Twenty-five years ago, we reported that agonist anti-CD137 monoclonal antibodies eradicated transplanted mouse tumors because of enhanced CD8+ T-cell antitumor immunity. Mouse models indicated that anti-CD137 agonist antibodies synergized with various other therapies. In the clinic, the agonist antibody urelumab showed evidence for single-agent activity against melanoma and non-Hodgkin lymphoma but caused severe liver inflammation in a fraction of the patients. CD137's signaling domain is included in approved chimeric antigen receptors conferring persistence and efficacy. A new wave of CD137 agonists targeting tumors, mainly based on bispecific constructs, are in early-phase trials and are showing promising safety and clinical activity.

Significance:

CD137 (4-1BB) is a costimulatory receptor of T and natural killer lymphocytes whose activity can be exploited in cancer immunotherapy strategies as discovered 25 years ago. Following initial attempts that met unacceptable toxicity, new waves of constructs acting agonistically on CD137 are being developed in patients, offering signs of clinical and pharmacodynamic activity with tolerable safety profiles.

The group of Byoung Kwon genetically searched for molecules expressed by activated but not resting T lymphocytes (1). A cDNA encoding a transmembrane protein was named 4-1BB (2). Cloning of the mouse and human homologs permitted the generation of monoclonal antibodies (mAb) that eventually led to the discovery of the protein (3) and its designation as CD137, becoming a new member of the tumor necrosis factor receptor (TNFR) gene family, termed TNFRSF9. The antibodies were functionally able to enhance proliferation and cytokine production while mitigating cell death in T-lymphocyte cultures (3, 4). The group of Robert Mittler, based at Bristol Myers Squibb, was able to show that the artificial in vivo costimulation of CD8+ T cells with agonist anti-CD137 mAb was able to enhance CD8+ T-cell responses while attenuating humoral responses through a CD4 T-cell–dependent mechanism (5).

Twenty-five years ago, we reported that an infusion of anti-CD137 agonist mAbs was able to eradicate a number of established transplantable mouse tumors including those derived from the Ag104 and P815 cell lines (6). Antitumor effects in every case required the presence in the mouse of CD8+ T cells, while the requirements for natural killer (NK) and CD4 T cells were tumor model–dependent (6–8).

In the meantime, scientists at the Immunex company were able to clone a TNF family natural ligand for CD137, termed CD137L (4-1BBL, TNFSF9), which also exerted costimulatory effects on T lymphocytes as a trimeric molecule (9). In this regard, we showed in mouse models that murine tumor cell transfectants expressing CD137L enhanced CD8 antitumor immune responses toward themselves and against untransfected concomitant counterparts in a manner that was enhanced if the fellow costimulation ligand CD80 was cotransfected to stimulate CD28 (10). Such results were improved if instead of the CD137L moiety, anti-CD137 mAb single-chain variable fragments (scFv) encompassing a transmembrane domain were transfected to transplantable tumor cell lines (11). Importantly, such effects could be recapitulated against established tumors by intratumoral injections of an adenovirus encoding such engineered surface scFv antibodies (12) or 4-1BBL (13, 14). These experiments were an application of the previously described costimulation with adenovirally transferred 4-1BBL of antiviral CD8+ T-cell responses (15).

These discoveries were coincidental in time with reports that CTLA-4 blockade with antagonist antibodies was able to eradicate a particular transplantable colon cancer (16). In this regard, to the best of our knowledge, the therapeutic use of CD137 mAb opened up the field of agonist immunomodulatory mAbs (17). Figure 1 shows a timeline of the key milestones regarding these and subsequent preclinical and clinical developments.

Figure 1.

Timeline of key discoveries and development in CD137 (4-1BB) cancer immunotherapy. Key scientific milestones in the history of CD137-based immunotherapy since the discovery of the target. CAR, chimeric antigen receptor; TIL, tumor-infiltrating lymphocyte.

Figure 1.

Timeline of key discoveries and development in CD137 (4-1BB) cancer immunotherapy. Key scientific milestones in the history of CD137-based immunotherapy since the discovery of the target. CAR, chimeric antigen receptor; TIL, tumor-infiltrating lymphocyte.

Close modal

Of utmost importance is the fact that CD137 expression is not constitutive on T cells but is induced following T-cell receptor (TCR)/CD3 activation on antigen-primed T lymphocytes. It can also be expressed by other activated leukocytes including activated NK cells, where it is stimulatory (8, 18), B cells (19), and other myeloid leukocytes including dendritic cells (20–22), where its functions remain elusive for the most part. As a result of being inducible, it can be used as a marker to identify and select T cells recently involved in antigen recognition (23, 24). Of note, costimulation by CD28 (10) and CD137 itself (25) notably enhances the levels of CD137 expression, as does reportedly hypoxia (26). Following massive CD137 cross-linking, internalization of the moiety occurs to endosomal compartments from where it may keep signaling (27). Consequences of internalization of CD137 are underexplored and might be important to optimize stimulation schedules, because chronic stimulation may desensitize the pathway, therefore favoring intermittent exposure to achieve maximal effects.

CD137 belongs to the TNFR subfamily that is devoid of cytoplasmic death domains (28) and therefore does not directly engage with the caspase activation machinery. The molecule is composed of four extracellular cysteine-rich domains (CRD), a transmembrane region, and a 42-aa-long cytoplasmic domain bereft of any known intrinsic enzymatic activity. As in the case of all other TNFR members, the activity of CD137 relies on the interaction with members of a family of adapter proteins termed TRAFs (TNF receptor–associated factors; ref. 29). The first pieces of evidence for TRAF2 interaction with CD137 were based on a two-hybrid methodology (30) and coimmunoprecipitation (31). CD137 interaction with TRAF2 and TRAF1, as well as TRAF3 and TRAF5, has been reported (29–33). Indeed, using high-resolution proteomics, we have recently demonstrated the presence of those four adapters in CD137 immunoprecipitates. In addition to these, cIAP1 and cIAP2 and the LUBAC members HOIP and HOIL-1L were coprecipitated (J. Glez-Vaz; submitted for publication).

Costimulatory signaling via 4-1BB is largely contingent upon cross-linking by ligand at least to trimerize or form larger order of magnitude lattice-shaped structures (29). Following cross-linking, K63 polyubiquitination reactions take place (27) that originally were thought to involve the E3 RING domain of TRAF2. However, this catalytic domain reportedly is structurally unable to mediate ubiquitination reactions (34), and other E3 ubiquitin ligases must be involved including cIAPs, which have been shown to avidly associate with TRAF2 (35) also in the 4-1BB signalosome (J. Glez-Vaz; submitted for publication). Of note, a dominant-negative cIAP transgene in mice abrogated T-cell costimulation by 4-1BB mAbs (36), and we now have evidence that cIAPs are required for downstream signaling and for the antitumor effects (J. Glez-Vaz; submitted for publication). McPherson and colleagues (37) had previously shown that knockdown of cIAP2 in cIAP1 knockout T cells abrogated 4-1BB–dependent NF-kB activation, indicating that either cIAP1 or cIAP2 was required for NF-kB1 activation. These authors also showed that 4-1BB signaling causes degradation of TRAF3 leading to activation of the NF-kB2 pathway that was delayed with respect to activation of the classical NF-kB pathway.

The system seems to be highly controlled by deubiquitinases such as A20 and CYLD that constitutively associate with the 4-1BB signalosome complex in which various substrates are ubiquitinated including TRAF2 (38).

The biochemical function of TRAF1, which is prominently induced by NF-κB activation, remains elusive, but reportedly together with TRAF2, it recruits cIAPs with more affinity to the signaling complex (35). TRAF1 has also been functionally linked to T-cell apoptosis inhibition through Bim downregulation (39). Once early ubiquitination takes place, docking sites appear for TAB1/2–TAK1, and subsequently the IKK complex and various MAPKs become activated. K63 polyubiquitination of IKKγ (NEMO) is also probably a key event in signaling via NF-κB1 (40). In any case, TRAF1 seems to enhance classical NF-kB1 activation. Of note, TRAF1 itself is a strong transcriptional target of NF-kB, suggesting an interesting positive feedback mechanism in this signaling route as a result of the inducibility of TRAF1 expression (41).

Remarkably, CD137 also activates noncanonical NF-κB through elusive mechanisms that reportedly involve TRAF1 (37) in a yet poorly understood fashion. In this regard, the presence of TRAF3 in the 4-1BB signalosome suggests that active 4-1BB could sequester TRAF3, thereby preventing the targeting of NIK for proteasomal degradation through K48 polyubiquitination. Signaling via the alternative NF-κB pathway (NF-κB2) may be a differential feature of CD137 as compared with other costimulatory TNFR family members (42). This is important because most of these signaling mechanisms are shared by other T cell–expressed TNFR family members, and it remains to be seen what the essential qualitative or quantitative differences among them are.

There are soluble CD137 (sCD137) forms dependent on variants splicing out the transmembrane domain (43) or on protein shedding by metalloprotease cleavage (44). Soluble forms resulting from T-cell activation can competitively block CD137L to tone down costimulatory functions as a negative feedback mechanism (45). The expression of the CD137 ligand seems to be restricted to activated dendritic cells, B cells, and macrophages (46, 47). Reverse signaling via CD137L has been studied and seems to be important at regulating myeloid leukocyte biology (48). CD137 molecular physiology and signaling are summarized in Fig. 2.

Figure 2.

Schematic representation of CD137 molecular physiology. Stimulation of CD137 with cognate ligand (CD137L) or agonist antibodies is represented. On the left, the activation of NF-κB1 and MAPKs is illustrated in conjunction with K63 polyubiquitination reactions, as well as the regulation by deubiquitinases. On the right, the postulated induction mechanism of NF-κB2 is depicted. APC, antigen-presenting cell.

Figure 2.

Schematic representation of CD137 molecular physiology. Stimulation of CD137 with cognate ligand (CD137L) or agonist antibodies is represented. On the left, the activation of NF-κB1 and MAPKs is illustrated in conjunction with K63 polyubiquitination reactions, as well as the regulation by deubiquitinases. On the right, the postulated induction mechanism of NF-κB2 is depicted. APC, antigen-presenting cell.

Close modal

Almost since its discovery, CD137 was found to be involved in fostering a number of functions in T cells. Once ligated, it increases T-cell proliferation, enhances cytokine production (3), reduces lymphocyte cell death (39), enhances memory differentiation (49), and may reverse anergy/exhaustion (50). CD137 ligation triggers transcriptomic changes that underlie these phenotypic observations with additional layers of complexity because CD137-induced cytokines may potentially contribute to these phenotypic changes in an autocrine/paracrine manner. In addition to transcriptional effects, experimental evidence indicated that CD137 elicits epigenetic chromatin reprogramming that controls DNA methylation (51) and histone-3 methylation/acetylation changes (52). CD137 costimulation potently reprograms metabolism fostering mitochondrial mass and function (53, 54). The costimulation via CD137 is not restricted to TCRαβ T lymphocytes because it is also functionally expressed by TCRγδ counterparts (55, 56).

Although somewhat of a surprise, CD137 or CD137L deficiency in mice does not drive a severe immunodeficient phenotype because such mice are mostly normal with only relatively mild defects in the setting of more severe model viral infections such as influenza or lymphocytic choriomeningitis virus (57–59) for which CTL responses are critical to clear the infections. One of the reasons for these mild phenotypes is probably related to the overlapping function of other costimulatory TNFR molecules such as OX40, CD27, and GITR. In humans, a few cases suffering homozygous loss-of-function mutations of CD137 have been associated with severe Epstein–Barr virus infections including lymphoproliferative syndromes (60–62).

It comes as a surprise that the same agonist mAbs that were able to elicit tumor regressions also improve the outcomes of autoimmune disease models in mice driven by autoreactive CD4 T cells such as experimental autoimmune encephalomyelitis (63), collagen-induced arthritis (64, 65), or lupus (66). The reasons for this are far from clear but were traced to the fostering of activation-induced cell death of such pathogenic CD4+ T cells (63, 65). In contrast, agonist anti-CD137 mAbs were shown to exacerbate CD8-mediated diabetes in NOD mice (67, 68) and caused discrete levels of T-cell infiltrates in the liver causing mild periportal hepatitis in the treated mice (69, 70). Of note, the pathogenesis in these models is mainly mediated by CD8+ T cells.

CD4+ Foxp3+ regulatory T cells (Treg) also acquire CD137 expression that is very prominent on the Tregs present in the tumor microenvironment (TME; ref. 71). Conspicuously, the result of CD137 ligation on Tregs seems not to increase or be detrimental for their immunosuppressive functions but apparently may contribute to their expansion (72, 73). Interestingly, in mouse models, CD137 can be used as a target to deplete Tregs from the TME with properly engineered mAbs (74).

Although costimulation of T-cell responses by CD137 mAbs in general requires antigen–TCR engagement, the effect of CD137 mAb on memory T cells can be antigen-independent as shown by proliferation and activation of memory T cells in mice without exposure to a specific antigen (75). Because memory T cells accumulate in the liver and bone marrow, it is possible that this antigen-independent CD8+ T-cell activation may be partially responsible for CD137 mAb–mediated side effects in theses organs.

The inhibition of T-cell apoptosis is perhaps the most prominent and relevant effect of CD137 costimulation. This has been ascribed to the NF-κB activation of Bcl-xL and Bfl-1 (76), as well as to downregulation of Bim (39). The inducible nature of the expression of 4-1BB and 4-1BBL mediating this prominent antiapoptosis role prompted T.H. Watts to propose 4-1BB signaling as a signal-4 in T-cell activation to distinguish it from constitutively operative CD28 costimulation (77). It was also important to establish that 4-1BB costimulation is actually independent from CD28, as shown in CD28 knockout mice (78).

NK cells are also able to prominently express CD137 on their plasma membrane upon activation (8). Other investigators have reported that CD137 ligation on NK cells leads to costimulation of cytokine secretion without increasing cytolytic activity at the effector phase including antibody-dependent cellular cytotoxicity (79) but strengthening their cytolytic machinery and making them resistant to TGFβ inhibition (80).

CD137 putative functional expression on other cell lineages has been documented. On endothelial cells in the tumor microvasculature in which it is expressed in response to hypoxia, its ligation promotes T-cell extravasation via ICAM-1 and VCAM upregulation (81). Expression and ligation on adipocytes mediate subtle metabolic consequences (82) and a role for CD137–CD137L interactions has been reported in the progression of atherosclerosis (83).

From the early discoveries of the antitumor effects exerted by CD137 agonistic mAbs, multiple attempts to enhance efficacy upon combination with other modalities of cancer therapy have been made (84, 85). For instance, chemotherapy can enhance efficacy against transplantable tumor models (86–88). This could result from the immunomodulatory effects of chemotherapy or because of the generation of more tumor antigen cross-presentation from dying tumor cells (89). Notably, cross-priming by cDC1 of CD8+ T cells is needed for anti–4-1BB mAbs to work (90, 91). Radiotherapy also synergizes with 4-1BB, possibly through similar mechanisms (92–95). Even surgery in neoadjuvant 4-1BB treatment settings synergized to prevent metastatic relapses (96).

A variety of immunotherapy strategies potently synergize with 4-1BB stimulation to enhance antitumor immunity. Among the most prominently efficacious in mouse tumor models is perhaps the combination with anti–PD-1/PD-L1 (97–99) and local delivery of IL12 (14, 100–102). Other combinations have also been shown to be synergistic including the use of an anti–CTLA-4 mAb (103) and the codepletion of Tregs (104). As a mechanism of mutual synergy, it is worth mentioning that CD137 costimulation fosters IFNγ secretion from T cells, and this cytokine in turn is arguably the main inducer of PD-L1 expression in the TME (105). Experimental cancer vaccines have also been shown to synergize with anti-CD137 mAbs (106–110). Other immunostimulatory mAbs may also act in synergy with agonist anti-CD137 mAb, as has been observed when targeting LAG3 (50), OX40 (110, 111), and CD40 (112). Reported synergies of CD137 agonists in preclinical and clinical research are summarized in Fig. 3.

Figure 3.

Reported evidence for synergistic effects of CD137-based immunotherapy with other modality treatments. The represented synergistic combinations are indicated by arrowheads with the corresponding citations to the literature given in brackets. ADCC, antibody-dependent cellular cytotoxicity.

Figure 3.

Reported evidence for synergistic effects of CD137-based immunotherapy with other modality treatments. The represented synergistic combinations are indicated by arrowheads with the corresponding citations to the literature given in brackets. ADCC, antibody-dependent cellular cytotoxicity.

Close modal

All things considered, arguably the most efficacious synergy of CD137 costimulation is probably established with adoptive T-cell therapies (113). Evidence for synergy includes TCR-transgenic T cells (111) and ex vivo cultures of tumor-infiltrating T lymphocytes (114). With regard to tumor-infiltrating lymphocytes (TIL), anti-CD137 mAb also helps at the preinfusion stages to select those T cells with tumor reactivity and to enhance the yield of the cultures (115, 116). In this regard, CD137 costimulation has been successfully exploited to expand NK-cell cultures for infusion purposes in clinical trials (117, 118). This is consistent with observations in which the presence of 4-1BBL in artificial antigen-presenting cells had a dramatic effect to induce T-cell expansion ex vivo in culture (119).

Adoptive T-cell therapy using the gene transfer of chimeric antigen receptors (CAR) against CD19 and BCMA has revolutionized the treatment of B-cell malignances (120). The group of Carl June introduced the 4-1BB signaling domain as part of the intracellular sequence of CARs, and showed that in CD8+ T cells it leads to longer persistence upon infusion as compared with those encompassing the CD28 signaling domain (121). Alternatively, the group of Michel Sadelain showed that 4-1BBL + CD80 gene transfer to the T cells cotransferred with CD28ζ CARs also gained much efficacy against prostate cancer and lymphoma xenografted to mice (122).

In the clinic, at least three 4-1BB containing CARs have received approval for commercialization in Western countries due to unprecedented efficacy against refractory cases of acute lymphoblastic leukemia, B-cell lymphomas, and multiple myeloma (120, 123). Reportedly, in CD4 T cells, the costimulation domain of ICOS surpasses the efficacy of CD137 (124), but in CD8+ T cells, CD137 seems more effective and perhaps leads to less severe cytokine release syndrome in the clinic as compared with CD28-containing CARs (125, 126). Recent efficacy evidence has been reported for allogeneic NK cells transduced with CARs following expansion in culture driven by cytokines and CD137 ligation. Such CARs expressed in NK cells also encompass the CD137 cytoplasmic domain for costimulation in addition to CD3ζ (127).

Chimeric membrane molecules other than conventional CARs can be envisaged using synthetic biology to convey CD137 costimulation. Among these, an extracellular FAS/intracellular 4-1BB chimera has shown remarkable potential in mice (128), both because of conferring 4-1BB costimulation and being a dominant-negative decoy ligand for FASL (129).

In summary, ways to confer CD137 costimulation are powerful allies of adoptive T-cell therapy. This could be especially important as adoptive T-cell therapy faces the formidable challenge of providing efficacy against solid malignances. In that regard, CD137 could be crucial due to its ability to prevent or reverse anergy (50, 130) in addition to promoting survival and longer persistence. Enhanced mitochondrial/metabolic fitness (131) and the NF-κB2 pathway (132) are probably key differential superiority elements in favor of CD137. Perhaps as a consequence of these mechanisms, CD137 tonic signaling from the CARs seems to be beneficial rather than detrimental for CAR T performance and has been associated with a unique transcriptional profile (130, 133). These reasons are probably behind the superiority of 4-1BB–based CARs over CD28 ones in terms of persistence and safety.

The Bristol Myers Squibb company led the initial development of a fully human IgG4 clinical-grade agonist anti-CD137 mAb (eventually termed urelumab; ref. 134). The mAb was safe in non-human primates but as was noted later, the affinity of urelumab for macaque CD137 was considerably lower as compared with the human protein. A conventional 3 + 3 dose-escalation phase I clinical trial as monotherapy was conducted in which dose-limiting toxicities were not encountered. Evidence of objective activity in cases of melanoma was observed, as well as evidence for pharmacodynamic activity, although transient elevations of transaminases were reported in some of these dose-scalation patients (135).

Phase II clinical trials were undertaken using every 3 week doses of 5 mg/kg, and it was observed that around 20% of patients developed grade ≥ 3 abnormalities in liver function tests, with most of them experiencing milder elevations in circulating transaminases (135). Indeed, two fatalities occurred. These serious drug-related adverse events, although apparently idiosyncratic and limited to a subset of patients, put the urelumab program on hold.

Liver toxicity could be modeled in mice, in which polyclonal CD8+ T-cell accumulations in the liver parenchyma were found following repeated treatment with agonist anti-mouse CD137 (69, 136). Fc receptors (137) and proinflammatory cytokines such as IFNγ and IL27 were also shown to be critical, perhaps involving myeloid cells in the liver (138). The exact mechanism of liver toxicity remains elusive, although baseline expression of 4-1BB has been substantiated on memory T cells in the bone marrow and in the liver (139). Intriguingly, liver inflammation was abrogated in GITR knockout mice (139). It is tempting to speculate that once the pathway is stimulated, it results in more 4-1BB expression on activated CD8+ T cells as a vicious circle.

In a parallel effort, Pfizer developed utomilumab, a fully human anti-CD137 mAb of the IgG2 isotype (140). In this case, the agonist activity on the receptor is moderate and is fully cross-reactive with macaque CD137. Following uneventful toxicology studies, dose-escalation clinical trials were performed with excellent tolerability and no dose-limiting toxicities (141). However, clinical activity as a single agent was modest, although at least two cases of Merkel carcinoma of the skin responded to treatment (141). A recent report confirmed marginal clinical activity of utomilumab for immune checkpoint inhibitor–experienced melanoma and non–small cell lung cancer patients with evidence for frequent induction (46.3%) of antidrug antibodies (142).

Of important note, urelumab is an IgG4 that keeps avidity for FcR-I, whereas utomilumab is an IgG2 with limited binding to Fc receptors (143). Conceivably, the agonist activity of these antibodies in vivo is contingent on Fc receptor–mediated cross-linking. However, in the case of urelumab, some level of costimulation is observed even without an apparent need for cross-linking.

Urelumab reentered clinical trials to document the safe doses that could be tolerable in terms of liver side effects, and a phase I study declared 0.1 mg/kg, 0.3 mg/kg, and a flat dose of 8 mg as safe (144). Skin reactions and neutropenia were also mitigated by dose reduction. However, at these doses, objective responses were restricted to 13% of 31 heavily pretreated non-Hodgkin B-lymphoma cases.

As mentioned above, preclinical research in mouse models demonstrated that combinations of CD137 agonists and PD-(L)1 checkpoint inhibitors were synergistic. In light of such results, the combinations of urelumab + nivolumab and utomilumab + pembrolizumab have been tested in pilot single-arm trials. In the case of urelumab, patients with melanoma responded to the combination [overall response rate (ORR) = 39%], but evidence of the contribution of low-dose urelumab in these patients with checkpoint inhibitor–naïve melanoma was not conclusive (145). In the case of utomilumab + pembrolizumab, several interesting objective responses were documented, including cases of complete response in one patient with anaplastic thyroid cancer and in another with small cell lung cancer (146). In a recent report, utomilumab in combination with rituximab demonstrated promising clinical activity (ORR = 21.2%) in patients with relapsed/refractory follicular lymphoma and other CD20+ non-Hodgkin lymphomas (147).

In a very interesting combination approach, the group of Elizabeth Jaffee has tested urelumab (8 mg doses) in combination with nivolumab and a GVAX vaccine in a neoadjuvant + adjuvant randomized clinical trial for resectable pancreatic cancer. Urelumab was found to enhance tumor CD8+ T-cell infiltration in the resected tumor tissue, with promising clinical outcomes of relapse-free survival (148).

However, the liver safety issues and lack of evidence of consistent clinical monotherapy activity deprioritized development of urelumab and utomilumab in such a manner that currently there are no active clinical trials with these agents. Nevertheless, several important conclusions can be drawn from these early-phase clinical experiences: (i) These agonist antibodies are pharmacodynamically active and can achieve certain clinical activity in monotherapy; (ii) liver inflammation is a key limiting factor to be overcome for this class of agonist agents; (iii) other less serious safety issues may appear in terms of leukopenia, as previously reported in mice (135, 149), and skin toxicity that are probably on-target; (iv) use of CD137 agonists and PD-(L)1 blockade is most likely feasible in combination regimens with no evidence for serious accumulative toxicity, and there is a rationale for therapeutic synergy of these combined regimens.

Biotechnology for therapeutic mAbs and recombinant protein constructs has advanced a great deal over the last 25 years. Several strategies have been followed to construct agents with multiple specificities and activities built into a single molecule (150). The main objective here is to attain meaningful agonist activity on CD137 while making it tolerable for the patient when given either as monotherapy or in combination with other treatments. As previously discussed, the main feature to exert agonistic activity on CD137 is to induce cross-linking by multimeric ligands or by means of attaching the ligand to a solid phase such as a juxtaposed plasma membrane. Hence, designing and making constructs that would only or selectively cause cross-linking in the TME and/or in tumor-draining lymph nodes is the goal of this strategy. Recent evidence supports the superiority of cross-linking CD137 from the plasma membrane of a cell that is presenting cognate antigen or is engaging CD3–TCR by other means such as a T-cell engager bispecific antibody (42). The superiority of the so-called “in cis” costimulation over “in trans” costimulation, when TCR–CD3 engagement occurs from different cells, speaks to some degree of receptor cross-talk between CD3 and CD137. The molecular underpinnings of such cross-talk are being clarified. Having said this, even if less molecularly active, intense trans costimulation might be as efficacious for cancer treatment.

Three main strategies are being followed: (i) bispecific antibodies that bind to tumor selectively expressed surface antigens; (ii) bispecific antibodies that bind to immune receptors or checkpoint ligands expressed in the TME; and (iii) conditional antibodies that become active only in the TME. Figure 4 summarizes the different alternatives that are being developed, and several of these agents have already entered early-phase clinical development. Ongoing and recently completed clinical trials with this new wave of CD137 agonists are included in Table 1.

Figure 4.

New wave of CD137 agonist agents. Schematic representation of the different antibody or protein constructs undergoing late preclinical (red) or clinical (green) development. The structure of the agents is schematized, and the postulated site of action in the TME is represented. APC, antigen-presenting cell; CAF, cancer associated fibroblast.

Figure 4.

New wave of CD137 agonist agents. Schematic representation of the different antibody or protein constructs undergoing late preclinical (red) or clinical (green) development. The structure of the agents is schematized, and the postulated site of action in the TME is represented. APC, antigen-presenting cell; CAF, cancer associated fibroblast.

Close modal
Table 1.

New wave of CD137 (4-1BB) agonists in the clinic

New wave of CD137 (4-1BB) agonists in the clinic
New wave of CD137 (4-1BB) agonists in the clinic

The formats of the multispecific compounds are variable and are represented in Fig. 4 for each agent. Conventional antibody chemistry still seems to be the most popular way, involving variations of antibody engineering both on constant regions and on the avidity of antigen-binding sites. However, other chemistries and constructs are used including nanobodies and calin- and collagen-based proteins. Both antibodies and trimeric forms of the natural ligand are under development. Details regarding these important aspects may account for different pharmacodynamics and tissue penetration features whose technical description is beyond this review (151).

Targeting CD137 Costimulation to Tumor-Expressed Molecules

This strategy is being pursued with several targets such as HER2 (152), EGFR (153, 154), 5T4 (42), PSMA (155), Glypican-3, Nectin-4 (156), Mesothelin (157), and B7-H3 (158), which are preferentially overexpressed by tumor cells. The principle of selective cross-linking and T-cell costimulation works with all these constructs in T-cell cocultures with the proper target-expressing cell lines. For hematologic malignancies, agents targeting CD137 agonists have been redirected to CD30 (159) and to CD19 (160). In these cases, the activity of the compounds is contingent on the expression of the different molecules on malignant cells. This feature is frequently disease-specific and variable even within a given malignancy. The key models to preclinically test these compounds are CD34- or peripheral blood mononuclear cell–humanized mice (161, 162) and human CD137 knock-in mice (163).

In the clinic, the only available clinical results are with HER2 × 4-1BB bispecific antibody (PRS-343; refs. 152, 164). The treatment as monotherapy is remarkably safe, signs of CD8+ T-cell functional stimulation have been found, and preliminary clinical activity has been reported (ORR = 40% at doses over 8 mg/kg) in HER2 cancer patients with trastuzumab-refractory disease (164).

Multispecific Constructs Stimulating CD137 upon Binding to FAP in the Tumor Stroma

Fibroblast-activated protein (FAP) is a transmembrane protein selectively and intensely expressed on tumor-associated fibroblasts in many solid malignancies. Targeting compounds to FAP could permit selective homing to tumors in mice and humans. Cross-linking of 4-1BB at this level provides significant costimulation to T cells and shows efficacy in mouse models (165). Two compounds are being developed in clinical trials based on these concepts: a FAP-binding antibody × trimeric CD137L (165, 166) developed by Roche and a more classic bispecific FAP–CD137 antibody that is being developed by Boehringer Ingelheim. In these cases, costimulation is provided in trans with respect to tumor antigens, but at least with T-cell engagers, these agents provide meaningful costimulation in vivo (165, 167).

The early-phase CD137L–FAP developed by Roche (RG7826 or RO7122290) has shown an excellent safety profile in monotherapy or when combined with PD-L1 blockade using atezolizumab. It is pharmacodynamically active, as it induces peripheral blood T-cell activation and proliferation as well as accumulation of activated CD8+ T cells in the TME (168, 169). There are signs of modest clinical activity in monotherapy and more promising evidence in combination with atezolizumab, but because responding patients were not previously refractory to anti–PD-(L)1 therapy, further evidence for clinical activity is needed. An ongoing clinical trial is testing the combination of a T-cell engager cotargeting CEACAM5 (CEA) and CD3 (cibisatamab) together with RG7826 based on interesting preclinical data in an attempt to provide both signal 1 and signal 2, thereby functionally mimicking CARs (165, 167).

Bispecific Antigens Binding CD137 and Other Immune Receptors

Given the preclinical synergy of CD137 agonists with PD-(L)1 blockade, various PD-L1 × CD137 combinations are being developed. The idea is to use PD-L1 both to target and agonistically cross-link CD137 in the TME while simultaneously blocking PD-L1 binding to PD-1 (170–173). The mouse surrogate of this idea shows evidence for immunologic and therapeutic activities (174–176), albeit the optimal doses for blockade and agonist activity are likely to be different. Several formats for such bispecific antibodies have been generated (Fig. 4).

In the clinic, results are presently available only for a molecule developed by Genmab (GEN1046). This agent has shown clear clinical signs of T-cell activation in patients. Regarding safety, around 20% of patients developed controllable abnormalities in liver function tests. Importantly, there is evidence for clinical activity in monotherapy against tumors previously having been determined to be refractory to anti–PD-(L)1 therapy, as well as clear signs of pharmacodynamic activity (170).

For hematologic malignancies, a trimeric CD137L binding to CD19 has been developed by Roche (RG6076 or RO7227166) to treat refractory B-cell malignancies. Preliminary reported evidence speaks of safety and frequent objective responses upon combination with a CD20 T-cell engager (177) in patients pretreated with obinutuzumab (an afucosylated anti-CD20 mAb) to deplete endogenous B cells and cytoreduce as much as possible the number of remaining malignant cells. A bispecific antibody binding to CD30 and CD137 is also being developed for hematologic malignancies, chiefly including cases of refractory Hodgkin lymphoma (159).

Interestingly, bispecific constructs can also exploit awakening the function of two immune receptors involved in triggering antitumor immunity. In this regard, it is worth considering the efforts using a CD40 × CD137 bispecific (GEN1042; ref. 178) already in the clinic (179). This bispecific antibody seems to mediate CD40 activation/maturation of dendritic cells while selectively cross-linking CD137 on T cells in contact with such dendritic cells in a bidirectional fashion (Fig. 4). This concept of cross-linking from dendritic cells that are cross-presenting tumor antigens is of much interest and presumably can also be achieved by PD-L1 × CD137 bispecifics because there is expression of PD-L1 on dendritic cells (C. Luri-Rey; manuscript in preparation).

Conditional CD137 Agonists

To attain selective activity in the TME and tumor-draining lymph nodes, probody technology has been applied. This consists of masked antibodies in which the antigen-binding site is not functional because of a masking cognate peptide. Such a peptide is tethered to the light chain of the antibody by a protease-sensitive linker that is cleaved by proteases selectively present in tumors. This strategy in mouse models with an anti-CD137 probody increases liver safety and synergizes with other treatments including PD-1 blockade and adoptive T-cell therapy (180).

Benefiting from the fact that the interstitial fluid of tumors is very rich in ATP concentrations, a conditional antibody binding and cross-linking CD137 only in ATP-rich media has been generated and has shown remarkable safety and efficacy in human CD137 knock-in mouse models (181, 182). Such a compound is progressing toward clinical development.

It is becoming increasingly clear that the costimulatory activity of CD137 is of great interest to enhance the effects of tumor immunotherapy, and the maximal effect is very likely to be found deploying the new agents in synergistic combination therapy schemes. The obvious combination with anti–PD-(L)1 therapy is ongoing in the clinic, but combinatorial approaches with adoptive T-cell therapy, cytokines, and chemotherapy should be clinically explored. Even in the case of PD-L1 × CD137 bispecific constructs, the combination with an additional antibody blocking PD-1 therapy is clearly worth considering given the fact that the optimal doses for CD137 stimulation might be lower than those for complete PD-L1 blockade. We have to bear in mind that to permit cross-linking of the two targets by a bispecific construct, optimal concentrations must be found, because an excess of the compound leads to saturation of each target with no opportunity for cross-linking (17). This is consistent with clinical findings in the sense that bimodal dose–response curves are observed for immune pharmacodynamic effects and efficacy.

Much of the success will depend on the discovery and validation of predictive biomarkers that are to be thoroughly investigated in clinical pretreatment and on-treatment samples. From the pharmacodynamic point of view, early evidence of T-cell activation is likely to be predictive of disease outcomes. Recent evidence indicates that CD137, either soluble or membrane bound, provides an accurate reflection of functional CD137 ligation (25). In peripheral blood, concentrations of IFNγ and CXCL10 as well as proliferating Ki-67+ CD8+ T cells are useful pharmacodynamic biomarkers in addition to T-cell infiltration and activation in on-treatment tumor biopsies (168, 170). Quantitatively significant expression of the targets for the bispecific compounds in the TME is likely to be a crucial condition to attain meaningful therapeutic effects.

The most relevant scientific questions in the field are: (i) to precisely decipher the molecular and cellular requirements to attain antitumor efficacy; (ii) to understand the role of 4-1BB costimulation in T-cell memory, dysfunction, and exhaustion; (iii) to understand and overcome CD137-related liver inflammation; (iv) to preclinically and clinically define the most synergistic partner treatments for combinations; (v) to extend the benefit of CD137-based immunotherapy to nonmalignant diseases; and (vi) to design and implement late-phase clinical trials to document benefit over standard of care.

Overall, CD137-based approaches hold much hope for cancer immunotherapy beyond their successful application in CAR T cells. We certainly expect that consistent efficacy in patients will be reached soon. Clinical science and pharmaceutical development are often unpredictable, but the magnitude of the current academic and industrial effort in CD137-based immunotherapy and the preliminary reports on the new wave of CD137 agonist compounds speak to a bright future, which will be harvesting the fruits of 25 years of continuous translational research efforts.

I. Melero reports grants and personal fees from Bristol Myers Squibb, Genmab, and Roche and personal fees from Alligator, F-Star, Merus, Pieris, and Boston Therapeutics during the conduct of the study, as well as grants and personal fees from Bristol Myers Squibb, Roche, AstraZeneca, Genmab, and PharmaMar and personal fees from F-Star, Gossamer, Alligator, Hotspot, Biolinerx, Bright Peak, Third Rock, Boston Therapeutics, Pieris, Servier, and Pierre Fabre outside the submitted work. M.F. Sanmamed reports grants from Bristol Myers Squibb and Roche during the conduct of the study, as well as grants and personal fees from Roche and Bristol Myers Squibb, and personal fees from Numab outside the submitted work. No disclosures were reported by the other authors.

I. Melero has been supported by grants PID2020-112892RB funded by MICIN/AEI/10.13039/501100011033 and SAF2017-83267-C2-1-R funded by MICIN/AEI/10.13039/501100011033 and by FEDER “Una manera de hacer Europa,” the Fundación de la Asociación Española Contra el Cáncer (AECC; HR21–00083), the Fundación La Caixa and Fundación BBVA, the Instituto de Salud Carlos III (PI20/00002 and PI19/01128), cofinanced by the Fondos FEDER “A way to make Europe” and Joint Translational Call for Proposals 2015 (JTC 2015), TRANSCAN456 2 (code TRS-2016-00000371), and the Gobierno de Navarra Proyecto LINTERNA (reference 0011-1411-2020-000075). M.F. Sanmamed has received a Miguel Servet I (MS17/00196) contract from the Instituto de Salud Carlos III cofinanced by the Fondo Social Europeo “Investing in your future” and a grant from Instituto de Salud Carlos III, Fondo de Investigacion Sanitaria (PI19/00668). J. Wang has received a sponsored research grant from RootPath Genomics in the past 12 months and is funded by NIH grants R21AI163924 and R37CA273333. L. Chen has received sponsored research grants from NextCure, DynamiCure, and Normunity in the past 12 months and is funded by NIH grants CA196530 and CA016359 and a United Technologies Corporation endowment in cancer research.

1.
Kwon
BS
,
Kim
GS
,
Prystowsky
MB
,
Lancki
DW
,
Sabath
DE
,
Pan
JL
, et al
.
Isolation and initial characterization of multiple species of T-lymphocyte subset cDNA clones
.
Proc Natl Acad Sci U S A
1987
;
84
:
2896
900
.
2.
Kwon
BS
,
Weissman
M
.
cDNA sequences of two inducible T-cell genes
.
Proc Natl Acad Sci U S A
1989
;
86
:
1963
7
.
3.
Pollok
KE
,
Kim
YJ
,
Zhou
Z
,
Hurtado
J
,
Kim
KK
,
Pickard
RT
, et al
.
Inducible T cell antigen 4-1BB: analysis of expression and function
.
J Immunol
1993
;
150
:
771
81
.
4.
Hurtado
JC
,
Kim
YJ
,
Kwon
BS
.
Signals through 4–1BB are costimulatory to previously activated splenic T cells and inhibit activation-induced cell death
.
J Immunol
1997
;
158
:
2600
9
.
5.
Mittler
RS
,
Bailey
TS
,
Klussman
K
,
Trailsmith
MD
,
Hoffmann
MK
.
Anti–4-1BB monoclonal antibodies abrogate T cell–dependent humoral immune responses in vivo through the induction of helper T cell anergy
.
J Exp Med
1999
;
190
:
1535
40
.
6.
Melero
I
,
Shuford
WW
,
Newby
SA
,
Aruffo
A
,
Ledbetter
JA
,
Hellström
KE
, et al
.
Monoclonal antibodies against the 4–1BB T-cell activation molecule eradicate established tumors
.
Nat Med
1997
;
3
:
682
5
.
7.
Miller
RE
,
Jones
J
,
Le
T
,
Whitmore
J
,
Boiani
N
,
Gliniak
B
, et al
.
4–1BB-specific monoclonal antibody promotes the generation of tumor-specific immune responses by direct activation of CD8 T cells in a CD40-dependent manner
.
J Immunol
2002
;
169
:
1792
800
.
8.
Melero
I
,
Johnston
JV
,
Shufford
WW
,
Mittler
RS
,
Chen
L
.
NK1.1 cells express 4–1BB (CDw137) costimulatory molecule and are required for tumor immunity elicited by anti-4-1BB monoclonal antibodies
.
Cell Immunol
1998
;
190
:
167
72
.
9.
Goodwin
RG
,
Din
WS
,
Davis-Smith
T
,
Anderson
DM
,
Gimpel
SD
,
Sato
TA
, et al
.
Molecular cloning of a ligand for the inducible T cell gene 4-1BB: a member of an emerging family of cytokines with homology to tumor necrosis factor
.
Eur J Immunol
1993
;
23
:
2631
41
.
10.
Melero
I
,
Bach
N
,
Hellström
KE
,
Aruffo
A
,
Mittler
RS
,
Chen
L
.
Amplification of tumor immunity by gene transfer of the co-stimulatory 4-1BB ligand: synergy with the CD28 co-stimulatory pathway
.
Eur J Immunol
1998
;
28
:
1116
21
.
11.
Ye
Z
,
Hellström
I
,
Hayden-Ledbetter
M
,
Dahlin
A
,
Ledbetter
JA
,
Hellström
KE
.
Gene therapy for cancer using single-chain Fv fragments specific for 4-1BB
.
Nat Med
2002
;
8
:
343
8
.
12.
Yang
Y
,
Yang
S
,
Ye
Z
,
Jaffar
J
,
Zhou
Y
,
Cutter
E
, et al
.
Tumor cells expressing anti-CD137 scFv induce a tumor-destructive environment
.
Cancer Res
2007
;
67
:
2339
44
.
13.
Martinet
O
,
Divino
CM
,
Zang
Y
,
Gan
Y
,
Mandeli
J
,
Thung
S
, et al
.
T cell activation with systemic agonistic antibody versus local 4-1BB ligand gene delivery combined with interleukin-12 eradicate liver metastases of breast cancer
.
Gene Ther
2002
;
9
:
786
92
.
14.
Xu
DP
,
Sauter
BV
,
Huang
TG
,
Meseck
M
,
Woo
SL
,
Chen
SH
.
The systemic administration of Ig-4-1BB ligand in combination with IL-12 gene transfer eradicates hepatic colon carcinoma
.
Gene Ther
2005
;
12
:
1526
33
.
15.
Bukczynski
J
,
Wen
T
,
Ellefsen
K
,
Gauldie
J
,
Watts
TH
.
Costimulatory ligand 4-1BBL (CD137L) as an efficient adjuvant for human antiviral cytotoxic T cell responses
.
Proc Natl Acad Sci U S A
2004
;
101
:
1291
6
.
16.
Leach
DR
,
Krummel
MF
,
Allison
JP
.
Enhancement of antitumor immunity by CTLA-4 blockade
.
Science
1996
;
271
:
1734
-
6
.
17.
Mayes
PA
,
Hance
KW
,
Hoos
A
.
The promise and challenges of immune agonist antibody development in cancer
.
Nat Rev Drug Discov
2018
;
17
:
509
27
.
18.
Kohrt
HE
,
Houot
R
,
Goldstein
MJ
,
Weiskopf
K
,
Alizadeh
AA
,
Brody
J
, et al
.
CD137 stimulation enhances the antilymphoma activity of anti-CD20 antibodies
.
Blood
2011
;
117
:
2423
32
.
19.
Zhang
X
,
Voskens
CJ
,
Sallin
M
,
Maniar
A
,
Montes
CL
,
Zhang
Y
, et al
.
CD137 promotes proliferation and survival of human B cells
.
J Immunol
2010
;
184
:
787
95
.
20.
Wilcox
RA
,
Chapoval
AI
,
Gorski
KS
,
Otsuji
M
,
Shin
T
,
Flies
DB
, et al
.
Cutting edge: expression of functional CD137 receptor by dendritic cells
.
J Immunol
2002
;
168
:
4262
7
.
21.
Heinisch
IV
,
Bizer
C
,
Volgger
W
,
Simon
HU
.
Functional CD137 receptors are expressed by eosinophils from patients with IgE-mediated allergic responses but not by eosinophils from patients with non-IgE-mediated eosinophilic disorders
.
J Allergy Clin Immunol
2001
;
108
:
21
8
.
22.
Heinisch
IV
,
Daigle
I
,
Knöpfli
B
,
Simon
HU
.
CD137 activation abrogates granulocyte-macrophage colony-stimulating factor-mediated anti-apoptosis in neutrophils
.
Eur J Immunol
2000
;
30
:
3441
6
.
23.
Gros
A
,
Robbins
PF
,
Yao
X
,
Li
YF
,
Turcotte
S
,
Tran
E
, et al
.
PD-1 identifies the patient-specific CD8(+) tumor-reactive repertoire infiltrating human tumors
.
J Clin Invest
2014
;
124
:
2246
59
.
24.
Parkhurst
M
,
Gros
A
,
Pasetto
A
,
Prickett
T
,
Crystal
JS
,
Robbins
P
, et al
.
Isolation of T-cell receptors specifically reactive with mutated tumor-associated antigens from tumor-infiltrating lymphocytes based on CD137 expression
.
Clin Cancer Res
2017
;
23
:
2491
505
.
25.
Glez-Vaz
J
,
Azpilikueta
A
,
Olivera
I
,
Cirella
A
,
Teijeira
A
,
Ochoa
MC
, et al
.
Soluble CD137 as a dynamic biomarker to monitor agonist CD137 immunotherapies
.
J Immunother Cancer
2022
;
10
:
e003532
.
26.
Palazon
A
,
Martinez-Forero
I
,
Teijeira
A
,
Morales-Kastresana
A
,
Alfaro
C
,
Sanmamed
MF
, et al
.
The HIF-1alpha hypoxia response in tumor-infiltrating T lymphocytes induces functional CD137 (4-1BB) for immunotherapy
.
Cancer Discov
2012
;
2
:
608
23
.
27.
Martinez-Forero
I
,
Azpilikueta
A
,
Bolanos-Mateo
E
,
Nistal-Villan
E
,
Palazon
A
,
Teijeira
A
, et al
.
T cell costimulation with anti-CD137 monoclonal antibodies is mediated by K63-polyubiquitin-dependent signals from endosomes
.
J Immunol
2013
;
190
:
6694
706
.
28.
Croft
M
.
Co-stimulatory members of the TNFR family: keys to effective T-cell immunity?
Nat Rev Immunol
2003
;
3
:
609
20
.
29.
Zapata
JM
,
Lefebvre
S
,
Reed
JC
.
Targeting TRAfs for therapeutic intervention
.
Adv Exp Med Biol
2007
;
597
:
188
201
.
30.
Arch
RH
,
Thompson
CB
.
4–1BB and Ox40 are members of a tumor necrosis factor (TNF)-nerve growth factor receptor subfamily that bind TNF receptor-associated factors and activate nuclear factor kappaB
.
Mol Cell Biol
1998
;
18
:
558
65
.
31.
Saoulli
K
,
Lee
SY
,
Cannons
JL
,
Yeh
WC
,
Santana
A
,
Goldstein
MD
, et al
.
CD28-independent, TRAF2-dependent costimulation of resting t cells by 4-1BB ligand
.
J Immunol
1998
;
187
:
1849
62
.
32.
Jang
IK
,
Lee
ZH
,
Kim
YJ
,
Kim
SH
,
Kwon
BS
.
Human 4–1BB (CD137) signals are mediated by TRAF2 and activate nuclear factor-kappa B
.
Biochem Biophys Res Commun
1998
;
242
:
613
20
.
33.
Sabbagh
L
,
Andreeva
D
,
Laramee
GD
,
Oussa
NA
,
Lew
D
,
Bisson
N
, et al
.
Leukocyte-specific protein 1 links TNF receptor-associated factor 1 to survival signaling downstream of 4–1BB in T cells
.
J Leukoc Biol
2013
;
93
:
713
21
.
34.
Yin
Q
,
Lamothe
B
,
Darnay
BG
,
Wu
H
.
Structural basis for the lack of E2 interaction in the RING domain of TRAF2
.
Biochemistry
2009
;
48
:
10558
67
.
35.
Zheng
C
,
Kabaleeswaran
V
,
Wang
Y
,
Cheng
G
,
Wu
H
.
Crystal structures of the TRAF2: cIAP2 and the TRAF1: TRAF2: cIAP2 complexes: affinity, specificity, and regulation
.
Mol Cell
2010
;
38
:
101
13
.
36.
Giardino Torchia
ML
,
Munitic
I
,
Castro
E
,
Herz
J
,
McGavern
DB
,
Ashwell
JD
.
c-IAP ubiquitin protein ligase activity is required for 4-1BB signaling and CD8(+) memory T-cell survival
.
Eur J Immunol
2015
;
45
:
2672
82
.
37.
McPherson
AJ
,
Snell
LM
,
Mak
TW
,
Watts
TH
.
Opposing roles for TRAF1 in the alternative versus classical NF-kappaB pathway in T cells
.
J Biol Chem
2012
;
287
:
23010
9
.
38.
Azpilikueta
A
,
Bolanos
E
,
Lang
V
,
Labiano
S
,
Aznar
MA
,
Etxeberria
I
, et al
.
Deubiquitinases A20 and CYLD modulate costimulatory signaling via CD137 (4-1BB)
.
Oncoimmunology
2017
;
7
:
e1368605
.
39.
Sabbagh
L
,
Pulle
G
,
Liu
Y
,
Tsitsikov
EN
,
Watts
TH
.
ERK-dependent Bim modulation downstream of the 4-1BB-TRAF1 signaling axis is a critical mediator of CD8 T cell survival in vivo
.
J Immunol
2008
;
180
:
8093
101
.
40.
Zhang
Q
,
Lenardo
MJ
,
Baltimore
D
.
30 Years of NF-kappaB: a blossoming of relevance to human pathobiology
.
Cell
2017
;
168
:
37
57
.
41.
Wang
CY
,
Mayo
MW
,
Korneluk
RG
,
Goeddel
DV
,
Baldwin
AS Jr
.
NF-kappaB antiapoptosis: induction of TRAF1 and TRAF2 and c-IAP1 and c-IAP2 to suppress caspase-8 activation
.
Science
1998
;
281
:
1680
3
.
42.
Otano
I
,
Azpilikueta
A
,
Glez-Vaz
J
,
Alvarez
M
,
Medina-Echeverz
J
,
Cortes-Dominguez
I
, et al
.
CD137 (4-1BB) costimulation of CD8(+) T cells is more potent when provided in cis than in trans with respect to CD3-TCR stimulation
.
Nat Commun
2021
;
12
:
7296
.
43.
Setareh
M
,
Schwarz
H
,
Lotz
M
.
A mRNA variant encoding a soluble form of 4-1BB, a member of the murine NGF/TNF receptor family
.
Gene
1995
;
164
:
311
5
.
44.
Seidel
J
,
Leitzke
S
,
Ahrens
B
,
Sperrhacke
M
,
Bhakdi
S
,
Reiss
K
.
Role of ADAM10 and ADAM17 in regulating CD137 function
.
Int J Mol Sci
2021
;
22
:
2730
.
45.
Labiano
S
,
Palazon
A
,
Bolanos
E
,
Azpilikueta
A
,
Sanchez-Paulete
AR
,
Morales-Kastresana
A
, et al
.
Hypoxia-induced soluble CD137 in malignant cells blocks CD137L-costimulation as an immune escape mechanism
.
Oncoimmunology
2016
;
5
:
e1062967
.
46.
Kwon
B
.
Is CD137 ligand (CD137L) signaling a fine tuner of immune responses?
Immune Netw
2015
;
15
:
121
4
.
47.
Futagawa
T
,
Akiba
H
,
Kodama
T
,
Takeda
K
,
Hosoda
Y
,
Yagita
H
, et al
.
Expression and function of 4-1BB and 4-1BB ligand on murine dendritic cells
.
Int Immunol
2002
;
14
:
275
86
.
48.
Shao
Z
,
Schwarz
H
.
CD137 ligand, a member of the tumor necrosis factor family, regulates immune responses via reverse signal transduction
.
J Leukoc Biol
2011
;
89
:
21
9
.
49.
Zhou
AC
,
Wagar
LE
,
Wortzman
ME
,
Watts
TH
.
Intrinsic 4–1BB signals are indispensable for the establishment of an influenza-specific tissue-resident memory CD8 T-cell population in the lung
.
Mucosal Immunol
2017
;
10
:
1294
309
.
50.
Williams
JB
,
Horton
BL
,
Zheng
Y
,
Duan
Y
,
Powell
JD
,
Gajewski
TF
.
The EGR2 targets LAG-3 and 4-1BB describe and regulate dysfunctional antigen-specific CD8+ T cells in the tumor microenvironment
.
J Exp Med
2017
;
214
:
381
400
.
51.
Aznar
MA
,
Labiano
S
,
Diaz-Lagares
A
,
Molina
C
,
Garasa
S
,
Azpilikueta
A
, et al
.
CD137 (4-1BB) costimulation modifies DNA methylation in CD8(+) T cell-relevant genes
.
Cancer Immunol Res
2018
;
6
:
69
78
.
52.
Ford
BR
,
Vignali
PDA
,
Rittenhouse
NL
,
Scharping
NE
,
Peralta
R
,
Lontos
K
, et al
.
Tumor microenvironmental signals reshape chromatin landscapes to limit the functional potential of exhausted T cells
.
Sci Immunol
2022
;
7
:
eabj9123
.
53.
Teijeira
A
,
Labiano
S
,
Garasa
S
,
Etxeberria
I
,
Santamaria
E
,
Rouzaut
A
, et al
.
Mitochondrial morphological and functional reprogramming following CD137 (4-1BB) costimulation
.
Cancer Immunol Res
2018
;
6
:
798
811
.
54.
Menk
AV
,
Scharping
NE
,
Rivadeneira
DB
,
Calderon
MJ
,
Watson
MJ
,
Dunstane
D
, et al
.
4–1BB costimulation induces T cell mitochondrial function and biogenesis enabling cancer immunotherapeutic responses
.
J Exp Med
2018
;
215
:
1091
100
.
55.
Lee
SJ
,
Kim
YH
,
Hwang
SH
,
Kim
YI
,
Han
IS
,
Vinay
DS
, et al
.
4-1BB signal stimulates the activation, expansion, and effector functions of gammadelta T cells in mice and humans
.
Eur J Immunol
2013
;
43
:
1839
48
.
56.
Cho
HW
,
Kim
SY
,
Sohn
DH
,
Lee
MJ
,
Park
MY
,
Sohn
HJ
, et al
.
Triple costimulation via CD80, 4-1BB, and CD83 ligand elicits the long-term growth of Vgamma9Vdelta2 T cells in low levels of IL-2
.
J Leukoc Biol
2016
;
99
:
521
9
.
57.
Lin
GHY
,
Sedgmen
BJ
,
Moraes
TJ
,
Snell
LM
,
Topham
DJ
,
Watts
TH
.
Endogenous 4-1BB ligand plays a critical role in protection from influenza-induced disease
.
J Immunol
2009
;
182
:
934
47
.
58.
DeBenedette
MA
,
Wen
T
,
Bachmann
MF
,
Ohashi
PS
,
Barber
BH
,
Stocking
KL
, et al
.
Analysis of 4-1BB ligand (4-1BBL)-deficient mice and of mice lacking both 4-1BBL and CD28 reveals a role for 4-1BBL in skin allograft rejection and in the cytotoxic T cell response to influenza virus
.
J Immunol
1999
;
163
:
4833
41
.
59.
Tan
JT
,
Whitmire
JK
,
Ahmed
R
,
Pearson
TC
,
Larsen
CP
.
4–1BB ligand, a member of the TNF family, is important for the generation of antiviral CD8 T cell responses
.
J Immunol
1999
;
163
:
4859
68
.
60.
Rodriguez
R
,
Fournier
B
,
Cordeiro
DJ
,
Winter
S
,
Izawa
K
,
Martin
E
, et al
.
Concomitant PIK3CD and TNFRSF9 deficiencies cause chronic active Epstein-Barr virus infection of T cells
.
J Exp Med
2019
;
216
:
2800
18
.
61.
Somekh
I
,
Thian
M
,
Medgyesi
D
,
Gülez
N
,
Magg
T
,
Duque
AG
, et al
.
CD137 deficiency causes immune dysregulation with predisposition to lymphomagenesis
.
Blood
2019
;
134
:
1510
6
.
62.
Alosaimi
MF
,
Hoenig
M
,
Jaber
F
,
Platt
CD
,
Jones
J
,
Wallace
J
, et al
.
Immunodeficiency and EBV-induced lymphoproliferation caused by 4-1BB deficiency
.
J Allergy Clin Immunol
2019
;
144
:
574
83
.
63.
Sun
Y
,
Lin
X
,
Chen
HM
,
Wu
Q
,
Subudhi
SK
,
Chen
L
, et al
.
Administration of agonistic anti-4-1BB monoclonal antibody leads to the amelioration of experimental autoimmune encephalomyelitis
.
J Immunol
2002
;
168
:
1457
65
.
64.
Foell
JL
,
Diez-Mendiondo
BI
,
Diez
OH
,
Holzer
U
,
Ruck
P
,
Bapat
AS
, et al
.
Engagement of the CD137 (4-1BB) costimulatory molecule inhibits and reverses the autoimmune process in collagen-induced arthritis and establishes lasting disease resistance
.
Immunology
2004
;
113
:
89
98
.
65.
Seo
SK
,
Choi
JH
,
Kim
YH
,
Kang
WJ
,
Park
HY
,
Suh
JH
, et al
.
4-1BB-mediated immunotherapy of rheumatoid arthritis
.
Nat Med
2004
;
10
:
1088
94
.
66.
Foell
J
,
McCausland
M
,
Burch
J
,
Corriazzi
N
,
Yan
XJ
,
Suwyn
C
, et al
.
CD137-mediated T cell co-stimulation terminates existing autoimmune disease in SLE-prone NZB/NZW F1 mice
.
Ann N Y Acad Sci
2003
;
987
:
230
5
.
67.
Forsberg
MH
,
Ciecko
AE
,
Bednar
KJ
,
Itoh
A
,
Kachapati
K
,
Ridgway
WM
, et al
.
CD137 plays both pathogenic and protective roles in type 1 diabetes development in NOD mice
.
J Immunol
2017
;
198
:
3857
68
.
68.
Sytwu
HK
,
Lin
WD
,
Roffler
SR
,
Hung
JT
,
Sung
HS
,
Wang
CH
, et al
.
Anti-4-1BB-based immunotherapy for autoimmune diabetes: lessons from a transgenic non-obese diabetic (NOD) model
.
J Autoimmun
2003
;
21
:
247
54
.
69.
Dubrot
J
,
Milheiro
F
,
Alfaro
C
,
Palazon
A
,
Martinez-Forero
I
,
Perez-Gracia
JL
, et al
.
Treatment with anti-CD137 mAbs causes intense accumulations of liver T cells without selective antitumor immunotherapeutic effects in this organ
.
Cancer Immunol Immunother
2010
;
59
:
1223
33
.
70.
Wang
J
,
Zhao
W
,
Cheng
L
,
Guo
M
,
Li
D
,
Li
X
, et al
.
CD137-mediated pathogenesis from chronic hepatitis to hepatocellular carcinoma in hepatitis B virus-transgenic mice
.
J Immunol
2010
;
185
:
7654
62
.
71.
Freeman
ZT
,
Nirschl
TR
,
Hovelson
DH
,
Johnston
RJ
,
Engelhardt
JJ
,
Selby
MJ
, et al
.
A conserved intratumoral regulatory T cell signature identifies 4-1BB as a pan-cancer target
.
J Clin Invest
2020
;
130
:
1405
16
.
72.
Zheng
G
,
Wang
B
,
Chen
A
.
The 4-1BB costimulation augments the proliferation of CD4+CD25+ regulatory T cells
.
J Immunol
2004
;
173
:
2428
34
.
73.
So
T
,
Lee
SW
,
Croft
M
.
Immune regulation and control of regulatory T cells by OX40 and 4-1BB
.
Cytokine Growth Factor Rev
2008
;
19
:
253
62
.
74.
Buchan
SL
,
Dou
L
,
Remer
M
,
Booth
SG
,
Dunn
SN
,
Lai
C
, et al
.
Antibodies to costimulatory receptor 4-1BB enhance anti-tumor immunity via T regulatory cell depletion and promotion of CD8 T cell effector function
.
Immunity
2018
;
49
:
958
70
.
75.
Zhu
Y
,
Zhu
G
,
Luo
L
,
Flies
AS
,
Chen
L
.
CD137 stimulation delivers an antigen-independent growth signal for T lymphocytes with memory phenotype
.
Blood
2007
;
109
:
4882
9
.
76.
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
8
.
77.
Chang
YH
,
Wang
KC
,
Chu
KL
,
Clouthier
DL
,
Tran
AT
,
Torres Perez
MS
, et al
.
Dichotomous expression of TNF superfamily ligands on antigen-presenting cells controls post-priming anti-viral CD4(+) T cell immunity
.
Immunity
2017
;
47
:
943
58
.
78.
DeBenedette
MA
,
Shahinian
A
,
Mak
TW
,
Watts
TH
.
Costimulation of CD28- T lymphocytes by 4-1BB ligand
.
J Immunol
1997
;
158
:
551
9
.
79.
Ochoa
MC
,
Perez-Ruiz
E
,
Minute
L
,
Onate
C
,
Perez
G
,
Rodriguez
I
, et al
.
Daratumumab in combination with urelumab to potentiate anti-myeloma activity in lymphocyte-deficient mice reconstituted with human NK cells
.
Oncoimmunology
2019
;
8
:
1599636
.
80.
Cabo
M
,
Santana-Hernandez
S
,
Costa-Garcia
M
,
Rea
A
,
Lozano-Rodriguez
R
,
Ataya
M
, et al
.
CD137 costimulation counteracts TGFbeta inhibition of NK-cell antitumor function
.
Cancer Immunol Res
2021
;
9
:
1476
90
.
81.
Palazon
A
,
Teijeira
A
,
Martinez-Forero
I
,
Hervas-Stubbs
S
,
Roncal
C
,
Penuelas
I
, et al
.
Agonist anti-CD137 mAb act on tumor endothelial cells to enhance recruitment of activated T lymphocytes
.
Cancer Res
2011
;
71
:
801
11
.
82.
Tu
TH
,
Kim
CS
,
Goto
T
,
Kawada
T
,
Kim
BS
,
Yu
R
.
4-1BB/4-1BBL interaction promotes obesity-induced adipose inflammation by triggering bidirectional inflammatory signaling in adipocytes/macrophages
.
Mediators Inflamm
2012
;
2012
:
972629
.
83.
Jeon
HJ
,
Choi
JH
,
Jung
IH
,
Park
JG
,
Lee
MR
,
Lee
MN
, et al
.
CD137 (4-1BB) deficiency reduces atherosclerosis in hyperlipidemic mice
.
Circulation
2010
;
121
:
1124
33
.
84.
Perez-Gracia
JL
,
Labiano
S
,
Rodriguez-Ruiz
ME
,
Sanmamed
MF
,
Melero
I
.
Orchestrating immune check-point blockade for cancer immunotherapy in combinations
.
Curr Opin Immunol
2014
;
27
:
89
97
.
85.
Melero
I
,
Berman
DM
,
Aznar
MA
,
Korman
AJ
,
Perez Gracia
JL
,
Haanen
J
.
Evolving synergistic combinations of targeted immunotherapies to combat cancer
.
Nat Rev Cancer
2015
;
15
:
457
72
.
86.
Kim
YH
,
Choi
BK
,
Oh
HS
,
Kang
WJ
,
Mittler
RS
,
Kwon
BS
.
Mechanisms involved in synergistic anticancer effects of anti-4-1BB and cyclophosphamide therapy
.
Mol Cancer Ther
2009
;
8
:
469
78
.
87.
Kim
YH
,
Choi
BK
,
Kim
KH
,
Kang
SW
,
Kwon
BS
.
Combination therapy with cisplatin and anti-4-1BB: synergistic anticancer effects and amelioration of cisplatin-induced nephrotoxicity
.
Cancer Res
2008
;
68
:
7264
9
.
88.
Ju
SA
,
Cheon
SH
,
Park
SM
,
Tam
NQ
,
Kim
YM
,
An
WG
, et al
.
Eradication of established renal cell carcinoma by a combination of 5-fluorouracil and anti-4-1BB monoclonal antibody in mice
.
Int J Cancer
2008
;
122
:
2784
90
.
89.
Salas-Benito
D
,
Perez-Gracia
JL
,
Ponz-Sarvise
M
,
Rodriguez-Ruiz
ME
,
Martinez-Forero
I
,
Castanon
E
, et al
.
Paradigms on immunotherapy combinations with chemotherapy
.
Cancer Discov
2021
;
11
:
1353
67
.
90.
Sanchez-Paulete
AR
,
Cueto
FJ
,
Martinez-Lopez
M
,
Labiano
S
,
Morales-Kastresana
A
,
Rodriguez-Ruiz
ME
, et al
.
Cancer immunotherapy with immunomodulatory anti-CD137 and anti–PD-1 monoclonal antibodies requires BATF3-dependent dendritic cells
.
Cancer Discov
2016
;
6
:
71
9
.
91.
Teijeira
A
,
Garasa
S
,
Luri-Rey
C
,
Cd
A
,
Gato
M
,
Molina
C
, et al
.
Depletion of conventional type-1 dendritic cells in established tumors suppresses immunotherapy efficacy
.
Cancer Res
2022
;
82
:
4373
85
.
92.
Rodriguez-Ruiz
ME
,
Rodriguez
I
,
Garasa
S
,
Barbes
B
,
Solorzano
JL
,
Perez-Gracia
JL
, et al
.
Abscopal effects of radiotherapy are enhanced by combined immunostimulatory mAbs and are dependent on CD8 T cells and crosspriming
.
Cancer Res
2016
;
76
:
5994
6005
.
93.
Shi
W
,
Siemann
DW
.
Augmented antitumor effects of radiation therapy by 4-1BB antibody (BMS-469492) treatment
.
Anticancer Res
2006
;
26
:
3445
53
.
94.
Verbrugge
I
,
Hagekyriakou
J
,
Sharp
LL
,
Galli
M
,
West
A
,
McLaughlin
NM
, et al
.
Radiotherapy increases the permissiveness of established mammary tumors to rejection by immunomodulatory antibodies
.
Cancer Res
2012
;
72
:
3163
74
.
95.
Belcaid
Z
,
Phallen
JA
,
Zeng
J
,
See
AP
,
Mathios
D
,
Gottschalk
C
, et al
.
Focal radiation therapy combined with 4-1BB activation and CTLA-4 blockade yields long-term survival and a protective antigen-specific memory response in a murine glioma model
.
PLoS One
2014
;
9
:
e101764
.
96.
Liu
J
,
Blake
SJ
,
Yong
MC
,
Harjunpaa
H
,
Ngiow
SF
,
Takeda
K
, et al
.
Improved efficacy of neoadjuvant compared to adjuvant immunotherapy to eradicate metastatic disease
.
Cancer Discov
2016
;
6
:
1382
99
.
97.
Azpilikueta
A
,
Agorreta
J
,
Labiano
S
,
Perez-Gracia
JL
,
Sanchez-Paulete
AR
,
Aznar
MA
, et al
.
Successful immunotherapy against a transplantable mouse squamous lung carcinoma with anti–PD-1 and anti-CD137 monoclonal antibodies
.
J Thorac Oncol
2016
;
11
:
524
36
.
98.
Hirano
F
,
Kaneko
K
,
Tamura
H
,
Dong
H
,
Wang
S
,
Ichikawa
M
, et al
.
Blockade of B7-H1 and PD-1 by monoclonal antibodies potentiates cancer therapeutic immunity
.
Cancer Res
2005
;
65
:
1089
96
.
99.
Wei
H
,
Zhao
L
,
Hellstrom
I
,
Hellstrom
KE
,
Guo
Y
.
Dual targeting of CD137 co-stimulatory and PD-1 co-inhibitory molecules for ovarian cancer immunotherapy
.
Oncoimmunology
2014
;
3
:
e28248
.
100.
Quetglas
JI
,
Dubrot
J
,
Bezunartea
J
,
Sanmamed
MF
,
Hervas-Stubbs
S
,
Smerdou
C
, et al
.
Immunotherapeutic synergy between anti-CD137 mAb and intratumoral administration of a cytopathic Semliki Forest virus encoding IL-12
.
Mol Ther
2012
;
20
:
1664
75
.
101.
Chen
SH
,
Pham-Nguyen
KB
,
Martinet
O
,
Huang
Y
,
Yang
W
,
Thung
SN
, et al
.
Rejection of disseminated metastases of colon carcinoma by synergism of IL-12 gene therapy and 4-1BB costimulation
.
Mol Ther
2000
;
2
:
39
46
.
102.
Martinet
O
,
Ermekova
V
,
Qiao
JQ
,
Sauter
B
,
Mandeli
J
,
Chen
L
, et al
.
Immunomodulatory gene therapy with interleukin 12 and 4-1BB ligand: long- term remission of liver metastases in a mouse model
.
J Natl Cancer Inst
2000
;
92
:
931
6
.
103.
Kocak
E
,
Lute
K
,
Chang
X
,
May
KF
Jr
,
Exten
KR
,
Zhang
H
, et al
.
Combination therapy with anti-CTL antigen-4 and anti-4-1BB antibodies enhances cancer immunity and reduces autoimmunity
.
Cancer Res
2006
;
66
:
7276
84
.
104.
Liu
J
,
Blake
SJ
,
Harjunpaa
H
,
Fairfax
KA
,
Yong
MC
,
Allen
S
, et al
.
Assessing immune-related adverse events of efficacious combination immunotherapies in preclinical models of cancer
.
Cancer Res
2016
;
76
:
5288
301
.
105.
Chen
L
,
Han
X
.
Anti–PD-1/PD-L1 therapy of human cancer: past, present, and future
.
J Clin Invest
2015
;
125
:
3384
91
.
106.
Wilcox
RA
,
Flies
DB
,
Zhu
G
,
Johnson
AJ
,
Tamada
K
,
Chapoval
AI
, et al
.
Provision of antigen and CD137 signaling breaks immunological ignorance, promoting regression of poorly immunogenic tumors
.
J Clin Invest
2002
;
109
:
651
9
.
107.
Bartkowiak
T
,
Singh
S
,
Yang
G
,
Galvan
G
,
Haria
D
,
Ai
M
, et al
.
Unique potential of 4-1BB agonist antibody to promote durable regression of HPV+ tumors when combined with an E6/E7 peptide vaccine
.
Proc Natl Acad Sci U S A
2015
;
112
:
E5290
9
.
108.
Ito
F
,
Li
Q
,
Shreiner
AB
,
Okuyama
R
,
Jure-Kunkel
MN
,
Teitz-Tennenbaum
S
, et al
.
Anti-CD137 monoclonal antibody administration augments the antitumor efficacy of dendritic cell-based vaccines
.
Cancer Res
2004
;
64
:
8411
9
.
109.
Tirapu
I
,
Arina
A
,
Mazzolini
G
,
Duarte
M
,
Alfaro
C
,
Feijoo
E
, et al
.
Improving efficacy of interleukin-12-transfected dendritic cells injected into murine colon cancer with anti-CD137 monoclonal antibodies and alloantigens
.
Int J Cancer
2004
;
110
:
51
60
.
110.
Cuadros
C
,
Dominguez
AL
,
Lollini
PL
,
Croft
M
,
Mittler
RS
,
Borgstrom
P
, et al
.
Vaccination with dendritic cells pulsed with apoptotic tumors in combination with anti-OX40 and anti-4-1BB monoclonal antibodies induces T cell-mediated protective immunity in Her-2/neu transgenic mice
.
Int J Cancer
2005
;
116
:
934
43
.
111.
Morales-Kastresana
A
,
Sanmamed
MF
,
Rodriguez
I
,
Palazon
A
,
Martinez-Forero
I
,
Labiano
S
, et al
.
Combined immunostimulatory monoclonal antibodies extend survival in an aggressive transgenic hepatocellular carcinoma mouse model
.
Clin Cancer Res
2013
;
19
:
6151
62
.
112.
Uno
T
,
Takeda
K
,
Kojima
Y
,
Yoshizawa
H
,
Akiba
H
,
Mittler
RS
, et al
.
Eradication of established tumors in mice by a combination antibody-based therapy
.
Nat Med
2006
;
12
:
693
8
.
113.
Weigelin
B
,
Bolanos
E
,
Rodriguez-Ruiz
ME
,
Martinez-Forero
I
,
Friedl
P
,
Melero
I
.
Anti-CD137 monoclonal antibodies and adoptive T cell therapy: a perfect marriage?
Cancer Immunol Immunother
2016
;
65
:
493
7
.
114.
Hernandez-Chacon
JA
,
Li
Y
,
Wu
RC
,
Bernatchez
C
,
Wang
Y
,
Weber
JS
, et al
.
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
50
.
115.
Tavera
RJ
,
Forget
MA
,
Kim
YU
,
Sakellariou-Thompson
D
,
Creasy
CA
,
Bhatta
A
, et al
.
Utilizing T-cell activation signals 1, 2, and 3 for tumor-infiltrating lymphocytes (TIL) expansion: the advantage over the sole use of interleukin-2 in cutaneous and uveal melanoma
.
J Immunother
2018
;
41
:
399
405
.
116.
Chacon
JA
,
Wu
RC
,
Sukhumalchandra
P
,
Molldrem
JJ
,
Sarnaik
A
,
Pilon-Thomas
S
, et al
.
Co-stimulation through 4-1BB/CD137 improves the expansion and function of CD8(+) melanoma tumor-infiltrating lymphocytes for adoptive T-cell therapy
.
PLoS One
2013
;
8
:
e60031
.
117.
Gurney
M
,
Kundu
S
,
Pandey
S
,
O'Dwyer
M
.
Feeder cells at the interface of natural killer cell activation, expansion and gene editing
.
Front Immunol
2022
;
13
:
802906
.
118.
Li
X
,
He
C
,
Liu
C
,
Ma
J
,
Ma
P
,
Cui
H
, et al
.
Expansion of NK cells from PBMCs using immobilized 4-1BBL and interleukin-21
.
Int J Oncol
2015
;
47
:
335
42
.
119.
Maus
MV
,
Thomas
AK
,
Leonard
DGB
,
Allman
D
,
Addya
K
,
Schlienger
K
, et al
.
Ex vivo expansion of polyclonal and antigen-specific cytotoxic T lymphocytes by artificial APCs expressing ligands for the T-cell receptor, CD28 and 4-1BB
.
Nat Biotechnol
2002
;
20
:
143
8
.
120.
Boyiadzis
MM
,
Dhodapkar
MV
,
Brentjens
RJ
,
Kochenderfer
JN
,
Neelapu
SS
,
Maus
MV
, et al
.
Chimeric antigen receptor (CAR) T therapies for the treatment of hematologic malignancies: clinical perspective and significance
.
J Immunother Cancer
2018
;
6
:
137
.
121.
Carpenito
C
,
Milone
MC
,
Hassan
R
,
Simonet
JC
,
Lakhal
M
,
Suhoski
MM
, et al
.
Control of large, established tumor xenografts with genetically retargeted human T cells containing CD28 and CD137 domains
.
Proc Natl Acad Sci U S A
2009
;
106
:
3360
5
.
122.
Stephan
MT
,
Ponomarev
V
,
Brentjens
RJ
,
Chang
AH
,
Dobrenkov
KV
,
Heller
G
, et al
.
T cell-encoded CD80 and 4-1BBL induce auto- and transcostimulation, resulting in potent tumor rejection
.
Nat Med
2007
;
13
:
1440
9
.
123.
Munshi
NC
,
Anderson
LD
Jr.
,
Shah
N
,
Madduri
D
,
Berdeja
J
,
Lonial
S
, et al
.
Idecabtagene vicleucel in relapsed and refractory multiple myeloma
.
N Engl J Med
2021
;
384
:
705
16
.
124.
Guedan
S
,
Posey
AD
Jr
,
Shaw
C
,
Wing
A
,
Da
T
,
Patel
PR
, et al
.
Enhancing CAR T cell persistence through ICOS and 4-1BB costimulation
.
JCI Insight
2018
;
3
:
e96976
.
125.
Cappell
KM
,
Kochenderfer
JN
.
A comparison of chimeric antigen receptors containing CD28 versus 4-1BB costimulatory domains
.
Nat Rev Clin Oncol
2021
;
18
:
715
27
.
126.
Ying
Z
,
He
T
,
Wang
X
,
Zheng
W
,
Lin
N
,
Tu
M
, et al
.
Parallel comparison of 4-1BB or CD28 Co-stimulated CD19-targeted CAR-T cells for B cell non-Hodgkin's lymphoma
.
Mol Ther Oncolytics
2019
;
15
:
60
8
.
127.
Liu
E
,
Marin
D
,
Banerjee
P
,
Macapinlac
HA
,
Thompson
P
,
Basar
R
, et al
.
Use of CAR-transduced natural killer cells in CD19-positive lymphoid tumors
.
N Engl J Med
2020
;
382
:
545
53
.
128.
Oda
SK
,
Anderson
KG
,
Ravikumar
P
,
Bonson
P
,
Garcia
NM
,
Jenkins
CM
, et al
.
A Fas-4-1BB fusion protein converts a death to a pro-survival signal and enhances T cell therapy
.
J Exp Med
2020
;
217
:
e20191166
.
129.
Melero
I
,
Berraondo
P
.
4-1BB (CD137) in anticancer chimeras
.
J Exp Med
2020
;
217
:
e20201562
.
130.
Long
AH
,
Haso
WM
,
Shern
JF
,
Wanhainen
KM
,
Murgai
M
,
Ingaramo
M
, et al
.
4-1BB costimulation ameliorates T cell exhaustion induced by tonic signaling of chimeric antigen receptors
.
Nat Med
2015
;
21
:
581
90
.
131.
Kawalekar
OU
,
O'Connor
RS
,
Fraietta
JA
,
Guo
L
,
McGettigan
SE
,
Posey
AD
Jr
, et al
.
Distinct signaling of coreceptors regulates specific metabolism pathways and impacts memory development in CAR T cells
.
Immunity
2016
;
44
:
380
90
.
132.
Philipson
BI
,
O'Connor
RS
,
May
MJ
,
June
CH
,
Albelda
SM
,
Milone
MC
.
4-1BB costimulation promotes CAR T cell survival through noncanonical NF-kB signalling
.
Sci Signal
2020
;
13
:
eaay8248
.
133.
Singh
N
,
Frey
NV
,
Engels
B
,
Barrett
DM
,
Shestova
O
,
Ravikumar
P
, et al
.
Antigen-independent activation enhances the efficacy of 4-1BB-costimulated CD22 CAR T cells
.
Nat Med
2021
;
27
:
842
50
.
134.
Ascierto
PA
,
Simeone
E
,
Sznol
M
,
Fu
YX
,
Melero
I
.
Clinical experiences with anti-CD137 and anti–PD-1 therapeutic antibodies
.
Semin Oncol
2010
;
37
:
508
16
.
135.
Segal
NH
,
Logan
TF
,
Hodi
FS
,
McDermott
D
,
Melero
I
,
Hamid
O
, et al
.
Results from an integrated safety analysis of urelumab, an agonist anti-CD137 monoclonal antibody
.
Clin Cancer Res
2017
;
23
:
1929
36
.
136.
Niu
L
,
Strahotin
S
,
Hewes
B
,
Zhang
B
,
Zhang
Y
,
Archer
D
, et al
.
Cytokine-mediated disruption of lymphocyte trafficking, hemopoiesis, and induction of lymphopenia, anemia, and thrombocytopenia in anti-CD137-treated mice
.
J Immunol
2007
;
178
:
4194
213
.
137.
Qi
X
,
Li
F
,
Wu
Y
,
Cheng
C
,
Han
P
,
Wang
J
, et al
.
Optimization of 4–1BB antibody for cancer immunotherapy by balancing agonistic strength with FcgammaR affinity
.
Nat Commun
2019
;
10
:
2141
.
138.
Bartkowiak
T
,
Jaiswal
AR
,
Ager
CR
,
Chin
R
,
Chen
CH
,
Budhani
P
, et al
.
Activation of 4-1BB on liver myeloid cells triggers hepatitis via an interleukin-27-dependent pathway
.
Clin Cancer Res
2018
;
24
:
1138
51
.
139.
Lin
GH
,
Snell
LM
,
Wortzman
ME
,
Clouthier
DL
,
Watts
TH
.
GITR-dependent regulation of 4-1BB expression: implications for T cell memory and anti-4-1BB-induced pathology
.
J Immunol
2013
;
190
:
4627
39
.
140.
Fisher
TS
,
Kamperschroer
C
,
Oliphant
T
,
Love
VA
,
Lira
PD
,
Doyonnas
R
, et al
.
Targeting of 4-1BB by monoclonal antibody PF-05082566 enhances T-cell function and promotes anti-tumor activity
.
Cancer Immunol Immunother
2012
;
61
:
1721
33
.
141.
Segal
NH
,
He
AR
,
Doi
T
,
Levy
R
,
Bhatia
S
,
Pishvaian
MJ
, et al
.
Phase I study of single-agent utomilumab (PF-05082566), a 4-1BB/CD137 agonist, in patients with advanced cancer
.
Clin Cancer Res
2018
;
24
:
1816
23
.
142.
Hong
DS
,
Gopal
AK
,
Shoushtari
AN
,
Patel
SP
,
He
AR
,
Doi
T
, et al
.
Utomilumab in patients with immune checkpoint inhibitor-refractory melanoma and non-small-cell lung cancer
.
Front Immunol
2022
;
13
:
897991
.
143.
Vonderheide
RH
,
Glennie
MJ
.
Agonistic CD40 antibodies and cancer therapy
.
Clin Cancer Res
2013
;
19
:
1035
43
.
144.
Timmerman
J
,
Herbaux
C
,
Ribrag
V
,
Zelenetz
AD
,
Houot
R
,
Neelapu
SS
, et al
.
Urelumab alone or in combination with rituximab in patients with relapsed or refractory B-cell lymphoma
.
Am J Hematol
2020
;
95
:
510
20
.
145.
Massarelli
E
,
Segal
NH
,
Ribrag
V
,
Melero
I
,
Gangadhar
TC
,
Urba
W
, et al
.
Clinical safety and efficacy assessment of the CD137 agonist urelumab alone and in combination with nivolumab in patients with hematologic and solid tumor malignancies
.
J Immunother Cancer
2016
;
4
Suppl 1
:
O7
.
Abstract nr
239
.
146.
Tolcher
AW
,
Sznol
M
,
Hu-Lieskovan
S
,
Papadopoulos
KP
,
Patnaik
A
,
Rasco
DW
, et al
.
Phase Ib study of utomilumab (PF-05082566), a 4-1BB/CD137 agonist, in combination with pembrolizumab (MK-3475) in patients with advanced solid tumors
.
Clin Cancer Res
2017
;
23
:
5349
57
.
147.
Gopal
AK
,
Levy
R
,
Houot
R
,
Patel
SP
,
Popplewell
L
,
Jacobson
C
, et al
.
First-in-human study of utomilumab, a 4-1BB/CD137 agonist, in combination with rituximab in patients with follicular and other CD20(+) non-Hodgkin lymphomas
.
Clin Cancer Res
2020
;
26
:
2524
34
.
148.
Zheng
L
,
Judkins
C
,
Hoare
J
,
Klein
R
,
Parkinson
R
,
Wang
H
, et al
.
Urelumab (anti-CD137 agonist) in combination with vaccine and nivolumab treatments is safe and associated with pathologic response as neoadjuvant and adjuvant therapy for resectable pancreatic cancer
.
J Immunother Cancer
2020
;
8
Suppl 3
:
A486
.
Abstract nr
812
.
149.
Lee
SW
,
Salek-Ardakani
S
,
Mittler
RS
,
Croft
M
.
Hypercostimulation through 4-1BB distorts homeostasis of immune cells
.
J Immunol
2009
;
182
:
6753
62
.
150.
Labrijn
AF
,
Janmaat
ML
,
Reichert
JM
,
Parren
P
.
Bispecific antibodies: a mechanistic review of the pipeline
.
Nat Rev Drug Discov
2019
;
18
:
585
608
.
151.
Carter
PJ
,
Lazar
GA
.
Next generation antibody drugs: pursuit of the ‘high-hanging fruit
’.
Nat Rev Drug Discov
2018
;
17
:
197
223
.
152.
Hinner
MJ
,
Aiba
RSB
,
Jaquin
TJ
,
Berger
S
,
Durr
MC
,
Schlosser
C
, et al
.
Tumor-localized costimulatory T-cell engagement by the 4-1BB/HER2 bispecific antibody-anticalin fusion PRS-343
.
Clin Cancer Res
2019
;
25
:
5878
89
.
153.
Compte
M
,
Harwood
SL
,
Erce-Llamazares
A
,
Tapia-Galisteo
A
,
Romero
E
,
Ferrer
I
, et al
.
An Fc-free EGFR-specific 4-1BB-agonistic trimerbody displays broad antitumor activity in humanized murine cancer models without toxicity
.
Clin Cancer Res
2021
;
27
:
3167
77
.
154.
Compte
M
,
Harwood
SL
,
Munoz
IG
,
Navarro
R
,
Zonca
M
,
Perez-Chacon
G
, et al
.
A tumor-targeted trimeric 4-1BB-agonistic antibody induces potent anti-tumor immunity without systemic toxicity
.
Nat Commun
2018
;
9
:
4809
.
155.
Legg
JW
.
Tumor dependent co-stimulation of CD137/4-1BB in PSMA positive tumors: preclinical characterization of CB307, a half-life extended PSMAxCD137 bispecific Humabody therapeutic
[abstract]. In
:
Proceedings of the Annual Meeting of the American Association for Cancer Research 2020
;
2020
Apr 27–28 and Jun 22–24
.
Philadelphia (PA)
:
AACR
;
Cancer Res 2020;80(16 Suppl):Abstract nr 3352
.
156.
Hurov
K
,
Lahdenranta
J
,
Upadhyaya
P
,
Haines
E
,
Cohen
H
,
Repash
E
, et al
.
BT7480, a novel fully synthetic Bicycle tumor-targeted immune cell agonist (Bicycle TICA) induces tumor localized CD137 agonism
.
J Immunother Cancer
2021
;
9
:
e002883
.
157.
Xu
C
,
Rabinovich
B
,
Deshpande
A
,
Zhou
X
,
Pipp
FC
,
Schweickhardt
R
, et al
.
M9657, a novel tumor-targeted conditional anti-CD137 agonist displays MSLN-dependent anti-tumor immunity
.
J Immunother Cancer
2021
;
9
Suppl 2
:
A792
.
Abstract nr
757
.
158.
You
G
,
Lee
Y
,
Kang
YW
,
Park
HW
,
Park
K
,
Kim
H
, et al
.
B7-H3×4-1BB bispecific antibody augments antitumor immunity by enhancing terminally differentiated CD8+ tumor-infiltrating lymphocytes
.
Sci Adv
2021
;
7
:
eaax3160
.
159.
Rajendran
S
,
Li
Y
,
Ngoh
E
,
Wong
HY
,
Cheng
MS
,
Wang
CI
, et al
.
Development of a bispecific antibody targeting CD30 and CD137 on hodgkin and reed-sternberg cells
.
Front Oncol
2019
;
9
:
945
.
160.
Hutchings
M
,
Offner
F
,
Bosch
F
,
Gritti
G
,
Carlo-Stella
C
,
Walter
H
, et al
.
Phase 1 study of CD19 targeted 4-1BBL costimulatory agonist to enhance T cell (glofitamab combination) or NK cell effector function (obinutuzumab combination) in relapsed/refractory B cell lymphoma
.
Blood
2020
;
136
Suppl 1
:
16
7
. Abstract nr 3269.
161.
Sanmamed
MF
,
Rodriguez
I
,
Schalper
KA
,
Onate
C
,
Azpilikueta
A
,
Rodriguez-Ruiz
ME
, et al
.
Nivolumab and urelumab enhance antitumor activity of human T lymphocytes engrafted in Rag2−/−IL2Rgammanull immunodeficient mice
.
Cancer Res
2015
;
75
:
3466
78
.
162.
Sanmamed
MF
,
Chester
C
,
Melero
I
,
Kohrt
H
.
Defining the optimal murine models to investigate immune checkpoint blockers and their combination with other immunotherapies
.
Ann Oncol
2016
;
27
:
1190
8
.
163.
Zhai
T
,
Wang
C
,
Xu
Y
,
Huang
W
,
Yuan
Z
,
Wang
T
, et al
.
Generation of a safe and efficacious llama single-domain antibody fragment (vHH) targeting the membrane-proximal region of 4-1BB for engineering therapeutic bispecific antibodies for cancer
.
J Immunother Cancer
2021
;
9
:
e002131
.
164.
Ku
G
,
Bendell
JC
,
Tolcher
AW
,
Hurvitz
SA
,
Krishnamurthy
A
,
El-Khoueiry
AB
, et al
.
A phase I dose escalation study of PRS-343, a HER2/4-1BB bispecific molecule, in patients with HER2-positive malignancies
.
Ann Oncol
2020
;
31
Suppl 4
:
S462
S3
.
Abstract nr
525O
.
165.
Claus
C
,
Ferrara
C
,
Xu
W
,
Sam
J
,
Lang
S
,
Uhlenbrock
F
, et al
.
Tumor-targeted 4-1BB agonists for combination with T cell bispecific antibodies as off-the-shelf therapy
.
Sci Transl Med
2019
;
11
:
eaav5989
.
166.
Trub
M
,
Uhlenbrock
F
,
Claus
C
,
Herzig
P
,
Thelen
M
,
Karanikas
V
, et al
.
Fibroblast activation protein-targeted-4-1BB ligand agonist amplifies effector functions of intratumoral T cells in human cancer
.
J Immunother Cancer
2020
;
8
:
e000238
.
167.
Teijeira
A
,
Migueliz
I
,
Garasa
S
,
Karanikas
V
,
Luri
C
,
Cirella
A
, et al
.
Three-dimensional colon cancer organoids model the response to CEA-CD3 T-cell engagers
.
Theranostics
2022
;
12
:
1373
87
.
168.
Moreno
V
,
Hernández
T
,
Melero
I
,
Sanmamed
MF
,
Spanggaard
I
,
Rohrberg
KS
, et al
.
Pharmacodynamic assessment of a novel FAP-targeted 4–1BB agonist, administered as single agent and in combination with atezolizumab to patients with advanced solid tumors
.
J Immunother Cancer
2020
;
8
Suppl 3
:
A225
.
Abstract nr
370
.
169.
Melero
I
,
Sanmamed
MF
,
Calvo
E
,
Moreno
I
,
Moreno
V
,
Guerrero
TCH
, et al
.
First-in-human (FIH) phase I study of RO7122290 (RO), a novel FAP-targeted 4-1BB agonist, administered as single agent and in combination with atezolizumab (ATZ) to patients with advanced solid tumours
.
Ann Oncol
2020
;
31
Suppl 4
:
S707
.
Abstract nr
1025MO
.
170.
Muik
A
,
Garralda
E
,
Altintas
I
,
Gieseke
F
,
Geva
R
,
Ben-Ami
E
, et al
.
Preclinical characterization and phase I trial results of a bispecific antibody targeting PD-L1 and 4-1BB (GEN1046) in patients with advanced refractory solid tumors
.
Cancer Discov
2022
;
12
:
1248
65
.
171.
Geuijen
C
,
Tacken
P
,
Wang
LC
,
Klooster
R
,
van Loo
PF
,
Zhou
J
, et al
.
A human CD137xPD-L1 bispecific antibody promotes anti-tumor immunity via context-dependent T cell costimulation and checkpoint blockade
.
Nat Commun
2021
;
12
:
4445
.
172.
Lakins
MA
,
Koers
A
,
Giambalvo
R
,
Munoz-Olaya
J
,
Hughes
R
,
Goodman
E
, et al
.
FS222, a CD137/PD-L1 tetravalent bispecific antibody, exhibits low toxicity and antitumor activity in colorectal cancer models
.
Clin Cancer Res
2020
;
26
:
4154
67
.
173.
Warmuth
S
,
Gunde
T
,
Snell
D
,
Brock
M
,
Weinert
C
,
Simonin
A
, et al
.
Engineering of a trispecific tumor-targeted immunotherapy incorporating 4-1BB co-stimulation and PD-L1 blockade
.
Oncoimmunology
2021
;
10
:
2004661
.
174.
Jeong
S
,
Park
E
,
Kim
HD
,
Sung
E
,
Kim
H
,
Jeon
J
, et al
.
Novel anti-4-1BBxPD-L1 bispecific antibody augments anti-tumor immunity through tumor-directed T-cell activation and checkpoint blockade
.
J Immunother Cancer
2021
;
9
:
e002428
.
175.
Peper-Gabriel
JK
,
Pavlidou
M
,
Pattarini
L
,
Morales-Kastresana
A
,
Jaquin
TJ
,
Gallou
C
, et al
.
The PD-L1/4-1BB bispecific antibody-anticalin fusion protein PRS-344/S095012 elicits strong T-cell stimulation in a tumor-localized manner
.
Clin Cancer Res
2022
;
28
:
3387
99
.
176.
Muik
A
,
Altintas
I
,
Gieseke
F
,
Schoedel
KB
,
Burm
SM
,
Toker
A
, et al
.
An Fc-inert PD-L1x4-1BB bispecific antibody mediates potent anti-tumor immunity in mice by combining checkpoint inhibition and conditional 4-1BB co-stimulation
.
Oncoimmunology
2022
;
11
:
2030135
.
177.
Herter
S
,
Sam
J
,
Ferrara Koller
C
,
Diggelmann
S
,
Bommer
E
,
Schönle
A
, et al
.
RG6076 (CD19-4-1BBL): CD19-targeted 4-1BB ligand combination with glofitamab as an off-the-shelf, enhanced T-cell redirection therapy for B-cell malignancies
.
Blood
2020
;
136
Suppl 1
:
40.
Abstract nr
625
.
178.
Muik
A
,
Adams 3rd
HC
,
Gieseke
F
,
Altintas
I
,
Schoedel
KB
,
Blum
JM
, et al
.
DuoBody-CD40x4-1BB induces dendritic-cell maturation and enhances T-cell activation through conditional CD40 and 4-1BB agonist activity
.
J Immunother Cancer
2022
;
10
:
e004322
.
179.
Muik
A
,
Kosoff
R
,
Gieseke
F
,
Altintas
I
,
Schödel
K
,
Burm
S
, et al
.
DuoBody-CD40×4-1BB (GEN1042) induces dendritic-cell maturation and enhances T-cell activation and effector functions in vitro by conditional CD40 and 4-1BB agonist activity
[abstract]. In:
Proceedings of the American Association for Cancer Research Annual Meeting 2021
;
2021
Apr 10–15 and May 17–21
.
Philadelphia (PA)
:
AACR
;
Cancer Res 2021;81(13_Suppl):Abstract nr 1846
.
180.
Etxeberria
I
,
Bolanos
E
,
Teijeira
A
,
Garasa
S
,
Yanguas
A
,
Azpilikueta
A
, et al
.
Antitumor efficacy and reduced toxicity using an anti-CD137 probody therapeutic
.
Proc Natl Acad Sci U S A
2021
;
118
:
e2025930118
.
181.
Kamata-Sakurai
M
,
Narita
Y
,
Hori
Y
,
Nemoto
T
,
Uchikawa
R
,
Honda
M
, et al
.
Antibody to CD137 activated by extracellular adenosine triphosphate is tumor selective and broadly effective in vivo without systemic immune activation
.
Cancer Discov
2021
;
11
:
158
75
.
182.
Sugyo
A
,
Tsuji
AB
,
Sudo
H
,
Narita
Y
,
Taniguchi
K
,
Nemoto
T
, et al
.
In vivo validation of the switch antibody concept: SPECT/CT imaging of the anti-CD137 switch antibody Sta-MB shows high uptake in tumors but low uptake in normal organs in human CD137 knock-in mice
.
Transl Oncol
2022
;
23
:
101481
.