Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2019 Apr 28.
Published in final edited form as: Int Rev Cell Mol Biol. 2018 Aug 20;342:1–25. doi: 10.1016/bs.ircmb.2018.07.003

Stimulating T cells against cancer with agonist immunostimulatory monoclonal antibodies

Xue Han 1, Matthew D Vesely 2
PMCID: PMC6487201  NIHMSID: NIHMS1021129  PMID: 30635089

Abstract

Elimination of cancer cells through anti-tumor immunity has been a long-sought after goal since Sir F. Macfarlane Burnet postulated the theory of immune surveillance against tumors in the 1950s. Finally, the use of immunotherapeutics against established cancer is becoming a reality in the past 5 years. Most notable are the monoclonal antibodies directed against inhibitory T cell receptors cytotoxic T lymphocyte antigen 4 (CTLA-4) and programmed death 1 (PD-1). The next generation of monoclonal antibodies targeting T cells are designed to stimulate co-stimulatory receptors on T cells. Here we review the recent progress on these immunostimulatory agonist antibodies against the costimulatory receptors CD137, GITR, OX40, and CD27.

Keywords: immunooncology, cancer immunothereapy, agonist, immunostimulatory

Introduction

In the past decade, there has been remarkable and stunning progress in our ability to modulate the immune system against human cancers in clinic. This progress is the result of the efforts from the previous few decades focusing on T cell activation and cancer immunoediting (Vesely, Kershaw et al., 2011). We now know there are numerous stimulatory and inhibitory receptors on T cells that help fine-tune the immune response after the T cell receptor (TCR) engages its cognate-major histocompatibility complex (MHC)/peptide ligand (Figure 1). The recent success of cancer immunotherapy is primarily due to monoclonal antibodies (mAbs) directed against inhibitory receptors such as cytotoxic T lymphocyte antigen 4 (CTLA-4) and programmed death 1 (PD-1). These molecules restrain T cell activation and function and are thus, often referred to as immune checkpoints. Therapeutic antibodies targeting these molecules, referred to as immune checkpoint inhibition, are often likened to “releasing the brakes” on the immune system (Alderson, Smith et al., 1994, Sharma and Allison, 2015). Despite recent successes of immune checkpoint inhibition, the majority of patients still fail therapy, necessitating the need for combinatorial therapies. A group of attractive combinatorial targets are T cell activation receptors, often referred to as costimulatory receptors. Treatment with agonist mAbs against these receptors provide stimulatory signals to T cells to enhance effector function against tumors and can be likened to “pressing on the gas” on the immune system (Figure 2). Here, we review recent progress on targeting T cell costimulatory molecules with agonist antibodies for the treatment of cancer.

Figure 1:

Figure 1:

Regulation of T cells by modulating TCR signals through co-stimulatory and co-inhibitory ligands and receptors.

Figure 2:

Figure 2:

An immune synapse between effector T cells and an antigen-presenting cell (APC). Co-stimulatory ligands and receptors belonging to B7/CD28, and TNF/TNFR families are expressed on antigen presenting cells and T cells. The agonist monoclonal antibodies under clinical development can mimic the ligand to engage the co-stimulatory receptors.

CD137

CD137, also known as 4–1BB or tumor necrosis factor receptor 9 (TNFR9), was originally discovered in 1989 as an inducible molecule on the surface of activated CD4+ and CD8+ T cells (Kwon and Weissman, 1989). It is a member of the TNFR superfamily and is expressed as a homotrimer. Its ligand, CD137L, or called 4–1BBL, is also expressed as a homotrimer on the surface of antigen presenting cells (APCs) (Alderson, Smith et al., 1994). Upon antigen-specific TCR activation, T cells express higher levels of CD137 which when engaged with its ligand CD137L on APCs, augments proliferation, cytokine secretion and survival, thereby enhancing effector functions (Sanmamed, Pastor et al., 2015).

In addition to activated T cells, CD137 is also expressed on regulatory T cells, B cells, myeloid cells and activated natural killer (NK) cells (Melero, Bach et al., 1998, Melero, Johnston et al., 1998, Melero, Murillo et al., 2008, Vinay and Kwon, 2011). Mice-deficient in CD137 were found to have reduced long-lived memory T cells to specific antigens (Willoughby, Kerr et al., 2014). Binding of CD137 to its ligand CD137L results in recruitment of the TNFR-associated factor (TRAF) 1 and TRAF2, resulting in downstream activation of the nuclear factor-kappa-light-chain-enhancer of activated B cells (NF-κB) and mitogen-activated protein (MAP) kinase signaling pathways (Martinez-Forero, Azpilikueta et al., 2013, Sabbagh, Pulle et al., 2008, Saoulli, Lee et al., 1998). Ultimately, this results in secretion of interleukin-2 (IL-2) and interferon-γ (IFN-γ) as well as upregulation of anti-apoptotic molecules Bcl-xL and Bfl-1 which contribute to T cell expansion, survival and function (So and Croft, 2013).

Preclinical studies in mice demonstrated strong anti-tumor response with agonist anti-CD137 mAbs (Melero, Shuford et al., 1997). In fact, stimulation of CD137 is one of the most powerful antibody-based cancer immunotherapeutic strategies in mouse models (Vinay and Kwon, 2012, Wilcox, Flies et al., 2002). In addition, agonist anti-CD137 synergizes with radiotherapy and chemotherapy (Ju, Cheon et al., 2008, Shi and Siemann, 2006). The anti-cancer effect of agonist anti-CD137 is explained not only by enhancing T cell function, but also its effects on NK cells. Stimulation of CD137 on NK cells results in NK cell activation as well as enhanced antibody-directed cellular cytotoxicity (ADCC) in murine models of lymphoma (Muntasell, Ochoa et al., 2017, Ochoa, Minute et al., 2017, Rajasekaran, Chester et al., 2015, Wang, Erbe et al., 2015). Uniquely, there is evidence that CD137 is internalized after ligation with its ligand and continues to signal through endosomal compartments (Martinez-Forero, Azpilikueta et al., 2013). Overall, CD137 is one of the most attractive targets for agonist immunotherapy. Its ability to potentiate T cell responses is underscored by the fact that third-generation chimeric antigen receptor (CAR) T cells use the intracellular CD137 signaling motif to generate the most successful CAR T cells date and outperforms CAR T cells with CD28 intracellular domains (June, O’Connor et al., 2018).

Two anti-CD137 agonists are furthest along in the treatment of human cancers, urelumab (BMS-663513) and utomilumab (PF-05082566) (Table I). Urelumab is a fully human IgG4 mAb that does not block the interaction between CD137 and its endogenous ligand, CD137L. Initial results in 2008 using urelumab were promising against advanced cancers (Segal, Logan et al., 2017), but a small number of patients experienced liver toxicity at the highest dose of urelumab. Thus, further clinical development was halted until 2012 when the promise of immunotherapy with agents targeting CTLA-4 and PD-1 was finally being realized. In subsequent clinical trials, a lower dose of urelumab was used in combination with rituximab, cetuximab, elotuzumab, and nivolumab that did not show liver toxicity (NCT01471210, NCT01775631, NCT02110082, NCT02252263, and NCT02253992). One particularly exciting preliminary result is the combination of urelumab with rituximab in patients with relapsed or refractory B cell non-Hodgkin lymphoma. These new trials with the lower dose of urelumab that have minimal liver toxicity are promising as we wait for the final results. The bioactivity of urelumab has been examined in many of these trials and found that urelumab in combination with cetuximab (anti-epidermal growth factor receptor) increases dendritic cell (DC) maturation and NK cell cytotoxicity (Kohrt, Colevas et al., 2014, Srivastava, Trivedi et al., 2017).

Table 1:

Ongoing clinical trials with costimulatory agonist monoclonal antibodies for cancer immunotherapy.

Reagent Company Diseases Phase Status Treatment # ClinicalTrials.gov First posted
OX40 agonists
9B12 AgonOx/Medimmune Solid tumors I Completed As a single agent NCT01644968 2012
GSK3174998 GlaxoSmithKline Solid tumors I Recruiting As a single agent or combined with pembrolizumab (a-PD-1) NCT02528357 2015
Solid tumors I Recruiting Combined with GSK1795091 (TLR4 agonist) NCT03447314 2018
INCAGN01949 Incyte Agenus Solid tumors I/II Recruiting As a single agent NCT02923349 2016
Solid tumors I/II Recruiting Combined with nivolumab(a-PD-1) +/−ipilimumab (a-CTLA4) NCT03241173 2017
MEDI-0562 MedImmune Solid tumors I Completed As a single agent NCT02318394 2014
Solid tumors I Recruiting Combined with tremelimumab (a-CTLA4) or durvalumab (a-PD-L1) NCT02705482 2016
MEDI-6469 MedImmune Solid tumors/B cell lymphoma I/II Ternimated As a single agent or combined with tremelimumab (a-CTLA4), durvalumab (a-PD-L1), or rituximab (a-CD20) NCT02205333 2014
Head and neck I Active, not recruiting Given prior to surgery NCT02274155 2014
Colorectal cancer I Active, not recruiting As a single agent NCT02559024 2015
MEDI-6383 (OX40L-Fc) MedImmune Solid tumors I Active, not recruiting As a single agent or combined with MEDI-4736 (a-PD-L1) NCT02221960 2014
MOXR0916 Genentech Solid tumors I Active, not recruiting As a single agent NCT02219724 2014
Solid tumors I Active, not recruiting Combined with atezolizumab(a-PD-L1) NCT02410512 2015
Urothelial carcinoma II Active, not recruiting Combined with atezolizumab (a-PD-L1) NCT03029832 2017
PF-04518600 Pfizer Solid tumors I Recruiting As signle agent or combined with PF-05082566 (a-4–1BB) NCT02315066 2014
Renal cell carcinoma II Recruiting Combined with axitinib (tyrosine kinase inhibitor) NCT03092856 2017
Solid tumors I/II Recruiting Combined with avelumab (a-PD-L1) NCT03217747 2017
Leukemia I/II Recruiting As signle agent or combined with avelumab (a-PD-L1 mAb) +/− azacitidine (analog of cytidine), or with utomilumab (a-41BB mAb) NCT03390296 2018
BMS-986178 Bristol-Myers Squibb Solid tumors I/II Recruiting As a sigle agent or combined with nivolumab (a-PD-1) +/− ipilimumab (a-CTLA4) NCT02737475 2016
B-Cell Non-Hodgkin Lymphomas I Not yet recruiting Combined with radiation therapy and TLR9 agonist NCT03410901 2018
 
CD27 agonists
Varlilumab Celldex Therapeutics Hematologic cancers and solid tumors I Completed As single agent NCT01460134 2011
Ovarian or Breast Cancer I Completed Combined with ONT-10 (MUC1-based cancer vaccine) NCT02270372 2014
Melanoma II Recruiting Combined with Glembatumumab Vedotin (cancer cell targeted toxic drug) NCT02302339 2014
Melanoma I/II Terminated (Feasibility concerns) Combined with ipilimumab (a-CTLA4) c/i CDX-1401(vaccine) and poly-ICLC) NCT02413827 2015
Renal cell carcinoma I/II Terminated (Portfolio re-prioritization) As a single agent or combined with sunitinib (receptor tyrosine kinase inhibitor) NCT02386111 2015
Solid tumors I/II Terminated (Portfolio re-prioritization) Combined with atezolizumab (a-PD-L1) NCT02543645 2015
Solid tumors I/II Active, not recruiting Combined with nivolumab (a-PD-1) NCT02335918 2015
Glioma I Recruiting Combined with IMA950 peptides and poly-ICLC NCT02924038 2016
B-cell lymphoma II Recruiting Combined with nivolumab (a-PD-1) NCT03038672 2017
B-cell lymphoma II Recruiting Combined with rituximab (a-CD20) NCT03307746 2017
 
CD137 agonists
Utomilumab Pfizer Diffuse large B-cell lymphoma I Recruiting Combined with avelumab (a-PD-L1), and rituximab (a-CD20) or azacitidine NCT02951156 2016
Advanced cancers I Active, not recruiting As a single agent and in combination with ritxuimab (a-CD20) NCT01307267 2011
Solid tumors I Recruiting Combined with mogamulizab (a-CCR4) NCT02444793 2015
Solid tumors I/II Recruiting Combined with avelumab (a-PD-L1) C/¡ PF-04518600 (a-OX40) NCT02554812 2015
Breast Cancer I Recruiting Combined with trastuzumab emtansine(a-HER2/neu + tubulin inhibitor) or trastuzumab (a-HER2/neu) NCT03364348 2017
Colorectal Cancer II Recruiting Combined with cetuximab (a-EGFR) and irinotecan (topoisomerase inhibitor)
 
Urelumab Bristol-Myers Squibb Glioblastoma I Recruiting As a single agent or combined with nivolumab (a-PD-1) NCT02658981 2016
Leukemia II Withdrawn Combined with rituximab (a-CD20) NCT02420938 2015
Solid tumors II Recruiting As a single agent or combined with nivolumab (a-PD-1) NCT02534506 2015
Urothelial carcinoma II Not yet recruiting Combined with nivolumab (a-PD-1) NCT02845323 2016
Melanoma I Recruiting Combined with nivolumab (a-PD-1) NCT02652455 2017
 
GITR agonist
AMG-228 Amgen Solid tumors I Terminated As a single agent NCT02437916 2015
 
BMS-986156 Bristol-Myers Squibb Solid tumors I/II Active, not recruiting As a single agent or combined with nivolumab (a-PD-1) NCT02598960 2015
 
GWN323 Novartis Lymphomas I Recruiting As a single agent or combined with PDR001 (a-PD-1) NCT02740270 2016
 
INCAGN01876 Incyte Solid tumors I/II Recruiting As a single agent NCT02697591 2016
Solid tumors I/II Recruiting Combined with nivolumab (a-PD-1) and/or ipilimumab (a-CTLA4) NCT03126110 2017
Solid tumors I/II Actine, not recruiting Combined with pembrolizumab (a-PD-1) and/or epacadostat (IDO inhibitor) NCT03277352 2017
 
MEDI-1873 MedImmune Solid tumors I Active, not recruiting As a single agent NCT02583165 2015
 
MK-1248 Merck Solid tumors I Active, not recruiting As a single agent or combined with pembrolizumab (a-PD-1) NCT02553499 2015
 
MK-4166 Merck Solid tumors I Recruiting As a single agent or combined with pembrolizumab (a-PD-1) NCT02132754 2014
 
TRX518 Leap Therapeutics Solid tumors I Recruiting As a single agent NCT02628574 2015
 
OMP-336B11 OncoMed Solid tumors I Recruiting As a single agent NCT03295942 2017

Utomilumab is a humanized IgG2 mAb that both activates CD137 and blocks engagement with CD137L. Furthermore, it appears that utomilumab has a superior safety profile since there does not appear to be significant liver toxicity. Initial phase I/II clinical trials are encouraging in that there are some durable responses in patients with follicular lymphomas treated with utomilumab (Segal, He et al., 2018, Tolcher, Sznol et al., 2017). With a favorable safety profile and some initial evidence of efficacy, many ongoing trials are using utomilumab in combination with other immunotherapies against a wide array of cancers such as non-small cell lung cancer, renal cell carcinoma, and head and neck cancer (Tolcher, Sznol et al., 2017). Perhaps the most promising malignancy for both urelumab and utomilumab appears to be rituximab-refractory or relapsed B cell lymphomas. We eagerly await the results of these ongoing trials (Table 1).

GITR

Glucocorticoid-induced TNFR-related protein (GITR, TNFSFR18, CD357) is another member of the TNFR superfamily that is induced after TCR engagement in CD4+ and CD8+ T cells (Gurney, Marsters et al., 1999, Nocentini, Giunchi et al., 1997). GITR was initially discovered in a screen of murine T cell hybridomas treated with dexamethasone (Nocentini, Giunchi et al., 1997), however subsequent studies have found that glucocorticoids have no effect on GITR expression in both mice and humans. In contrast to CD4+ and CD8+ effector T cells where GITR expression is induced after TCR signaling, regulatory T cells (Tregs) display constitutively high levels of GITR (Shimizu, Yamazaki et al., 2002). Whereas GITR signaling stimulates effector T cell function, it appears to function as an inhibitory receptor on Tregs, thereby limiting the suppressive function of Tregs (McHugh, Whitters et al., 2002). Like the other costimulatory receptors discussed in this review, expression of GITR is not limited to T cells and can be induced on NK cells, B cells, and myeloid cells (Schaer, Murphy et al., 2012). The binding of GITR with its ligand GITRL results in activation of the NF-kB and MAPK pathways mediated by TRAF5 and TRAF2 adapter proteins (Snell, McPherson et al., 2010). Ultimately, GITR-induced signaling results in enhanced T cell proliferation and effector function through the production of IL-2 and IFN-γ. In addition, GITR signaling seems to protect against activation-induced cell death (Turk, Guevara-Patino et al., 2004). On CD8+ T cells, GITR signaling appears to lower the threshold of CD28 signaling and induces expression of CD137 (4–1BB) on memory CD8+ T cells (Lin, Snell et al., 2013, Ronchetti, Nocentini et al., 2007). There also seems to be a unique feature of agonist-GITR mAbs to elicit IL-9 producing CD4+ T cells (Th9) which are critical to for CT26 colon carcinoma rejection in mice (Kim, Kim et al., 2015).

Preclinical studies using agonist anti-GITR mAbs have shown antitumor effect against of number of murine cancers. The most commonly used mAb, clone DTA-1 has been used to successfully treat mice against B16 melanoma, CT26 colon carcinoma and sarcomas (Cohen, Diab et al., 2006, Ko, Yamazaki et al., 2005, Zhou, L’Italien et al., 2007). Synergistic effects beyond monotherapy were also achieved with CTLA-4, Toll-like-receptor (TLR) ligands and vaccines. This antitumor effect of agonist anti-GITR mAbs is thought to be due to enhancing effector T cell function as well as inhibiting Tregs. The anti-GITR mAb DTA-1 has been reported to directly deplete Tregs through an FcγR-dependent mechanism (Coe, Begom et al., 2010). With this compelling, preclinical data, humanized agonist anti-GITR antibodies were developed for clinical use.

Currently there are several agents targeting GITR in ongoing clinical trials (Knee, Hewes et al., 2016) (Table 1). The first agonist anti-GITR mAb, (TRX518) against malignant melanoma showed little toxicity, but also little efficacy. Repeat phase I trial with TRX518 against multiple advanced cancers started in 2015 with no results reported yet. Subsequently, a number of agonist anti-GITR mAbs have entered phase I/II clinical trials including INCAGN01876 (Incyte), MK-4166 (Merck), AMG 228 (Amgen) and BMS-986156 (BMS). Furthermore, Medimmune with Astra-Zeneca has developed a hexamerix GITRL protein (MEDI1873) currently in phase I clinical trials that was designed to maximize stimulating effector T cells while inhibiting or depleting regulatory T cells. Results from these trials are still pending.

OX40

OX40, also known as CD134 or TNFRSF4, is a type I transmembrane protein and a member of TNFR superfamily. It was initially discovered using a panel of monoclonal antibodies generated against rat CD4+ T cell blasts. One antibody in the panel, MRC-OX40, increased CD4+ T-cell proliferation under either concanavalin A or alloantigen stimulation in vitro and the target was subsequently named OX40 (Mallett, Fossum et al., 1990, Paterson, Jefferies et al., 1987). In addition to CD4+ T cells, OX40 is also expressed on CD8+ T cells following activation (Mallett, Fossum et al., 1990). In mice, Tregs constitutively express OX40, while in humans, OX40 expression is upregulated on Tregs upon activation (Valzasina, Guiducci et al., 2005). NK, NKT cells and neutrophils also express OX40(Baumann, Yousefi et al., 2004).

OX40L, the ligand of OX40, also known as TNFSF4 and CD252, was first identified as a new glycoprotein on T-cell leukemia virus type-I transformed lymphocytes (Tanaka, Inoi et al., 1985) and later found to bind OX40 (Baum, Gayle et al., 1994, Godfrey, Fagnoni et al., 1994). OX40L is not constitutively expressed but, rather is induced on activated APCs including DCs (Ohshima, Tanaka et al., 1997), B cells (Stuber, Neurath et al., 1995) and macrophages (Weinberg, Wegmann et al., 1999). The expression of OX40L on APCs is in line with its function in controlling the extent of T cell priming following recognition of antigen (Gramaglia, Jember et al., 2000, Gramaglia, Weinberg et al., 1998). OX40 ligation with OX40L recruits TRAF2 and TRAF3 to the intracellular domain of OX40, leading to activation of both the canonical and non-canonical NF-κB pathways (Kawamata, Hori et al., 1998). Downstream signaling ultimately leads to the expression of pro-survival molecules including Bcl-xL and Bcl-2, increased cytokine production associated with enhanced T-cell expansion, differentiation, and the generation of long-lived memory cells (Rogers, Song et al., 2001, Song, So et al., 2005).

Agonist anti-OX40 mAbs have been reported to reverse CD4+ T-cell tolerance by overturning the anergic state induced by antigenic peptides under non-inflammatory conditions (Bansal-Pakala, Jember et al., 2001). Engagement of OX40 increases tumor immunity against multiple transplantable syngeneic tumors including sarcomas, melanoma, colon carcinoma, and glioma in experiments using in vivo gene transfer of OX40 ligand to tumor cells or administration of OX40L-Fc or OX40 agonist mAbs (Andarini, Kikuchi et al., 2004, Kjaergaard, Tanaka et al., 2000, Weinberg, Rivera et al., 2000). However, anti-OX40 administration shows very limited impact on the growth of poorly immunogenic tumors (Kjaergaard, Tanaka et al., 2000). In this context, combinational strategies could be important to increase OX40 agonist antitumor efficacy. For example, in preclinical studies, OX40 stimulation has been demonstrated to enhance antitumor effects when combined with multiple therapeutic strategies including cytokines (Redmond, Triplett et al., 2012, Ruby, Montler et al., 2008), adjuvants (Gough, Crittenden et al., 2010, Houot and Levy, 2009, Voo, Foglietta et al., 2014), vaccinations (Murata, Ladle et al., 2006), chemotherapy (Hirschhorn-Cymerman, Rizzuto et al., 2009), or radiotherapy (Young, Baird et al., 2016). In addition, anti-OX40 antibodies have been combined with immunomodulatory antibodies against other costimulatory receptors (Lee, Myers et al., 2004, Morales-Kastresana, Sanmamed et al., 2013, Pan, Zang et al., 2002), or blocking coinhibitory pathways (Linch, Kasiewicz et al., 2016, Messenheimer, Jensen et al., 2017, Redmond, Linch et al., 2014) to treat lymphomas, sarcomas, colon metastases, and spontaneous hepatocellular carcinoma.

One of the main advantages of targeting OX40 is that OX40 signaling can prevent Treg-mediated suppression of antitumor immune responses. Three potential mechanisms have been described. First, OX40 signaling reduces the induction of adaptive Tregs. Mice-deficient in OX40 had normal development of naturally arising CD4+Foxp3+ Tregs in vivo, but OX40 costimulation prevents the induction of inducible Foxp3+ Tregs from either naïve or effector T cells in vitro (So and Croft, 2007, Vu, Xiao et al., 2007). Second, OX40 signaling reduces Treg suppressive activity. Triggering OX40 signaling on Tregs using either agonist antibody or OX40L overexpressed on APCs inhibits Treg capacity to suppress, allowing for greater effector T-cell proliferation and production of IL-2 and other cytokines (Valzasina, Guiducci et al., 2005, Vu, Xiao et al., 2007). For example, in mice bearing CT26 transplanted tumors, intratumoral injection of agonist anti-OX40 mAb resulted in reduced Treg function, more infiltrating DCs and an influx of tumor-specific cytotoxic T lymphocytes (Piconese, Valzasina et al., 2008). Thirdly, anti-OX40 mAbs can directly deplete Tregs. In a recent report, agonist anti-OX40 OX86 administration resulted in the depletion of intratumoral Tregs in an FcγR-dependent manner, which correlated with tumor regression (Bulliard, Jolicoeur et al., 2014).

The murine anti-human OX40 mAb (clone 9B12) was the first OX40 agonistic reagent tested in a clinical trial of 30 patients with advanced solid tumors. In this phase I study, although none of the patients showed an objective response by RECIST criteria, some immune responses like Ki67-staining by antigen-experienced CD4+ and CD8+ T cells in blood was increased, suggesting enhanced activation of T cells. In addition, upregulation of OX40 by tumor-infiltrating Tregs was detected. Overall, agonist anti-OX40 mAb 9B12 was well tolerated with mild to moderate side effects (Curti, Kovacsovics-Bankowski et al., 2013). To increase the anti-tumor response, combinations of 9B12 with chemotherapy and/or radiotherapy in clinical trials are currently ongoing in different solid tumors. However, elevated levels of neutralizing human anti-mouse immunoglobulin antibodies were detected in treated patients and is considered one of the most important limitations in this clinical trial. Thus, the development of fully human OX40 agonists may overcome this limitation. Currently, agonistic OX40 monoclonal antibodies (e.g. MOXR0916, PF-04518600, MEDI0562, MEDI6469, INCAGN01949, and GSK3174998) are being evaluated in several Phase I/II clinical trials either as monotherapy or in combination with other immunomodulating agents. In addition, OX40L-Fc has demonstrated promising results in preclinical model and MEDI-6383, a human OX40L-Fc is also being evaluated in clinical trials currently (Table 1).

CD27

CD27, also known as TNFRSF7, is a type I membrane protein and a member of TNFR family. CD27 was first cloned and characterized in 1987 as a novel T cell differentiation antigen by using an antibody directed against human T cells (van Lier, Borst et al., 1987). Murine and human CD27 are expressed on T cells, B cells, and NK cells (Gravestein, Nieland et al., 1995, Klein, Rajewsky et al., 1998, Sugita, Robertson et al., 1992, van Lier, Borst et al., 1987). A unique feature of CD27 among TNFR family members is its constitutive expression on the majority of T cells, which suggests that CD27 may play a role during T cell priming. Naive T cells constitutively expresses CD27, and TCR signaling further upregulates the CD27 expression (de Jong, Loenen et al., 1991). Interestingly, besides αβ T cells, CD27 is also highly expressed on a subset of γδ T cells, Vγ9Vδ2 T cells, that is thought to have antitumor activity (DeBarros, Chaves-Ferreira et al., 2011). CD27 is also expressed on subsets of NK cells. In mice, CD27 expression differentiates the mature CD11b high NK cell pool into two functionally distinct subsets. The CD27high NK cell subset has a greater effector function compared to CD27low NK cells (Hayakawa and Smyth, 2006). While in humans, the loss of CD27 on NK cells is associated with enhanced cytotoxicity (Vossen, Matmati et al., 2008). Importantly, CD27 is expressed by both effector and regulatory T-cell populations at sites of inflammation and cancer, indicating that both T cell populations are potential targets for CD27 immunomodulation.

CD70 is the only known ligand of CD27. The interaction between CD27 and its ligand CD70 provides a costimulatory signal for T cell expansion. Under physiologic conditions, the tight regulation of CD70 expression ensures the transient availability of this costimulatory signal on activated APCs, T cells, and NK cells. On human peripheral DCs, CD70 is absent at steady-state, but is upregulated by TNF-α, TLR agonists or irradiation (Huang, Wang et al., 2011, Krause, Bruckner et al., 2009, Oosterhoff, Heusinkveld et al., 2013).

On T cells, the simultaneous engagement of the TCR by MHC/peptide and CD27 by CD70 provides an important signal for T cell expansion. Mice constitutively expressing CD70 on B cells or DCs had higher number of peripheral T cells were a greater percentage were activated effector T cells (Arens, Tesselaar et al., 2001, Keller, Schildknecht et al., 2008). CD70-CD27 interaction leads to recruitment of TNFR-associated factor (TRAF) proteins TRAF2 and TRAF5 to the intracellular domain of CD27 (Akiba, Nakano et al., 1998, Gravestein, Amsen et al., 1998). Subsequent activation of NF-κβ and c-Jun pathways promotes CD8+ T cell priming, proliferation, survival, and cytotoxicity (Carr, Carrasco et al., 2006, Ramakrishnan, Wang et al., 2004, Rowley and Al-Shamkhani, 2004, Taraban, Rowley et al., 2006). In addition to the function on T cells, CD70/CD27 also promotes germinal center formation of B cells at the centroblast stage (Xiao, Hendriks et al., 2004). Additionally, a subset of NK cells express CD27, and engagement of CD27 results in NK cell proliferation and IFN-γ production (Takeda, Oshima et al., 2000).

The anti-tumor effect of CD27 pathway has been demonstrated in multiple mouse tumor models. In a mouse B cell lymphoma model, blocking CD27 costimulation by using a blocking mAb against CD70 completely abolished the anti-CD40 mAb therapeutic effect due to severely impairing CD8 T-cell expansion. Furthermore, it was shown in two different lymphoma models that agonistic anti-CD27 mAb given as a single reagent could completely protect tumor-bearing mice (French, Taraban et al., 2007). In an established mouse melanoma model, agonistic anti-CD27 antibody as monotherapy led a substantial reduction in the outgrowth of both subcutaneous tumors and experimental lung metastases. Some potential mechanisms include survival of tumor-specific CD8+ T cells, reducing the frequency of regulatory T cells within tumors, and enhancing the function of NK cells and CD8+ CTLs (Roberts, Franklin et al., 2010). In a recent study, the potential of anti-CD27 monotherapy for cancer immunotherapy was demonstrated in murine models of lymphoma (Turaj, Hussain et al., 2017). In this study, an extended panel of immunomodulatory mAbs, either as a single agent or combined with the direct tumor-targeting mAb anti-CD20, was used to treat murine lymphoma. Among all the immunomodulatory mAbs tested, including mAbs against costimulatory receptors OX40, 4–1BB, GITR, CD27 or checkpoint blockers TIGIT, PD-L1, PD-1, or CTLA-4, only anti-4–1BB and anti-CD27 showed efficacy as single reagent. Combination of anti-4–1BB/CD20 didn’t show any synergistic effect and combination of anti-CD20 with anti-OX40, GITR, TIGIT, PD-L1, PD-1, or CTLA-4 only showed a moderate benefit compared with anti-CD20 therapy alone. Remarkably, combination of anti-CD20/CD27 showed a complete cure. This effect was apparent in multiple lymphoma models, as well as in a human CD27 transgenic mice using the anti-human CD27 mAb, varlilumab. Single-cell RNA sequencing demonstrated that anti-CD27 stimulated CD8+ T and natural killer cells to release myeloid chemo-attractants and IFN-γ, to elicit myeloid infiltration and macrophage activation. This study demonstrates the therapeutic advantage of using an immunomodulatory mAb to regulate lymphoid cells, which then recruit and activate myeloid cells for enhanced killing of mAb-opsonized tumors.

Varlilumab (CDX-1127) is a fully human mAb that targets and stimulates CD27. In both in vitro assays and in vivo preclinical models, variliumab showed direct antitumor activity against a CD27-expressing lymphoma or leukemia by stimulating effector T cells and depleting T regulatory cells (Ramakrishna, Sundarapandiyan et al., 2015, Vitale, He et al., 2012). In addition, varlilumab showed synergistic effects when combined with either anti-CD20 or PD-1/PD-L1 blockade in pre-clinical lymphoma and melanoma tumor models. A Phase I study of varlilumab in patients with both solid cancers and hematologic malignancies demonstrated that varlilumab is well tolerated and has anti-tumor efficacy (Burris, Infante et al., 2017). Currently, there are more clinical trials of varlilumab as a cancer therapeutic that are ongoing (Table 1).

Emerging agonists: ICOS and CD40

In addition to CD137, GITR, OX40 and CD27, there are additional costimulatory receptors being targeted for cancer immunotherapy (Cabo, Offringa et al., 2017, Marin-Acevedo, Dholaria et al., 2018). One such receptor is inducible costimulator (ICOS or CD278) which is expressed on activated CD4+ T cells and member of the B7-CD28 family of proteins (Hutloff, Dittrich et al., 1999). Engagement of ICOS with its ligand ICOSL results in greater T cell proliferation and cytokine production (Simpson, Quezada et al., 2010). Of note, its levels are increased in response to anti-CTLA-4 treatment and thus current efforts are underway to use anti-CTLA-4 in combination with agonist anti-ICOS.

Agonist mAbs directed against TNFR family member CD40 have also emerged as a potentially potent anti-tumor therapy, especially in combination with either CTLA-4 or PD-1 blockade (Vonderheide and Glennie, 2013). CD40 is expressed on APCs including B cells and its ligand CD40L (CD152) is expressed on activated T cells. Initial safety concerns including the association of thromboembolic events and cytokine storm release have limited clinical development of agonist anti-CD40 (Dempke, Fenchel et al., 2017). Nevertheless, a series of agonist anti-CD40 reagents are currently in clinical trials and more are in clinical development.

CD28 superagonist as a cautionary tale

The excitement of cancer immunotherapy has been a boom for both academic and biotechnology firms in the development of novel targets for immunomodulation. Importantly, this has provided many patients with advanced or metastatic cancer a chance for survival when standard therapies fail. But it is important to remember that agonists stimulating the immune system can have dangerous side effects. The most infamous of these cases is the development of superagonist anti-CD28 TGN1412 (Hunig, 2012, Hunig, 2016). In March 2006, during a phase I clinical trial, six healthy volunteers who were given TGN1412 developed profound cytokine storm and nearly died. This occurred at a time before any successes with immunotherapy and sent shockwaves throughout hospitals, doctor’s offices, board rooms, and of course, patient’s homes. The lesson was clear: stimulate the immune response at your own risk and with great caution.

Conclusions

We are now in the next phase of immune-oncology, where T cells can be modulated by both blocking antibodies against inhibitory receptors and stimulating antibodies against activating receptors. The number of agonists stimulating T cells is growing and exciting early clinical trial data shows great promise. However, most of these agents are in phase I trials and there is no significant efficacy data from phase II or phase III clinical trials yet. Identifying the most appropriate cancer population subsets and what combinatorial therapy is most effective with the least tolerable toxicity remains on ongoing challenge. But what is certain is that costimulatory agonists such as those targeting CD137, GITR, OX40, and CD27 will be among our new weapons against cancer.

References

  1. Akiba H, et al. (1998). CD27, a member of the tumor necrosis factor receptor superfamily, activates NF-kappaB and stress-activated protein kinase/c-Jun N-terminal kinase via TRAF2, TRAF5, and NF-kappaB-inducing kinase. J Biol Chem 273(21) 13353–13358. [DOI] [PubMed] [Google Scholar]
  2. Alderson MR, et al. (1994). Molecular and biological characterization of human 4–1BB and its ligand. Eur J Immunol 24(9) 2219–2227. [DOI] [PubMed] [Google Scholar]
  3. Andarini S, et al. (2004). Adenovirus vector-mediated in vivo gene transfer of OX40 ligand to tumor cells enhances antitumor immunity of tumor-bearing hosts. Cancer Res 64(9) 3281–3287. [DOI] [PubMed] [Google Scholar]
  4. Arens R, et al. (2001). Constitutive CD27/CD70 interaction induces expansion of effector-type T cells and results in IFNgamma-mediated B cell depletion. Immunity 15(5) 801–812. [DOI] [PubMed] [Google Scholar]
  5. Bansal-Pakala P, Jember AG and Croft M (2001). Signaling through OX40 (CD134) breaks peripheral T-cell tolerance. Nat Med 7(8) 907–912. [DOI] [PubMed] [Google Scholar]
  6. Baum PR, et al. (1994). 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 13(17) 3992–4001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  7. Baumann R, et al. (2004). Functional expression of CD134 by neutrophils. Eur J Immunol 34(8) 2268–2275. [DOI] [PubMed] [Google Scholar]
  8. Bulliard Y, et al. (2014). OX40 engagement depletes intratumoral Tregs via activating FcgammaRs, leading to antitumor efficacy. Immunol Cell Biol 92(6) 475–480. [DOI] [PubMed] [Google Scholar]
  9. Burris HA, et al. (2017). Safety and Activity of Varlilumab, a Novel and First-in-Class Agonist Anti-CD27 Antibody, in Patients With Advanced Solid Tumors. J Clin Oncol 35(18) 2028–2036. [DOI] [PubMed] [Google Scholar]
  10. Cabo M, et al. (2017). Trial Watch: Immunostimulatory monoclonal antibodies for oncological indications. Oncoimmunology 6(12) e1371896. [DOI] [PMC free article] [PubMed] [Google Scholar]
  11. Carr JM, et al. (2006). CD27 mediates interleukin-2-independent clonal expansion of the CD8+ T cell without effector differentiation. Proc Natl Acad Sci U S A 103(51) 19454–19459. [DOI] [PMC free article] [PubMed] [Google Scholar]
  12. Coe D, et al. (2010). Depletion of regulatory T cells by anti-GITR mAb as a novel mechanism for cancer immunotherapy. Cancer Immunol Immunother 59(9) 1367–1377. [DOI] [PMC free article] [PubMed] [Google Scholar]
  13. Cohen AD, et al. (2006). Agonist anti-GITR antibody enhances vaccine-induced CD8(+) T-cell responses and tumor immunity. Cancer Res 66(9) 4904–4912. [DOI] [PMC free article] [PubMed] [Google Scholar]
  14. Curti BD, et al. (2013). OX40 is a potent immune-stimulating target in late-stage cancer patients. Cancer Res 73(24) 7189–7198. [DOI] [PMC free article] [PubMed] [Google Scholar]
  15. de Jong R, et al. (1991). Regulation of expression of CD27, a T cell-specific member of a novel family of membrane receptors. J Immunol 146(8) 2488–2494. [PubMed] [Google Scholar]
  16. DeBarros A, et al. (2011). CD70-CD27 interactions provide survival and proliferative signals that regulate T cell receptor-driven activation of human gammadelta peripheral blood lymphocytes. Eur J Immunol 41(1) 195–201. [DOI] [PubMed] [Google Scholar]
  17. Dempke WCM, et al. (2017). Second- and third-generation drugs for immuno-oncology treatment-The more the better? Eur J Cancer 74 55–72. [DOI] [PubMed] [Google Scholar]
  18. French RR, et al. (2007). Eradication of lymphoma by CD8 T cells following anti-CD40 monoclonal antibody therapy is critically dependent on CD27 costimulation. Blood 109(11) 4810–4815. [DOI] [PubMed] [Google Scholar]
  19. Godfrey WR, et al. (1994). Identification of a human OX-40 ligand, a costimulator of CD4+ T cells with homology to tumor necrosis factor. J Exp Med 180(2) 757–762. [DOI] [PMC free article] [PubMed] [Google Scholar]
  20. Gough MJ, et al. (2010). Adjuvant therapy with agonistic antibodies to CD134 (OX40) increases local control after surgical or radiation therapy of cancer in mice. J Immunother 33(8) 798–809. [DOI] [PMC free article] [PubMed] [Google Scholar]
  21. Gramaglia I, et al. (2000). The OX40 costimulatory receptor determines the development of CD4 memory by regulating primary clonal expansion. J Immunol 165(6) 3043–3050. [DOI] [PubMed] [Google Scholar]
  22. Gramaglia I, et al. (1998). Ox-40 ligand: a potent costimulatory molecule for sustaining primary CD4 T cell responses. J Immunol 161(12) 6510–6517. [PubMed] [Google Scholar]
  23. Gravestein LA, et al. (1998). The TNF receptor family member CD27 signals to Jun N-terminal kinase via Traf-2. Eur J Immunol 28(7) 2208–2216. [DOI] [PubMed] [Google Scholar]
  24. Gravestein LA, et al. (1995). Novel mAbs reveal potent co-stimulatory activity of murine CD27. Int Immunol 7(4) 551–557. [DOI] [PubMed] [Google Scholar]
  25. Gurney AL, et al. (1999). Identification of a new member of the tumor necrosis factor family and its receptor, a human ortholog of mouse GITR. Curr Biol 9(4) 215–218. [DOI] [PubMed] [Google Scholar]
  26. Hayakawa Y and Smyth MJ (2006). CD27 dissects mature NK cells into two subsets with distinct responsiveness and migratory capacity. J Immunol 176(3) 1517–1524. [DOI] [PubMed] [Google Scholar]
  27. Hirschhorn-Cymerman D, et al. (2009). OX40 engagement and chemotherapy combination provides potent antitumor immunity with concomitant regulatory T cell apoptosis. J Exp Med 206(5) 1103–1116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  28. Houot R and Levy R (2009). T-cell modulation combined with intratumoral CpG cures lymphoma in a mouse model without the need for chemotherapy. Blood 113(15) 3546–3552. [DOI] [PMC free article] [PubMed] [Google Scholar]
  29. Huang J, et al. (2011). Irradiation enhances human T-cell function by upregulating CD70 expression on antigen-presenting cells in vitro. J Immunother 34(4) 327–335. [DOI] [PMC free article] [PubMed] [Google Scholar]
  30. Hunig T (2012). The storm has cleared: lessons from the CD28 superagonist TGN1412 trial. Nat Rev Immunol 12(5) 317–318. [DOI] [PubMed] [Google Scholar]
  31. Hunig T (2016). The rise and fall of the CD28 superagonist TGN1412 and its return as TAB08: a personal account. FEBS J 283(18) 3325–3334. [DOI] [PubMed] [Google Scholar]
  32. Hutloff A, et al. (1999). ICOS is an inducible T-cell co-stimulator structurally and functionally related to CD28. Nature 397(6716) 263–266. [DOI] [PubMed] [Google Scholar]
  33. Ju SA, et al. (2008). Eradication of established renal cell carcinoma by a combination of 5-fluorouracil and anti-4–1BB monoclonal antibody in mice. Int J Cancer 122(12) 2784–2790. [DOI] [PubMed] [Google Scholar]
  34. June CH, et al. (2018). CAR T cell immunotherapy for human cancer. Science 359(6382) 1361–1365. [DOI] [PubMed] [Google Scholar]
  35. Kawamata S, et al. (1998). Activation of OX40 signal transduction pathways leads to tumor necrosis factor receptor-associated factor (TRAF) 2- and TRAF5-mediated NF-kappaB activation. J Biol Chem 273(10) 5808–5814. [DOI] [PubMed] [Google Scholar]
  36. Keller AM, et al. (2008). Expression of costimulatory ligand CD70 on steady-state dendritic cells breaks CD8+ T cell tolerance and permits effective immunity. Immunity 29(6) 934–946. [DOI] [PubMed] [Google Scholar]
  37. Kim IK, et al. (2015). Glucocorticoid-induced tumor necrosis factor receptor-related protein co-stimulation facilitates tumor regression by inducing IL-9-producing helper T cells. Nat Med 21(9) 1010–1017. [DOI] [PubMed] [Google Scholar]
  38. Kjaergaard J, et al. (2000). Therapeutic efficacy of OX-40 receptor antibody depends on tumor immunogenicity and anatomic site of tumor growth. Cancer Res 60(19) 5514–5521. [PubMed] [Google Scholar]
  39. Klein U, Rajewsky K and Kuppers R (1998). Human immunoglobulin (Ig)M+IgD+ peripheral blood B cells expressing the CD27 cell surface antigen carry somatically mutated variable region genes: CD27 as a general marker for somatically mutated (memory) B cells. J Exp Med 188(9) 1679–1689. [DOI] [PMC free article] [PubMed] [Google Scholar]
  40. Knee DA, Hewes B and Brogdon JL (2016). Rationale for anti-GITR cancer immunotherapy. Eur J Cancer 67 1–10. [DOI] [PubMed] [Google Scholar]
  41. Ko K, et al. (2005). Treatment of advanced tumors with agonistic anti-GITR mAb and its effects on tumor-infiltrating Foxp3+CD25+CD4+ regulatory T cells. J Exp Med 202(7) 885–891. [DOI] [PMC free article] [PubMed] [Google Scholar]
  42. Kohrt HE, et al. (2014). Targeting CD137 enhances the efficacy of cetuximab. J Clin Invest 124(6) 2668–2682. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  43. Krause P, et al. (2009). Prostaglandin E(2) enhances T-cell proliferation by inducing the costimulatory molecules OX40L, CD70, and 4–1BBL on dendritic cells. Blood 113(11) 2451–2460. [DOI] [PubMed] [Google Scholar]
  44. Kwon BS and Weissman SM (1989). cDNA sequences of two inducible T-cell genes. Proc Natl Acad Sci U S A 86(6) 1963–1967. [DOI] [PMC free article] [PubMed] [Google Scholar]
  45. Lee SJ, et al. (2004). 4–1BB and OX40 dual costimulation synergistically stimulate primary specific CD8 T cells for robust effector function. J Immunol 173(5) 3002–3012. [DOI] [PubMed] [Google Scholar]
  46. Lin GH, et al. (2013). GITR-dependent regulation of 4–1BB expression: implications for T cell memory and anti-4–1BB-induced pathology. J Immunol 190(9) 4627–4639. [DOI] [PubMed] [Google Scholar]
  47. Linch SN, et al. (2016). Combination OX40 agonism/CTLA-4 blockade with HER2 vaccination reverses T-cell anergy and promotes survival in tumor-bearing mice. Proc Natl Acad Sci U S A 113(3) E319–327. [DOI] [PMC free article] [PubMed] [Google Scholar]
  48. Mallett S, Fossum S and Barclay AN (1990). Characterization of the MRC OX40 antigen of activated CD4 positive T lymphocytes--a molecule related to nerve growth factor receptor. EMBO J 9(4) 1063–1068. [DOI] [PMC free article] [PubMed] [Google Scholar]
  49. Marin-Acevedo JA, et al. (2018). Next generation of immune checkpoint therapy in cancer: new developments and challenges. J Hematol Oncol 11(1) 39. [DOI] [PMC free article] [PubMed] [Google Scholar]
  50. Martinez-Forero I, et al. (2013). T cell costimulation with anti-CD137 monoclonal antibodies is mediated by K63-polyubiquitin-dependent signals from endosomes. J Immunol 190(12) 6694–6706. [DOI] [PubMed] [Google Scholar]
  51. Matter M, et al. (2006). Elimination of chronic viral infection by blocking CD27 signaling. J Exp Med 203(9) 2145–2155. [DOI] [PMC free article] [PubMed] [Google Scholar]
  52. McHugh RS, et al. (2002). CD4(+)CD25(+) immunoregulatory T cells: gene expression analysis reveals a functional role for the glucocorticoid-induced TNF receptor. Immunity 16(2) 311–323. [DOI] [PubMed] [Google Scholar]
  53. Melero I, et al. (1998). Amplification of tumor immunity by gene transfer of the co-stimulatory 4–1BB ligand: synergy with the CD28 co-stimulatory pathway. Eur J Immunol 28(3) 1116–1121. [DOI] [PubMed] [Google Scholar]
  54. Melero I, et al. (1998). NK1.1 cells express 4–1BB (CDw137) costimulatory molecule and are required for tumor immunity elicited by anti-4–1BB monoclonal antibodies. Cell Immunol 190(2) 167–172. [DOI] [PubMed] [Google Scholar]
  55. Melero I, et al. (2008). Multi-layered action mechanisms of CD137 (4–1BB)-targeted immunotherapies. Trends Pharmacol Sci 29(8) 383–390. [DOI] [PubMed] [Google Scholar]
  56. Melero I, et al. (1997). Monoclonal antibodies against the 4–1BB T-cell activation molecule eradicate established tumors. Nat Med 3(6) 682–685. [DOI] [PubMed] [Google Scholar]
  57. Messenheimer DJ, et al. (2017). Timing of PD-1 Blockade Is Critical to Effective Combination Immunotherapy with Anti-OX40. Clin Cancer Res 23(20) 6165–6177. [DOI] [PMC free article] [PubMed] [Google Scholar]
  58. Morales-Kastresana A, et al. (2013). Combined immunostimulatory monoclonal antibodies extend survival in an aggressive transgenic hepatocellular carcinoma mouse model. Clin Cancer Res 19(22) 6151–6162. [DOI] [PubMed] [Google Scholar]
  59. Muntasell A, et al. (2017). Targeting NK-cell checkpoints for cancer immunotherapy. Curr Opin Immunol 45 73–81. [DOI] [PubMed] [Google Scholar]
  60. Murata S, et al. (2006). OX40 costimulation synergizes with GM-CSF whole-cell vaccination to overcome established CD8+ T cell tolerance to an endogenous tumor antigen. J Immunol 176(2) 974–983. [DOI] [PubMed] [Google Scholar]
  61. Nocentini G, et al. (1997). 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 94(12) 6216–6221. [DOI] [PMC free article] [PubMed] [Google Scholar]
  62. Ochoa MC, et al. (2017). Antibody-dependent cell cytotoxicity: immunotherapy strategies enhancing effector NK cells. Immunol Cell Biol 95(4) 347–355. [DOI] [PubMed] [Google Scholar]
  63. Ohshima Y, et al. (1997). Expression and function of OX40 ligand on human dendritic cells. J Immunol 159(8) 3838–3848. [PubMed] [Google Scholar]
  64. Oosterhoff D, et al. (2013). Intradermal delivery of TLR agonists in a human explant skin model: preferential activation of migratory dendritic cells by polyribosinic-polyribocytidylic acid and peptidoglycans. J Immunol 190(7) 3338–3345. [DOI] [PubMed] [Google Scholar]
  65. Pan PY, et al. (2002). OX40 ligation enhances primary and memory cytotoxic T lymphocyte responses in an immunotherapy for hepatic colon metastases. Mol Ther 6(4) 528–536. [DOI] [PubMed] [Google Scholar]
  66. Paterson DJ, et al. (1987). Antigens of activated rat T lymphocytes including a molecule of 50,000 Mr detected only on CD4 positive T blasts. Mol Immunol 24(12) 1281–1290. [DOI] [PubMed] [Google Scholar]
  67. Piconese S, Valzasina B and Colombo MP (2008). OX40 triggering blocks suppression by regulatory T cells and facilitates tumor rejection. J Exp Med 205(4) 825–839. [DOI] [PMC free article] [PubMed] [Google Scholar]
  68. Rajasekaran N, et al. (2015). Enhancement of antibody-dependent cell mediated cytotoxicity: a new era in cancer treatment. Immunotargets Ther 4 91–100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  69. Ramakrishna V, et al. (2015). Characterization of the human T cell response to in vitro CD27 costimulation with varlilumab. J Immunother Cancer 3 37. [DOI] [PMC free article] [PubMed] [Google Scholar]
  70. Ramakrishnan P, Wang W and Wallach D (2004). Receptor-specific signaling for both the alternative and the canonical NF-kappaB activation pathways by NF-kappaB-inducing kinase. Immunity 21(4) 477–489. [DOI] [PubMed] [Google Scholar]
  71. Redmond WL, Linch SN and Kasiewicz MJ (2014). Combined targeting of costimulatory (OX40) and coinhibitory (CTLA-4) pathways elicits potent effector T cells capable of driving robust antitumor immunity. Cancer Immunol Res 2(2) 142–153. [DOI] [PMC free article] [PubMed] [Google Scholar]
  72. Redmond WL, et al. (2012). Dual anti-OX40/IL-2 therapy augments tumor immunotherapy via IL-2R-mediated regulation of OX40 expression. PLoS One 7(4) e34467. [DOI] [PMC free article] [PubMed] [Google Scholar]
  73. Roberts DJ, et al. (2010). Control of established melanoma by CD27 stimulation is associated with enhanced effector function and persistence, and reduced PD-1 expression of tumor infiltrating CD8(+) T cells. J Immunother 33(8) 769–779. [DOI] [PMC free article] [PubMed] [Google Scholar]
  74. Rogers PR, et al. (2001). OX40 promotes Bcl-xL and Bcl-2 expression and is essential for long-term survival of CD4 T cells. Immunity 15(3) 445–455. [DOI] [PubMed] [Google Scholar]
  75. Ronchetti S, et al. (2007). Glucocorticoid-induced TNFR-related protein lowers the threshold of CD28 costimulation in CD8+ T cells. J Immunol 179(9) 5916–5926. [DOI] [PubMed] [Google Scholar]
  76. Rowley TF and Al-Shamkhani A (2004). Stimulation by soluble CD70 promotes strong primary and secondary CD8+ cytotoxic T cell responses in vivo. J Immunol 172(10) 6039–6046. [DOI] [PubMed] [Google Scholar]
  77. Ruby CE, et al. (2008). IL-12 is required for anti-OX40-mediated CD4 T cell survival. J Immunol 180(4) 2140–2148. [DOI] [PubMed] [Google Scholar]
  78. Sabbagh L, et al. (2008). 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 180(12) 8093–8101. [DOI] [PubMed] [Google Scholar]
  79. Sanmamed MF, et al. (2015). Agonists of Co-stimulation in Cancer Immunotherapy Directed Against CD137, OX40, GITR, CD27, CD28, and ICOS. Semin Oncol 42(4) 640–655. [DOI] [PubMed] [Google Scholar]
  80. Saoulli K, et al. (1998). CD28-independent, TRAF2-dependent costimulation of resting T cells by 4–1BB ligand. J Exp Med 187(11) 1849–1862. [DOI] [PMC free article] [PubMed] [Google Scholar]
  81. Schaer DA, Murphy JT and Wolchok JD (2012). Modulation of GITR for cancer immunotherapy. Curr Opin Immunol 24(2) 217–224. [DOI] [PMC free article] [PubMed] [Google Scholar]
  82. Segal NH, et al. (2018). Phase I Study of Single-Agent Utomilumab (PF-05082566), a 4–1BB/CD137 Agonist, in Patients with Advanced Cancer. Clin Cancer Res [DOI] [PubMed]
  83. Segal NH, et al. (2017). Results from an Integrated Safety Analysis of Urelumab, an Agonist Anti-CD137 Monoclonal Antibody. Clin Cancer Res 23(8) 1929–1936. [DOI] [PubMed] [Google Scholar]
  84. Sharma P and Allison JP (2015). Immune checkpoint targeting in cancer therapy: toward combination strategies with curative potential. Cell 161(2) 205–214. [DOI] [PMC free article] [PubMed] [Google Scholar]
  85. Shi W and Siemann DW (2006). Augmented antitumor effects of radiation therapy by 4–1BB antibody (BMS-469492) treatment. Anticancer Res 26(5A) 3445–3453. [PubMed] [Google Scholar]
  86. Shimizu J, et al. (2002). Stimulation of CD25(+)CD4(+) regulatory T cells through GITR breaks immunological self-tolerance. Nat Immunol 3(2) 135–142. [DOI] [PubMed] [Google Scholar]
  87. Simpson TR, Quezada SA and Allison JP (2010). Regulation of CD4 T cell activation and effector function by inducible costimulator (ICOS). Curr Opin Immunol 22(3) 326–332. [DOI] [PubMed] [Google Scholar]
  88. Snell LM, et al. (2010). CD8 T cell-intrinsic GITR is required for T cell clonal expansion and mouse survival following severe influenza infection. J Immunol 185(12) 7223–7234. [DOI] [PubMed] [Google Scholar]
  89. So T and Croft M (2007). Cutting edge: OX40 inhibits TGF-beta- and antigen-driven conversion of naive CD4 T cells into CD25+Foxp3+ T cells. J Immunol 179(3) 1427–1430. [DOI] [PubMed] [Google Scholar]
  90. So T and Croft M (2013). Regulation of PI-3-Kinase and Akt Signaling in T Lymphocytes and Other Cells by TNFR Family Molecules. Front Immunol 4 139. [DOI] [PMC free article] [PubMed] [Google Scholar]
  91. Song J, et al. (2005). Sustained survivin expression from OX40 costimulatory signals drives T cell clonal expansion. Immunity 22(5) 621–631. [DOI] [PubMed] [Google Scholar]
  92. Srivastava RM, et al. (2017). CD137 Stimulation Enhances Cetuximab-Induced Natural Killer: Dendritic Cell Priming of Antitumor T-Cell Immunity in Patients with Head and Neck Cancer. Clin Cancer Res 23(3) 707–716. [DOI] [PMC free article] [PubMed] [Google Scholar]
  93. Stuber E, et al. (1995). Cross-linking of OX40 ligand, a member of the TNF/NGF cytokine family, induces proliferation and differentiation in murine splenic B cells. Immunity 2(5) 507–521. [DOI] [PubMed] [Google Scholar]
  94. Sugita K, et al. (1992). Participation of the CD27 antigen in the regulation of IL-2-activated human natural killer cells. J Immunol 149(4) 1199–1203. [PubMed] [Google Scholar]
  95. Takeda K, et al. (2000). CD27-mediated activation of murine NK cells. J Immunol 164(4) 1741–1745. [DOI] [PubMed] [Google Scholar]
  96. Tanaka Y, et al. (1985). A glycoprotein antigen detected with new monoclonal antibodies on the surface of human lymphocytes infected with human T-cell leukemia virus type-I (HTLV-I). Int J Cancer 36(5) 549–555. [DOI] [PubMed] [Google Scholar]
  97. Taraban VY, et al. (2006). Requirement for CD70 in CD4+ Th cell-dependent and innate receptor-mediated CD8+ T cell priming. J Immunol 177(5) 2969–2975. [DOI] [PubMed] [Google Scholar]
  98. Tolcher AW, et al. (2017). 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 23(18) 5349–5357. [DOI] [PubMed] [Google Scholar]
  99. Turaj AH, et al. (2017). Antibody Tumor Targeting Is Enhanced by CD27 Agonists through Myeloid Recruitment. Cancer Cell 32(6) 777–791 e776. [DOI] [PMC free article] [PubMed] [Google Scholar]
  100. Turk MJ, et al. (2004). Concomitant tumor immunity to a poorly immunogenic melanoma is prevented by regulatory T cells. J Exp Med 200(6) 771–782. [DOI] [PMC free article] [PubMed] [Google Scholar]
  101. Valzasina B, et al. (2005). 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 105(7) 2845–2851. [DOI] [PubMed] [Google Scholar]
  102. van Lier RA, et al. (1987). Tissue distribution and biochemical and functional properties of Tp55 (CD27), a novel T cell differentiation antigen. J Immunol 139(5) 1589–1596. [PubMed] [Google Scholar]
  103. Vesely MD, et al. (2011). Natural innate and adaptive immunity to cancer. Annu Rev Immunol 29 235–271. [DOI] [PubMed] [Google Scholar]
  104. Vinay DS and Kwon BS (2011). 4–1BB signaling beyond T cells. Cell Mol Immunol 8(4) 281–284. [DOI] [PMC free article] [PubMed] [Google Scholar]
  105. Vinay DS and Kwon BS (2012). Immunotherapy of cancer with 4–1BB. Mol Cancer Ther 11(5) 1062–1070. [DOI] [PubMed] [Google Scholar]
  106. Vitale LA, et al. (2012). Development of a human monoclonal antibody for potential therapy of CD27-expressing lymphoma and leukemia. Clin Cancer Res 18(14) 3812–3821. [DOI] [PubMed] [Google Scholar]
  107. Vonderheide RH and Glennie MJ (2013). Agonistic CD40 antibodies and cancer therapy. Clin Cancer Res 19(5) 1035–1043. [DOI] [PMC free article] [PubMed] [Google Scholar]
  108. Voo KS, et al. (2014). Selective targeting of Toll-like receptors and OX40 inhibit regulatory T-cell function in follicular lymphoma. Int J Cancer 135(12) 2834–2846. [DOI] [PMC free article] [PubMed] [Google Scholar]
  109. Vossen MT, et al. (2008). CD27 defines phenotypically and functionally different human NK cell subsets. J Immunol 180(6) 3739–3745. [DOI] [PubMed] [Google Scholar]
  110. Vu MD, et al. (2007). OX40 costimulation turns off Foxp3+ Tregs. Blood 110(7) 2501–2510. [DOI] [PMC free article] [PubMed] [Google Scholar]
  111. Wang W, et al. (2015). NK Cell-Mediated Antibody-Dependent Cellular Cytotoxicity in Cancer Immunotherapy. Front Immunol 6 368. [DOI] [PMC free article] [PubMed] [Google Scholar]
  112. Weinberg AD, et al. (2000). Engagement of the OX-40 receptor in vivo enhances antitumor immunity. J Immunol 164(4) 2160–2169. [DOI] [PubMed] [Google Scholar]
  113. Weinberg AD, et al. (1999). Blocking OX-40/OX-40 ligand interaction in vitro and in vivo leads to decreased T cell function and amelioration of experimental allergic encephalomyelitis. J Immunol 162(3) 1818–1826. [PubMed] [Google Scholar]
  114. Wilcox RA, et al. (2002). Provision of antigen and CD137 signaling breaks immunological ignorance, promoting regression of poorly immunogenic tumors. J Clin Invest 109(5) 651–659. [DOI] [PMC free article] [PubMed] [Google Scholar]
  115. Willoughby JE, et al. (2014). Differential impact of CD27 and 4–1BB costimulation on effector and memory CD8 T cell generation following peptide immunization. J Immunol 193(1) 244–251. [DOI] [PubMed] [Google Scholar]
  116. Xiao Y, et al. (2004). CD27 is acquired by primed B cells at the centroblast stage and promotes germinal center formation. J Immunol 172(12) 7432–7441. [DOI] [PubMed] [Google Scholar]
  117. Young KH, et al. (2016). Optimizing Timing of Immunotherapy Improves Control of Tumors by Hypofractionated Radiation Therapy. PLoS One 11(6) e0157164. [DOI] [PMC free article] [PubMed] [Google Scholar]
  118. Zhou P, et al. (2007). Pivotal roles of CD4+ effector T cells in mediating agonistic anti-GITR mAb-induced-immune activation and tumor immunity in CT26 tumors. J Immunol 179(11) 7365–7375. [DOI] [PubMed] [Google Scholar]

RESOURCES