Young DA, Shevach EM, Collins M, Byrne MC: CD4(+)CD25(+) immunoregulatory T cells: gene expression analysis reveals a functional role for the glucocorticoid-induced TNF receptor. Immunity. 2002, 16: 311-323. 10.1016/S1074-7613(02)00280-7.

    CAS  PubMed  Google Scholar 

  35. Vinay DS, Kwon BS: 4-1BB signaling beyond T cells. Cell Mol Immunol. 2011, 8: 281-284. 10.1038/cmi.2010.82.

    CAS  PubMed  PubMed Central  Google Scholar 

  36. Vinay DS, Kwon BS: Immunotherapy of cancer with 4-1BB. Mol Cancer Ther. 2012, 11: 1062-1070. 10.1158/1535-7163.MCT-11-0677.

    CAS  PubMed  Google Scholar 

  37. Saoulli K, Lee SY, Cannons JL, Yeh WC, Santana A, Goldstein MD, Bangia N, DeBenedette MA, Mak TW, Choi Y, Watts TH: CD28-independent, TRAF2-dependent costimulation of resting T cells by 4–1BB ligand. J Exp Med. 1998, 187: 1849-1862. 10.1084/jem.187.11.1849.

    CAS  PubMed  PubMed Central  Google Scholar 

  38. Lee HW, Park SJ, Choi BK, Kim HH, Nam KO, Kwon BS: 4-1BB promotes the survival of CD8+ T lymphocytes by increasing expression of Bcl-xL and Bfl-1. J Immunol. 2002, 169: 4882-4888. 10.4049/jimmunol.169.9.4882.

    PubMed  Google Scholar 

  39. Stärck L, Scholz C, Dörken B, Daniel PT: Costimulation by CD137/4-1BB inhibits T cell apoptosis and induces Bcl-xL and c-FLIP(short) via phosphatidylinositol 3-kinase and AKT/protein kinase B. Eur J Immunol. 2005, 35: 1257-1266. 10.1002/eji.200425686.

    PubMed  Google Scholar 

  40. Hernandez-Chacon JA, Li Y, Wu RC, Bernatchez C, Wang Y, Weber J, Hwu P, Radvanyi LG: Costimulation through the CD137/4-1BB pathway protects human melanoma tumor-infiltrating lymphocytes from activation-induced cell death and enhances antitumor effector function. J Immunother. 2011, 34: 236-250. 10.1097/CJI.0b013e318209e7ec.

    CAS  PubMed  PubMed Central  Google Scholar 

  41. Melero I, Shuford WW, Newby SA, Aruffo A, Ledbetter JA, Hellström KE, Mittler RS, Chen L: Monoclonal antibodies against the 4-1BB T-cell activation molecule eradicate established tumors. Nat Med. 1997, 3: 682-685. 10.1038/nm0697-682.

    CAS  PubMed  Google Scholar 

  42. Sabel MS, Conway TF, Chen FA, Bankert RB: Monoclonal antibodies directed against the T-cell activation molecule CD137 (interleukin-A or4-1BB) block human lymphocyte-mediated suppression of tumor xenografts in severe combined immunodeficient mice. J Immunother. 2000, 23: 362-368. 10.1097/00002371-200005000-00009.

    CAS  PubMed  Google Scholar 

  43. Kim JA, Averbook BJ, Chambers K, Rothchild K, Kjaergaard J, Papay R, Shu S: Divergent effects of 4-1BB antibodies on antitumor immunity and on tumor-reactive T-cell generation. Cancer Res. 2001, 61: 2031-2037.

    CAS  PubMed  Google Scholar 

  44. Lin GH, Liu Y, Ambagala T, Kwon BS, Ohashi PS, Watts TH: Evaluating the cellular targets of anti-4-1BB agonist antibody during immunotherapy of a pre-established tumor in mice. PLoS One. 2010, 5: e11003-10.1371/journal.pone.0011003.

    PubMed  PubMed Central  Google Scholar 

  45. Uno T, Takeda K, Kojima Y, Yoshizawa H, Akiba H, Mittler RS, Gejyo F, Okumura K, Yagita H, Smyth MJ: Eradication of established tumors in mice by a combination antibody-based therapy. Nat Med. 2006, 12: 693-698. 10.1038/nm1405.

    CAS  PubMed  Google Scholar 

  46. Takeda K, Kojima Y, Uno T, Hayakawa Y, Teng MW, Yoshizawa H, Yagita H, Gejyo F, Okumura K, Smyth MJ: Combination therapy of established tumors by antibodies targeting immune activating and suppressing molecules. J Immunol. 2010, 184: 5493-5501. 10.4049/jimmunol.0903033.

    CAS  PubMed  Google Scholar 

  47. Curran MA, Kim M, Montalvo W, Al-Shamkhani A, Allison JP: Combination CTLA-4 blockade and 4-1BB activation enhances tumor rejection by increasing T-cell infiltration, proliferation, and cytokine production. PLoS One. 2011, 6: e19499-10.1371/journal.pone.0019499.

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Youlin K, Li Z, Xiaodong W, Xiuheng L, Hengchen Z: Combination immunotherapy with 4–1 BBL and CTLA-4 blockade for the treatment of prostate cancer. Clin Dev Immunol. 2012, 2012: 439235-

    PubMed  PubMed Central  Google Scholar 

  49. Kohrt HE, Houot R, Weiskopf K, Goldstein MJ, Scheeren F, Czerwinski D, Colevas AD, Weng WK, Clarke MF, Carlson RW, Stockdale FE, Mollick JA, Chen L, Levy R: Stimulation of natural killer cells with a CD137-specific antibody enhances trastuzumab efficacy in xenotransplant models of breast cancer. J Clin Invest. 2012, 122: 1066-1075. 10.1172/JCI61226.

    CAS  PubMed  PubMed Central  Google Scholar 

  50. Shi W, Siemann DW: Augmented antitumor effects of radiation therapy by 4-1BB antibody (BMS-469492) treatment. Anticancer Res. 2006, 26: 3445-3453.

    CAS  PubMed  Google Scholar 

  51. Verbrugge I, Hagekyriakou J, Sharp LL, Galli M, West A, McLaughlin NM, Duret H, Yagita H, Johnstone RW, Smyth MJ, Haynes NM: Radiotherapy increases the permissiveness of established mammary tumors to rejection by immunomodulatory antibodies. Cancer Res. 2012, 72: 3163-3174. 10.1158/0008-5472.CAN-12-0210.

    CAS  PubMed  Google Scholar 

  52. Dubrot J, Palazón A, Alfaro C, Azpilikueta A, Ochoa MC, Rouzaut A, Martinez-Forero I, Teijeira A, Berraondo P, Le Bon A, Hervás-Stubbs S, Melero I: Intratumoral injection of interferon-α and systemic delivery of agonist anti-CD137 monoclonal antibodies synergize for immunotherapy. Int J Cancer. 2011, 128: 105-118. 10.1002/ijc.25333.

    CAS  PubMed  Google Scholar 

  53. Lee H, Park HJ, Sohn HJ, Kim JM, Kim SJ: Combinatorial therapy for liver metastatic colon cancer: dendritic cell vaccine and low-dose agonistic anti-4-1BB antibody co-stimulatory signal. J Surg Res. 2011, 169: e43-e50. 10.1016/j.jss.2011.03.067.

    CAS  PubMed  Google Scholar 

  54. Ascierto PA, Simeone E, Sznol M, Fu YX, Melero I: Clinical experiences with anti-CD137 and anti-PD1 therapeutic antibodies. Semin Oncol. 2010, 37: 508-516. 10.1053/j.seminoncol.2010.09.008.

    CAS  PubMed  Google Scholar 

  55. Hwu W-J: Targeted therapy for metastatic melanoma: from bench to bedside.http://www.healio.com/hematology-oncology/melanoma-skin-cancer/news/print/hematology-oncology/%7B77E71A11-1FD1-4193-A2F7-7C50C6121C1F%7D/Targeted-therapy-for-metastatic-melanoma-From-bench-to-bedside,

  56. Porter DL, Levine BL, Kalos M, Bagg A, June CH: Chimeric antigen receptor-modified T cells in chronic lymphoid leukemia. N Engl J Med. 2011, 365: 725-733. 10.1056/NEJMoa1103849.

    CAS  PubMed  PubMed Central  Google Scholar 

  57. Paterson DJ, Jefferies WA, Green JR, Brandon MR, Corthesy P, Puklavec M, Williams AF: Antigens of activated rat T lymphocytes including a molecule of 50,000 Mr detected only on CD4 positive T blasts. Mol Immunol. 1987, 24: 1281-1290. 10.1016/0161-5890(87)90122-2.

    CAS  PubMed  Google Scholar 

  58. Baum PR, Gayle RB, Ramsdell F, Srinivasan S, Sorensen RA, Watson ML, Seldin MF, Baker E, Sutherland GR, Clifford KN, Alderson MR, Goodwin RG, Fanslow WC: Molecular characterization of murine and human OX40/OX40 ligand systems: identification of a human OX40 ligand as the HTLV-1-regulated protein gp34. EMBO J. 1994, 13: 3992-4001.

    CAS  PubMed  PubMed Central  Google Scholar 

  59. Godfrey WR, Fagnoni FF, Harara MA, Buck D, Engleman EG: Identification of a human OX-40 ligand, a costimulator of CD4+ T cells with homology to tumor necrosis factor. J Exp Med. 1994, 180: 757-762. 10.1084/jem.180.2.757.

    CAS  PubMed  Google Scholar 

  60. Weinberg AD, Morris NP, Kovacsovics-Bankowski M, Urba WJ, Curti BD: Science gone translational: the OX40 agonist story. Immunol Rev. 2011, 244: 218-231. 10.1111/j.1600-065X.2011.01069.x.

    CAS  PubMed  PubMed Central  Google Scholar 

  61. Pardee AD, McCurry D, Alber S, Hu P, Epstein AL, Storkus WJ: A therapeutic OX40 agonist dynamically alters dendritic, endothelial, and T cell subsets within the established tumor microenvironment. Cancer Res. 2010, 70: 9041-9052. 10.1158/0008-5472.CAN-10-1369.

    CAS  PubMed  PubMed Central  Google Scholar 

  62. Jensen SM, Maston LD, Gough MJ, Ruby CE, Redmond WL, Crittenden M, Li Y, Puri S, Poehlein CH, Morris N, Kovacsovics-Bankowski M, Moudgil T, Twitty C, Walker EB, Hu HM, Urba WJ, Weinberg AD, Curti BD, Fox BA: Signaling through OX40 enhances antitumor immunity. Semin Oncol. 2010, 37: 524-532. 10.1053/j.seminoncol.2010.09.013.

    CAS  PubMed  PubMed Central  Google Scholar 

  63. Takeda I, Ine S, Killeen N, Ndhlovu LC, Murata K, Satomi S, Sugamura K, Ishii N: Distinct roles for theOX40-40 ligand interaction in regulatory and nonregulatory T cells. J Immunol. 2004, 172: 3580-3589. 10.4049/jimmunol.172.6.3580.

    CAS  PubMed  Google Scholar 

  64. Redmond WL, Triplett T, Floyd K, Weinberg AD: Dual anti-OX40/IL-2 therapy augments tumor immunotherapy via IL-2R-mediated regulation of OX40 expression. PLoS One. 2012, 7: e34467-10.1371/journal.pone.0034467.

    CAS  PubMed  PubMed Central  Google Scholar 

  65. Griseri T, Asquith M, Thompson C, Powrie F: OX40 is required for regulatory T cell-mediated control of colitis. J Exp Med. 2010, 207: 699-709. 10.1084/jem.20091618.

    CAS  PubMed  PubMed Central  Google Scholar 

  66. Piconese S, Pittoni P, Burocchi A, Gorzanelli A, Carè A, Tripodo C, Colombo MP: A non-redundant role for OX40 in the competitive fitness of Treg in response to IL-2. Eur J Immunol. 2010, 40: 2902-2913. 10.1002/eji.201040505.

    PubMed  Google Scholar 

  67. Ruby CE, Yates MA, Hirschhorn-Cymerman D, Chlebeck P, Wolchok JD, Houghton AN, Offner H, Weinberg AD: Cutting edge: OX40agonists can drive regulatory T cell expansion if the cytokine milieu is right. J Immunol. 2009, 183: 4853-4857. 10.4049/jimmunol.0901112.

    CAS  PubMed  PubMed Central  Google Scholar 

  68. Valzasina B, Guiducci C, Dislich H, Killeen N, Weinberg AD, Colombo MP: Triggering of OX40 (CD134) on CD4 + CD25+ T cells blocks their inhibitory activity: a novel regulatory role for OX40 and its comparison with GITR. Blood. 2005, 105: 2845-2851. 10.1182/blood-2004-07-2959.

    CAS  PubMed  Google Scholar 

  69. Croft M: Control of immunity by the TNFR- related molecule OX40 (CD134). Annu Rev Immunol. 2010, 28: 57-78. 10.1146/annurev-immunol-030409-101243.

    CAS  PubMed  PubMed Central  Google Scholar 

  70. Weinberg AD, Rivera MM, Prell R, Morris A, Ramstad T, Vetto JT, Urba WJ, Alvord G, Bunce C, Shields J: Engagement of the OX-40 receptor in vivo enhances antitumor immunity. J Immunol. 2000, 164: 2160-2169. 10.4049/jimmunol.164.4.2160.

    CAS  PubMed  Google Scholar 

  71. Gough MJ, Crittenden MR, Sarff MC, Pang P, Seung SK, Vetto JT, Hu HM, Redmond WL, Holland J, Weinberg AD: Adjuvant therapy with agonistic antibodies to CD134 (OX40) increases local control after surgical or radiation therapy of cancer in mice. J Immunother. 2010, 33: 798-809. 10.1097/CJI.0b013e3181ee7095.

    CAS  PubMed  PubMed Central  Google Scholar 

  72. Pan PY, Zang Y, Weber K, Meseck ML, Chen SH: OX40 ligation enhances primary and memory cytotoxic T lymphocyte responses in an immunotherapy for hepatic colon metastases. Mol Ther. 2002, 6: 528-536. 10.1006/mthe.2002.0699.

    CAS  PubMed  Google Scholar 

  73. Houot R, Levy R: T-cell modulation combined with intratumoral CpG cures lymphoma in a mouse model without the need for chemotherapy. Blood. 2009, 113: 3546-3552. 10.1182/blood-2008-07-170274.

    CAS  PubMed  PubMed Central  Google Scholar 

  74. Watanabe A, Hara M, Chosa E, Nakamura K, Sekiya R, Shimizu T, Onitsuka T: Combination of adoptive cell transfer and antibody injection can eradicate established tumors in mice–an in vivo study using anti-OX40mAb, anti-CD25mAb and anti-CTLA4mAb. Immunopharmacol Immunotoxicol. 2010, 32: 238-245. 10.3109/08923970903222355.

    CAS  PubMed  Google Scholar 

  75. Garrison K, Hahn T, Lee WC, Ling LE, Weinberg AD, Akporiaye ET: The small molecule TGF-β signaling inhibitor SM16 synergizes with agonistic OX40 antibody to suppress established mammary tumors and reduce spontaneous metastasis. Cancer Immunol Immunother. 2012, 61: 511-521. 10.1007/s00262-011-1119-y.

    CAS  PubMed  PubMed Central  Google Scholar 

  76. Hirschhorn-Cymerman D, Rizzuto GA, Merghoub T, Cohen AD, Avogadri F, Lesokhin AM, Weinberg AD, Wolchok JD, Houghton AN: OX40 engagement and chemotherapy combination provides potent antitumor immunity with concomitant regulatory T cell apoptosis. J Exp Med. 2009, 206: 1103-1116. 10.1084/jem.20082205.

    CAS  PubMed  PubMed Central  Google Scholar 

  77. Curti BD, Kovacsovics-Bankowski M, Morris N, Walker E, Chisholm L, Floyd K, Walker J, Gonzalez I, Meeuwsen T, Fox BA, Moudgil T, Miller W, Haley D, Coffey T, Fisher B, Delanty-Miller L, Rymarchyk N, Kelly T, Crocenzi T, Bernstein E, Sanborn R, Urba WJ, Weinberg AD: OX40 is a potent immune-stimulating target in late-stage cancer patients. Cancer Res. 2013, 73: 7189-98. 10.1158/0008-5472.CAN-12-4174.

    CAS  PubMed  PubMed Central  Google Scholar 

  78. Voo KS, Bover L, Harline ML, Vien LT, Facchinetti V, Arima K, Kwak LW, Liu YJ: Antibodies targeting human OX40 expand effector T cells and block inducible and natural regulatory T cell function. J Immunol. 2013, 191: 3641-3650. 10.4049/jimmunol.1202752.

    CAS  PubMed  Google Scholar 

  79. Morris NP, Peters C, Montler R, Hu HM, Curti BD, Urba WJ, Weinberg AD: Development and characterization of recombinant human Fc:OX40L fusion protein linked via a coiled-coil trimerization domain. Mol Immunol. 2007, 44: 3112-3121. 10.1016/j.molimm.2007.02.004.

    CAS  PubMed  PubMed Central  Google Scholar 

  80. Nocentini G, Giunchi L, Ronchetti S, Krausz LT, Bartoli A, Moraca R, Migliorati G, Riccardi C: A new member of the tumor necrosis factor/nerve growth factor receptor family inhibits T cell receptor-induced apoptosis. Proc Natl Acad Sci U S A. 1997, 94: 6216-6221. 10.1073/pnas.94.12.6216.

    CAS  PubMed  PubMed Central  Google Scholar 

  81. Nocentini G, Ronchetti S, Petrillo MG, Riccardi C: Pharmacological modulation of GITRL/GITR system: therapeutic perspectives. Br J Pharmacol. 2012, 165: 2089-2099. 10.1111/j.1476-5381.2011.01753.x.

    CAS  PubMed  PubMed Central  Google Scholar 

  82. Schaer DA, Murphy JT, Wolchok JD: Modulation of GITR for cancer immunotherapy. Curr Opin Immunol. 2012, 24: 217-224. 10.1016/j.coi.2011.12.011.

    CAS  PubMed  PubMed Central  Google Scholar 

  83. Lacal PM, Petrillo MG, Ruffini F, Muzi A, Bianchini R, Ronchetti S, Migliorati G, Riccardi C, Graziani G, Nocentini G: Glucocorticoid-induced tumor necrosis factor receptor family-related ligand triggering upregulates vascular cell adhesion molecule-1 and intercellular adhesion molecule-1 and promotes leukocyte adhesion. J Pharmacol Exp Ther. 2013, 347: 164-172. 10.1124/jpet.113.207605.

    CAS  PubMed  Google Scholar 

  84. Chattopadhyay K, Ramagopal UA, Brenowitz M, Nathenson SG, Almo SC: Evolution of GITRL immune function: murine GITRL exhibits unique structural and biochemical properties within the TNF superfamily. Proc Natl Acad Sci U S A. 2008, 105: 635-640. 10.1073/pnas.0710529105.

    CAS  PubMed  PubMed Central  Google Scholar 

  85. Zhou Z, Tone Y, Song X, Furuuchi K, Lear JD, Waldmann H, Tone M, Greene MI, Murali R: Structural basis for ligand-mediated mouse GITR activation. Proc Natl Acad Sci USA. 2008, 105: 641-645. 10.1073/pnas.0711206105.

    CAS  PubMed  PubMed Central  Google Scholar 

  86. Shimizu J, Yamazaki S, Takahashi T, Ishida Y, Sakaguchi S: Stimulation of CD25(+)CD4(+) regulatory T cells through GITR breaks immunological self-tolerance. Nat Immunol. 2002, 3: 135-142. 10.1038/ni759.

    CAS  PubMed  Google Scholar 

  87. Stephens GL, McHugh RS, Whitters MJ, Young DA, Luxenberg D, Carreno BM, Collins M, Shevach EM: Engagement of glucocorticoid-induced TNFR family-related receptor on effector T cells by its ligand mediates resistance to suppression by CD4 + CD25+ T cells. J Immunol. 2004, 173: 5008-5020. 10.4049/jimmunol.173.8.5008.

    CAS  PubMed  Google Scholar 

  88. Cohen AD, Schaer DA, Liu C, Li Y, Hirschhorn-Cymmerman D, Kim SC, Diab A, Rizzuto G, Duan F, Perales MA, Merghoub T, Houghton AN, Wolchok JD: Agonist anti-GITR monoclonal antibody induces melanoma tumor immunity in mice by altering regulatory T cell stability and intra-tumor accumulation. PLoS One. 2010, 5: e10436-10.1371/journal.pone.0010436.

    PubMed  PubMed Central  Google Scholar 

  89. Turk MJ, Guevara-Patiño JA, Rizzuto GA, Engelhorn ME, Sakaguchi S, Houghton AN: Concomitant tumor immunity to a poorly immunogenic melanoma is prevented by regulatory T cells. J Exp Med. 2004, 200: 771-782. 10.1084/jem.20041130.

    CAS  PubMed  PubMed Central  Google Scholar 

  90. Ko K, Yamazaki S, Nakamura K, Nishioka T, Hirota K, Yamaguchi T, Shimizu J, Nomura T, Chiba T, Sakaguchi S: Treatment of advanced tumors with agonistic anti-GITR mAb and its effects on tumor-infiltrating Foxp3 + CD25 + CD4+ regulatory T cells. J Exp Med. 2005, 202: 885-891. 10.1084/jem.20050940.

    CAS  PubMed  PubMed Central  Google Scholar 

  91. Piao J, Kamimura Y, Iwai H, Cao Y, Kikuchi K, Hashiguchi M, Masunaga T, Jiang H, Tamura K, Sakaguchi S, Azuma M: Enhancement of T-cell-mediated anti-tumour immunity via the ectopically expressed glucocorticoid-induced tumour necrosis factor receptor-related receptor ligand (GITRL) on tumours. Immunology. 2009, 127: 489-499. 10.1111/j.1365-2567.2008.03036.x.

    CAS  PubMed  PubMed Central  Google Scholar 

  92. Schaer DA, Budhu S, Liu C, Bryson CF, Malandro NM, Cohen AD, Zhong H, Yang X, Houghton AN, Merghoub T, Wolchok JD: GITR pathway activation abrogates tumor immune suppression through loss of regulatory T cell lineage stability. Cancer Immunol Res. 2013, 1: 320-331. 10.1158/2326-6066.CIR-13-0086.

    CAS  PubMed  Google Scholar 

  93. Cohen AD, Diab A, Perales MA, Wolchok JD, Rizzuto G, Merghoub T, Huggins D, Liu C, Turk MJ, Restifo NP, Sakaguchi S, Houghton AN: Agonist anti-GITR antibody enhances vaccine-induced CD8(+) T-cell responses and tumor immunity. Cancer Res. 2006, 66: 4904-4912. 10.1158/0008-5472.CAN-05-2813.

    CAS  PubMed  PubMed Central  Google Scholar 

  94. Bulliard Y, Jolicoeur R, Windman M, Rue SM, Ettenberg S, Knee DA, Wilson NS, Dranoff G, Brogdon JL: Activating Fc γ receptors contribute to the antitumor activities of immunoregulatory receptor-targeting antibodies. J Exp Med. 2013, 210: 1685-1693. 10.1084/jem.20130573.

    CAS  PubMed  PubMed Central  Google Scholar 

  95. Nishikawa H, Kato T, Hirayama M, Orito Y, Sato E, Harada N, Gnjatic S, Old LJ, Shiku H: Regulatory T cell-resistant CD8+ T cells induced by glucocorticoid-induced tumor necrosisfactor receptor signaling. Cancer Res. 2008, 68: 5948-5954. 10.1158/0008-5472.CAN-07-5839.

    CAS  PubMed  Google Scholar 

  96. Boczkowski D, Lee J, Pruitt S, Nair S: Dendritic cells engineered to secrete anti-GITR antibodies are effective adjuvants to dendritic cell-based immunotherapy. Cancer Gene Ther. 2009, 16: 900-911. 10.1038/cgt.2009.39.

    CAS  PubMed  Google Scholar 

  97. Hoffmann C, Stanke J, Kaufmann AM, Loddenkemper C, Schneider A, Cichon G: Combining T-cell vaccination and application of agonistic anti-GITR mAb (DTA-1) induces complete eradication of HPV oncogene expressing tumors in mice. J Immunother. 2010, 33: 136-145. 10.1097/CJI.0b013e3181badc46.

    CAS  PubMed  Google Scholar 

  98. Imai N, Ikeda H, Tawara I, Wang L, Wang L, Nishikawa H, Kato T, Shiku H: Glucocorticoid-induced tumor necrosis factor receptor stimulation enhances the multifunctionality of adoptively transferred tumor antigen-specific CD8+ T cells with tumor regression. Cancer Sci. 2009, 100: 1317-1325. 10.1111/j.1349-7006.2009.01179.x.

    CAS  PubMed  Google Scholar 

  99. Mitsui J, Nishikawa H, Muraoka D, Wang L, Noguchi T, Sato E, Kondo S, Allison JP, Sakaguchi S, Old LJ, Kato T, Shiku H: Two distinct mechanisms of augmented antitumor activity by modulation of immunostimulatory/inhibitory signals. Clin Cancer Res. 2010, 16: 2781-2791. 10.1158/1078-0432.CCR-09-3243.

    CAS  PubMed  Google Scholar 

  100. Pruitt SK, Boczkowski D, de Rosa N, Haley NR, Morse MA, Tyler DS, Dannull J, Nair S: Enhancement of anti-tumor immunity through local modulation of CTLA-4 and GITR by dendritic cells. Eur J Immunol. 2011, 41: 3553-3563. 10.1002/eji.201141383.

    CAS  PubMed  Google Scholar 

  101. Baessler T, Krusch M, Schmiedel BJ, Kloss M, Baltz KM, Wacker A, Schmetzer HM, Salih HR: Glucocorticoid-induced tumor necrosis factor receptor-related protein ligand subverts immunosurveillance of acute myeloid leukemia in humans. Cancer Res. 2009, 69: 1037-1045.

    CAS  PubMed  Google Scholar 

  102. Baltz KM, Krusch M, Bringmann A, Brossart P, Mayer F, Kloss M, Baessler T, Kumbier I, Peterfi A, Kupka S, Kroeber S, Menzel D, Radsak MP, Rammensee HG, Salih HR: Cancer immunoediting by GITR (glucocorticoid-induced TNF-related protein) ligand in humans: NK cell/tumor cell interactions. FASEB J. 2007, 21: 2442-2454. 10.1096/fj.06-7724com.

    CAS  PubMed  Google Scholar 

  103. Rosenzweig M, Ponte J, Apostolou I, Doty D, Guild G, Slavonic M, Ponath P, Vaickus L: Development of TRX518, an aglycosyl humanized monoclonal antibody (Mab) agonist of huGITR [abstract]. J Clin Oncol. 2010, 28: e13028-

    Google Scholar 

  104. Kwon BS, Tan KB, Ni J, Oh KO, Lee ZH, Kim KK, Kim YJ, Wang S, Gentz R, Yu GL, Harrop J, Lyn SD, Silverman C, Porter TG, Truneh A, Young PR: A newly identified member of the tumor necrosis factor receptor superfamily with a wide tissue distribution and involvement in lymphocyte activation. J Biol Chem. 1997, 272: 14272-14276. 10.1074/jbc.272.22.14272.

    CAS  PubMed  Google Scholar 

  105. Pasero C, Speiser DE, Derré L, Olive D: The HVEM network: new directions in targeting novel costimulatory/co-inhibitory molecules for cancer therapy. Curr Opin Pharmacol. 2012, 12: 475-485.

    Google Scholar 

  106. Morel Y, Truneh A, Sweet RW, Olive D, Costello RT: The TNF superfamily members LIGHT and CD154 (CD40 ligand) costimulate induction of dendritic cell maturation and elicit specific CTL activity. J Immunol. 2001, 167: 2479-2486. 10.4049/jimmunol.167.5.2479.

    CAS  PubMed  Google Scholar 

  107. Cai G, Freeman GJ: The CD160, BTLA, LIGHT/HVEM pathway: a bidirectional switch regulating T-cell activation. Immunol Rev. 2009, 229: 244-258. 10.1111/j.1600-065X.2009.00783.x. 107

    CAS  PubMed  Google Scholar 

  108. Derré L, Rivals JP, Jandus C, Pastor S, Rimoldi D, Romero P, Michielin O, Olive D, Speiser DE: BTLA mediates inhibition of human tumor-specific CD8+ T cells that can be partially reversed by vaccination. J Clin Invest. 2010, 120: 157-167. 10.1172/JCI40070.

    PubMed  PubMed Central  Google Scholar 

  109. Fourcade J, Sun Z, Pagliano O, Guillaume P, Luescher IF, Sander C, Kirkwood JM, Olive D, Kuchroo V, Zarour HM: 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, 72: 887-896. 10.1158/0008-5472.CAN-11-2637.

    CAS  PubMed  PubMed Central  Google Scholar 

  110. Cheung KJ, Johnson NA, Affleck JG, Severson T, Steidl C, Ben- Neriah S, Schein J, Morin RD, Moore R, Shah SP, Qian H, Paul JE, Telenius A, Relander T, Lam W, Savage K, Connors JM, Brown C, Marra MA, Gascoyne RD, Horsman DE: Acquired TNFRSF14 mutations in follicular lymphoma are associated with worse prognosis. Cancer Res. 2010, 70: 9166-9174. 10.1158/0008-5472.CAN-10-2460.

    CAS  PubMed  Google Scholar 

  111. Park JJ, Anand S, Zhao Y, Matsumura Y, Sakoda Y, Kuramasu A, Strome SE, Chen L, Tamada K: Expression of anti-HVEM single-chain antibody on tumor cells induces tumor-specific immunity with long-term memory. Cancer Immunol Immunother. 2012, 61: 203-214. 10.1007/s00262-011-1101-8.

    CAS  PubMed  PubMed Central  Google Scholar 

  112. van Lier RA, Borst J, Vroom TM, Klein H, Van Mourik P, Zeijlemaker WP, Melief CJ: Tissue distribution and biochemical and functional properties of Tp55 (CD27), a novel T cell differentiation antigen. J Immunol. 1987, 139: 1589-1596.

    CAS  PubMed  Google Scholar 

  113. Hendriks J, Gravestein LA, Tesselaar K, van Lier RA, Schumacher TN, Borst J: CD27 is required for generation and long-term maintenance of T cell immunity. Nat Immunol. 2000, 1: 433-440. 10.1038/80877.

    CAS  PubMed  Google Scholar 

  114. Denoeud J, Moser M: Role of CD27/CD70 pathway of activation in immunity and tolerance. J Leukoc Biol. 2011, 89: 195-203. 10.1189/jlb.0610351.

    CAS  PubMed  Google Scholar 

  115. Croft M: The role of TNF superfamily members in T-cell function and diseases. Nat Rev Immunol. 2009, 9: 271-285. 10.1038/nri2526.

    CAS  PubMed  PubMed Central  Google Scholar 

  116. Ruprecht CR, Gattorno M, Ferlito F, Gregorio A, Martini A, Lanzavecchia A, Sallusto F: Coexpression of CD25 and CD27 identifies FoxP3+ regulatory T cells in inflamed synovia. J Exp Med. 2005, 201: 1793-1803. 10.1084/jem.20050085.

    CAS  PubMed  PubMed Central  Google Scholar 

  117. Coenen JJ, Koenen HJ, van Rijssen E, Hilbrands LB, Joosten I: Rapamycin, and not cyclosporin A, preserves the highly suppressive CD27+ subset of human CD4 + CD25+ regulatory T cells. Blood. 2006, 107: 1018-1023.

    CAS  PubMed  Google Scholar 

  118. Jung J, Choe J, Li L, Choi YS: Regulation of CD27 expression in the course of germinal center B cell differentiation: the pivotal role of IL-10. Eur J Immunol. 2000, 30: 2437-2443. 10.1002/1521-4141(2000)30:8<2437::AID-IMMU2437>3.0.CO;2-M.

    CAS  PubMed  Google Scholar 

  119. Oshima H, Nakano H, Nohara C, Kobata T, Nakajima A, Jenkins NA, Gilbert DJ, Copeland NG, Muto T, Yagita H, Okumura K: Characterization of murine CD70 by molecular cloning and mAb. Int Immunol. 1998, 10: 517-526. 10.1093/intimm/10.4.517.

    CAS  PubMed  Google Scholar 

  120. Peperzak V, Xiao Y, Veraar EA, Borst J: CD27 sustains survival of CTLs in virus-infected nonlymphoid tissue in mice by inducing autocrine IL-2 production. J Clin Invest. 2010, 120: 168-178. 10.1172/JCI40178.

    CAS  PubMed  PubMed Central  Google Scholar 

  121. Song DG, Ye Q, Poussin M, Harms GM, Figini M, Powell DJ: CD27 costimulation augments the survival and antitumor activity of redirected human T cells in vivo. Blood. 2012, 119: 696-706. 10.1182/blood-2011-03-344275.

    CAS  PubMed  Google Scholar 

  122. Kelly JM, Darcy PK, Markby JL, Godfrey DI, Takeda K, Yagita H, Smyth MJ: Induction of tumor-specific T cell memory by NK cell-mediated tumor rejection. Nat Immunol. 2002, 3: 83-90.

    CAS  PubMed  Google Scholar 

  123. Claus C, Riether C, Schürch C, Matter MS, Hilmenyuk T, Ochsenbein AF: CD27 signaling increases the frequency of regulatory T cells and promotes tumor growth. Cancer Res. 2012, 72: 3664-3676. 10.1158/0008-5472.CAN-11-2791.

    CAS  PubMed  Google Scholar 

  124. Schürch C, Riether C, Matter MS, Tzankov A, Ochsenbein AF: CD27 signaling on chronic myelogenous leukemia stem cells activates Wnt target genes and promotes disease progression. J Clin Invest. 2012, 122: 624-638. 10.1172/JCI45977.

    PubMed  PubMed Central  Google Scholar 

  125. Celldex Therapeutics initiates CDX-1127 phase 1 study in malignant solid tumors [press release].http://www.news-medical.net/news/20111108/Celldex-Therapeutics-initiates-CDX-1127-Phase-1-study-in-malignant-solid-tumors.aspx,

  126. He L-Z, Thomas L, Weidlick J, Vitale L, O’Neill T, Prostak N, Sundarapandiyan K, Marsh H, Yellin M, Davis TA, Keler T: Development of a human anti-CD27 antibody with efficacy in lymphoma and leukemia models by two distinct mechanisms [Abstract]. Blood. 2011, 118: abstr 2861-

    Google Scholar 

  127. Vanneman M, Dranoff G: Combining immunotherapy and targeted therapies in cancer treatment. Nature Rev Cancer. 2012, 12: 237-251. 10.1038/nrc3237.

    CAS  Google Scholar 

  128. Suntharalingam G, Perry MR, Ward S, Brett SJ, Castello-Cortes A, Brunner MD, Panoskaltsis N: Cytokine storm in a phase 1 trial of the anti-CD28 monoclonal antibody TGN1412. N Engl J Med. 2006, 355: 1018-1028. 10.1056/NEJMoa063842.

    CAS  PubMed  Google Scholar 

Download references

Acknowledgments

This study was partially supported by NIH grants R01CA56821, P01CA33049, and P01CA59350, D.A.S received support from the NIH Clinical Training for Scholar Grant K12 CA120121-01, NIH/NCI Immunology Training GrantT32 CA09149-30 and John D. Proctor Foundation: Margaret A. Cunningham Immune Mechanisms in Cancer Research Fellowship Award; Swim Across America; the Mr. William H. Goodwin and Mrs. Alice Goodwin and the Commonwealth Cancer Foundation for Research and the Experimental Therapeutics Center of Memorial Sloan-Kettering Cancer Center (to J.D.W.). Professional medical writing and editorial assistance was provided by Cailin Moira Wilke, PhD, at StemScientific, and was funded by Bristol-Myers Squibb.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Jedd D Wolchok.

Additional information

Competing interests

DS and DH declare no competing interest. JW discloses that he is a paid consultant for Medimmune and Bristol-Myers Squibb.

Authors’ contributions

DS, DH and JW contributed equally to the development of this manuscript. All authors read and approved the final manuscript.

David A Schaer, Daniel Hirschhorn-Cymerman contributed equally to this work.

Authors’ original submitted files for images

Below are the links to the authors’ original submitted files for images.

Authors’ original file for figure 1

Rights and permissions

Open Access This article is published under license to BioMed Central Ltd. This is an Open Access article is distributed under the terms of the Creative Commons Attribution License ( https://creativecommons.org/licenses/by/2.0 ), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly credited. The Creative Commons Public Domain Dedication waiver ( https://creativecommons.org/publicdomain/zero/1.0/ ) applies to the data made available in this article, unless otherwise stated.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Schaer, D.A., Hirschhorn-Cymerman, D. & Wolchok, J.D. Targeting tumor-necrosis factor receptor pathways for tumor immunotherapy. j. immunotherapy cancer 2, 7 (2014). https://doi.org/10.1186/2051-1426-2-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/2051-1426-2-7

Keywords