Skip to main content
Springer logoLink to Springer
. 2024 Oct 4;43(4):1137–1163. doi: 10.1007/s10555-024-10206-6

Tumor necrosis factor superfamily signaling: life and death in cancer

Obada Ababneh 1,, Daisuke Nishizaki 2, Shumei Kato 2,#, Razelle Kurzrock 3,4,5,✉,#
PMCID: PMC11554763  PMID: 39363128

Abstract

Immune checkpoint inhibitors have shaped the landscape of cancer treatment. However, many patients either do not respond or suffer from later progression. Numerous proteins can control immune system activity, including multiple tumor necrosis factor (TNF) superfamily (TNFSF) and TNF receptor superfamily (TNFRSF) members; these proteins play a complex role in regulating cell survival and death, cellular differentiation, and immune system activity. Notably, TNFSF/TNFRSF molecules may display either pro-tumoral or anti-tumoral activity, or even both, depending on tumor type. Therefore, TNF is a prototype of an enigmatic two-faced mediator in oncogenesis. To date, multiple anti-TNF agents have been approved and/or included in guidelines for treating autoimmune disorders and immune-related toxicities after immune checkpoint blockade for cancer. A confirmed role for the TNFSF/TNFRSF members in treating cancer has proven more elusive. In this review, we highlight the cancer-relevant TNFSF/TNFRSF family members, focusing on the death domain-containing and co-stimulation members and their signaling pathways, as well as their complicated role in the life and death of cancer cells.

Graphical Abstract

graphic file with name 10555_2024_10206_Figa_HTML.jpg

Keywords: TNFSF, TNFRSF, Immunotherapy, Novel immunotherapy targets, Personalized oncology, Tumor necrosis factor, Cancer

Introduction

The clinical development of immune checkpoint inhibitors (ICIs), mainly anti-PD1 or anti-CTLA4 agents, has been a game changer for the therapy of several types of cancer. However, ICIs are not a one-size-fit-all treatment and many patients do not respond or develop resistance to them [1]. Thus, other strategies are being explored, including blocking co-inhibitory checkpoints such as LAG-3, enhancing the activities of immune co-stimulatory receptors, and tumor microenvironment modifications [24]. In this regard, the tumor necrosis factor (TNF) protein family members are of enormous importance in cancer research due to their diverse biological and oncogenic activity and as potential targets for treatment [5].

The TNF proteins are divided into two superfamilies: the TNF receptor superfamily (TNFRSF) and the TNF superfamily (TNFSF) (Table 1) [618]. TNFSF is a group of soluble or membrane-bound proteins that bind to TNFRSF on target cells. There are at least 19 TNFSF ligands and they bind to at least 29 TNFRSFs [19]. Some of the best-known TNFSF ligands include the FAS ligand (FASL) and TNFSF10 (TRAIL). Upon ligand binding, TNFRSF induces intracellular signaling pathways of different TNF receptor-associated factor (TRAF) subtypes, which in turn regulates key cellular biologic processes. These interactions play a crucial role in the orchestration of cell survival, inflammation, proliferation, differentiation, and apoptosis.

Table 1.

Examples of cancer-relevant members of TNFSF and TNFRSF and selected biologic activities

TNF superfamily gene name Protein name TNF receptor superfamily gene name TNF receptor superfamily protein name Typical downstream action and comments References
TNF TNF-alpha TNFRSF1A TNFR1, TNFR2 Induces apoptosis, inflammation, and immune response. Plays an important role in cachexia [6]
TNFSF4 OX40L TNFRSF4 OX40 (CD134) Enhances T cell survival and proliferation [7]
TNFSF5 CD40L TNFRSF5 CD40 Activates B cells, immunoglobulin production and enhances immune response [8]
TNFSF6 FasL (CD95L/CD178) FAS FAS (CD95) Triggers apoptosis and immune system regulation [9]
TNFSF7 CD70 TNFRSF7 CD27 Enhances T cell activation and survival [10]
TNFSF8 CD30L TNFRSF8 CD30 Modulates cell survival and immune response [11]
TNFSF9 4-1BB ligand (CD137L) TNFRSF9 4-1BB (CD137) Enhances T cell proliferation and survival [12]
TNFSF10 TRAIL (CD253) TNFRSF10A, TNFRSF10B, TNFRSF10C, TNFRSF10D DR4 (CD261), DR5 (CD262), DcR1 (CD263), DcR2 (CD264) Induces apoptosis and immune system regulation [13]
TNFSF12 TWEAK TNFRSF12A Fn14 (CD266) Modulates cell survival, proliferation, and inflammation [14]
TNFSF13 APRIL (CD256) TNFRSF13B, TNFRSF17 TACI (CD267), BCMA (CD257) Regulates B-cell survival and antibody production [15]
TNFSF13B BAFF (CD257) TNFRSF13C, TNFRSF17 BAFF-R (CD268), BCMA (CD269) Promotes B cell survival, and maturation proliferation [15]
TNFSF14 LIGHT (CD258) TNFRSF3, TNFRSF14 LTBR (CD18), HVEM (CD270), DcR3 Regulates immune response and inflammation and lymphoid tissue development. Uniquely, HVEM can also bind to two non-TNFSF members of the Ig superfamily, BTLA and CD160 [16]
TNFSF15 TL1A TNFRSF25 DR3 Modulates T cell activation and inflammation in mucosal tissue. Stimulates natural killer cells [17]
TNFSF18 GITRL TNFRSF18 GITR (CD357) Modulates T cell activation and immune response [18]

Abbreviations: APRIL a proliferation-inducing ligand, BAFF B-cell activating factor, BAFF-R BAFF receptor, BCMA B-cell maturation antigen, CD40L CD40 ligand, CD70 CD70 molecule, DcR1 decoy receptor 1, DcR2 decoy receptor 2, DcR3 decoy receptor 3, DR3 death receptor 3, DR4 death receptor 4, DR5 death receptor 5, FasL Fas ligand, Fn14 fibroblast growth factor-inducible 14, GITR glucocorticoid-induced TNFR-related protein, GITRL glucocorticoid-induced TNFR-related protein ligand, HVEM herpesvirus entry mediator, LIGHT lymphotoxin-like, exhibits inducible expression, and competes with HSV glycoprotein D for HVEM, a receptor expressed by T lymphocytes, LTBR lymphotoxin beta receptor, OX40L OX40 ligand, TACI transmembrane activator and calcium modulator and cyclophilin ligand interactor, TL1A TNF-like ligand 1A, TNF-alpha tumor necrosis factor-alpha, TNFR1 tumor necrosis factor receptor 1, TNFR2 tumor necrosis factor receptor 2, TNFRSF tumor necrosis factor receptor superfamily, TNFSF tumor necrosis factor superfamily, TRAIL TNF-related apoptosis-inducing ligand

In this review, we provide a succinct overview of the TNF pathway, including TNFSF/TFNRSF members, their signaling cascades, current and ongoing clinical trials, and the role of anti-TNF in managing immune-related adverse events.

TNF pathways in cancer

In the cancer realm, TNF signaling reflects a two-faced family of molecules. On one hand, TNF family members can function as tumor promoters, because they can stimulate malignant cell proliferation, invasion and metastasis, and tumor angiogenesis. On the other hand, TNF family members can induce cancer cell apoptosis via death domains and other properties. TNFSF and TNFRSF play critical roles in controlling the balance of inflammation [20]. Figure 1 shows some of the interactions between TNFSF and TNFRSF in the tumor microenvironment (TME).

Fig. 1.

Fig. 1

The interaction of TNFSF and TNFRSF and their downstream effectors. Upper panel: the interactions of TNFSF and TNFRSF members in tumor, lymphocytes, and stromal cells. The expression of some of these members results in multiple different effects. Lower panel: the downstream impact of the activation of TNFSF/TNFRSF leads to FADD or TRAF signals. The activation of FADD leads to subsequent cell death while the activation of TRAFs can lead to either cell survival or apoptosis, partially explaining the variability of the biologic impact of some TNFSF/TNFRSF members. Figure created with BioRender.com. Abbreviations: FADD, Fas-associated protein with death domain; TNFSF, tumor necrosis factor superfamily; TNFRSF, tumor necrosis factor receptor superfamily; TRAF, tumor necrosis factor (TNF) receptor-associated factor

Death domain-containing members

A number of TNFRSF molecules can induce programmed cell death—apoptosis—as they contain a death domain. These different proteins share similar downstream signaling after their activation. They often stimulate Fas-associated protein with death domain (FADD) and death-inducing signaling complex (DISC) formation, and they recruit different caspases, mainly caspase 3, 8, and 10 [21].

TNFR1, TNFR2, and TNF

TNF-α has diverse roles in carcinogenesis. In 1975, Carswell et al. discovered a novel protein in the hemorrhagic necrosis of a mouse tumor model, leading to the name “tumor necrosis factor” [22]. TNF binds two receptors: TNFR1, which is expressed in most cells; and TNFR2 which is mainly expressed in hematopoietic and immune cells [23]. TNFR1 is activated by binding to soluble TNF found in blood and extracellular space, while TNFR2 binds to both the transmembrane and soluble form of TNF [24]. The transformation of transmembrane TNF to a soluble TNF is mediated by the metalloprotease TNFα-converting enzyme (TACE) [25].

TNF binding can result in two distinct responses dependent on the activated receptors. Only TNFR1 has a death domain, which enables it to activate the signaling that leads to apoptosis and promotes inflammation [26]. On the other hand, TNFR2 lacks a death domain and instead prompts TNFR-associated factor 2 (TRAF2) recruitment, which mainly promotes cell proliferation and survival, cell activation, and migration [6, 27]. In addition, TNFR2 is expressed by multiple tumor types such as renal cell carcinoma, breast cancer, lung cancer, ovarian cancer, multiple myeloma, and esophageal carcinoma [2730]. The diverse and paradoxical actions of TNF may largely depend on the bound receptor. For instance, while mainly known as an anti-tumor factor (as indicated by its name—“tumor necrosis factor”), Kulbe et al. noticed that TNF secretion by ovarian cancer cells stimulated cytokine release, neo-angiogenesis, and peritoneum invasion, indicating a pro-tumor effect [31]. These findings led to different strategies for targeting TNF in cancer. Additionally, TNFR2 also has a role in carcinogenesis by promoting inflammation. CD8 + effector T cells use TNFR2 for activation and cytotoxicity during the early immune response [32]. TNFR2 can also promote apoptosis signals to terminate the immune response [33]. Moreover, TNFR2 can stimulate the suppressive effects of CD4 + Foxp3 + regulatory T cells (Tregs) and CD8 + Foxp3 + Tregs by inducing their proliferation and activation [34, 35]. Thus, we suspect that TNFR2 works at first to promote inflammation at early carcinogenesis and then it shifts toward a suppressed immune microenvironment to maintain cancer cell survival. He et al. demonstrated that the loss of the TNFR2 allele promotes the development of breast cancer in mice with a more aggressive phenotype and metastatic potential [36]. Such possible effects might be due to promoting autocrine production of TNFα and the preferential activation of the canonical NF-κB signaling pathway. In addition, TNFR2 activation expressed on myeloid-derived suppressor cells was found to promote liver metastasis in murine colon and lung cancer models [37]. Moreover, losing the TNFR2 gene led to decreased lung metastases and Treg infiltration in a melanoma mouse model [38].

Both TNF and anti-TNF molecules have been used in the clinic. For example, in part to attenuate systemic side effects, local infusion strategies utilizing TNF, such as isolated limb perfusion, have been studied. Isolated limb perfusion of TNF with melphalan had activity in metastatic melanoma and unresectable sarcomas [39, 40]. Elia et al. demonstrated that the use of combined modified TNF, ICI, and adoptive cell therapy achieved substantial tumor shrinkage and immune cell infiltration in melanoma and prostate cancer models [41]. On the other side of the camp, the TICIMEL (NTC03293784) trial assessed the benefit of adding an anti-TNF to the standard of care combinational immunotherapy in advanced melanoma. The authors found that the use of concurrent nivolumab, ipilimumab, and a TNF blocker, either infliximab or certolizumab, had a good safety profile and showed activity [42, 43]. However, only a small number of patients were treated; 13 patients received ipilimumab/nivolumab/infliximab and 20 patients ipilimumab/nivolumab/certolizumab. The use of soluble TNF receptors also showed modest activity in cutaneous T-cell lymphomas [44]. Badran et al. reported that in patients with immune-related enterocolitis due to ICI, the use of concurrent ICI and immunosuppressive therapy was associated with a possible decreased risk of enterocolitis recurrence and no treatment effect [45]. Although these studies are limited by their size and design, they offer new strategies to mitigate adverse events without compromising survival. Future studies are needed to investigate the validity of the TNF inhibitor approach and the potential underlying mechanisms. Table 2 summarizes examples of different TNF-α based therapies that reached the clinical trials stage [39, 40, 42, 4649].

Table 2.

Examples of treatment strategies targeting TNF-α in cancer clinical trials

Strategies Major results Tumor types References
Systemic recombinant TNF Systemic single-agent recombinant TNF had low objective response rates Advanced tumors [46, 49]
TNF combined with IFNγ Low objective response rate and significant toxicity Multiple cancer types [47]
Isolated limb infusion/perfusion Objective response rates range between 49 to 90%. This approach is limited by the site of the tumor, the presence of metastases, local toxicity, and the need for specialized centers

Melanoma

Sarcoma

[39, 40]
TNFerade

TNFerade is an adenovector-based gene therapy activated by radiation to induce translation of the human TNF-α gene specifically in cancer cells

In a randomized phase III of locally advanced pancreatic cancer, TNFerade with the standard of care did not show a significant survival benefit compared to the standard of care alone

Advanced solid tumors

Pancreatic cancer

[48]
Anti-TNF drugs TICIMEL explored adding anti-TNF agents’ infliximab or certolizumab to nivolumab and ipilimumab in advanced melanoma. The ORR of the certolizumab and infliximab cohorts were 63% and 46%, respectively. Both combinations were safe with the certolizumab cohort having higher rates of adverse events compared to infliximab Melanoma [42, 43]

Abbreviations: IFN interferon, ORR objective response rate, TNF tumor necrosis factor

Targeting TNFR2 has also been explored. Several preclinical models showed the anti-tumor activity of anti-TNFR2 antibodies in acute myeloid leukemia, ovarian cancer, breast cancer, and colorectal cancer [5053]. The mechanism behind this activity seems to revolve around selective depletion of T regulatory (Treg) cells while sparing T effector (Teff) cells. Thus, anti-TNFR2 could be a future tumor microenvironment (TME)-specific cancer therapy [6]. Clinical trials, such as NCT04752826 and NCT05238883, using anti-TNRF2 in solid tumors are ongoing.

FAS and FAS ligand (FASL)

FASL is another death domain-containing member of the TNFSF family that plays a critical role in immune surveillance and elimination of damaged or infected cells [9]. FASL binds to the FAS receptor, leading to the formation of a death-inducing signaling complex (DISC) and subsequent activation of apoptotic pathways [54]. Dysregulation of the FAS-FASL pathway has been observed in various cancers, contributing to tumor immune escape and resistance to apoptosis [55]. A high FASL expression level was also closely associated with the development of gastric cancer, especially poorly differentiated gastric carcinoma [56]. In some cases, cancer cells can downregulate FAS expression or release soluble FASL, which can act as a decoy receptor and inhibit FAS-mediated apoptosis [5759]. This evasion of cell death can promote tumor survival and progression. Interestingly, some studies suggested a “tumor counterattack” phenomenon where tumor cells expressing FASL can induce apoptosis of FAS-expressing tumor-infiltrating lymphocytes [6062]. Although there is considerable evidence that this counterattack took place, its existence is still debatable [63]. In addition, FASL expressed on the tumor endothelium was found to reduce CD8 T-cell infiltration into the tumor [64]. Moreover, FAS is capable of the maintenance and survival of cancer stem cells (CSCs) and inducing the epithelial-to-mesenchymal transition (EMT) [6568]. Thus, once cancer cells are unresponsive to FAS apoptotic signaling, they can exploit the FAS-mediated non-apoptotic oncogenic functions [9].

Several attempts have been made to develop FAS-agnostic molecules. The use of FAS agonist antibodies systemically administered led to severe hepatotoxicity in treated mice [69]. Another strategy was the use of FASL fusion proteins. ACRP30:FasL, also known as MegaFasL, is a fusion protein of the stalk region of adipocyte complement-related protein (ACRP30) and FasL [70]. CRP30:FasL was found to have an anti-tumor synergistic effect with imatinib in gastrointestinal stromal tumors [71]. NCT00437736 is the only phase I trial investigating MegaFasL in solid tumors. Although the study started in 2007, we could not find any online published report of the trial. CTLA4:FasL and CD40:FasL are other examples of fusion proteins. Orbach et al. showed that both CD40:FasL and CTLA4:FasL can induce cell pro-apoptotic signal of malignant cells of lymphatic origin and inhibit anti-apoptotic proteins: cFLIP, caspase 8, caspase 9, and caspase 3 [72]. We are not aware of any current clinical trials of such strategies.

TRAIL and its receptors

TRAIL, also known as Apo2L, is a member of the TNFSF that selectively induces apoptosis in cancer cells while sparing normal cells [73]. TRAIL binds to two groups of receptors: decoy receptors and death receptors. There are three decoy receptors, DcR1, DcR2, and OPG. DcR1 and DcR2 are surface proteins that block the transmission of the apoptotic signal, while OPG is a soluble secreted receptor that can also mitigate the apoptotic signals [13, 74, 75]. The death receptors DR4 (TRAIL-R1) and DR5 (TRAIL-R2) contain a death domain that leads to the activation of apoptotic signaling cascades upon binding [76]. Thus, TRAIL-mediated apoptosis is regulated by the balance between pro-apoptotic and anti-apoptotic proteins. Critically, naïve T cells are resistant to TRAIL-induced apoptosis [77]. However, T cells may become susceptible to TRAIL-induced apoptosis after repeated or prolonged activation [78]. In TRAIL knockout mice, there is an increased susceptibility to the development of different tumors such as lymphoma, fibrosarcoma breast cancer, and fibrosarcoma [79, 80]. In addition, the use of anti-TRAIL agents led to increased liver metastases in different TRAIL-sensitive cell lines, but this effect was not observed in NK-depleted and IFN-γ models, suggesting the need for both immune competency and TRAIL expression to prevent metastases [81].

Utilizing the TRAIL pathway for therapeutics started with developing TRAIL-receptor agonists (TRAs) as either recombinant TRAIL (rTRAIL) or death receptor agonistic antibodies [82]. Ashkenazi et al. were one of the first to describe the anti-tumor activity of recombinant TRAIL without evoking systematic toxicity in vitro and in vivo [83]. Moreover, other reports showed the efficacy of recombinant TRAIL [84, 85]. Herbst et al. conducted the first phase I, an open-label, dose-escalation, clinical trial of dulanermin, a rTRAIL, in patients with advanced cancer [86]. Although dulanermin was well tolerated, its anticancer activity was limited. Interestingly, two of five patients with chondrosarcoma achieved partial responses. One of the two patients was found to have high BCL-2, protein levels in resistance tissue (limited necrosis) compared to tumor necrotic tissue [87]. Also, dulanermin can bind to both decoy and death receptors, which may lead to diverging activity. In addition, resistance to dulanermin might be driven by alterations to TRAIL receptor 1 (DR4). Horak et al. found low DR4 expression driven by hypermethylation of the DR4 gene [88]. The authors also noted a short mean terminal phase half-life of dulanermin (0.56–1.02 h). Thus, combining dulanermin with other agents such as anti-BCL2 (e.g., venetoclax) and/or hypomethylating agents (such as decitabine and azacitidine) may alleviate resistance to rTRAIL and improve anti-tumor activity. Subsequently, DR5-targeted antibodies (TRAIL-R2 antibodies) were also under development. Lexatumumab (HGS-ETR2), a DR5-targeted antibody, was found to promote DR5 expression and induce apoptosis in renal cell carcinoma in a mice model [89]. In a phase I trial of lexatumumab in patients with advanced cancers, lexatumumab was safe and had a mean plasma half-life of more than 2 weeks at the recommended dose [90]. Overall, no patient achieved an objective response. Mapatumumab, a DR4-targeted antibody, was well tolerated, but no objective responses were observed in a phase I trial in patients with advanced solid tumors [91]. The results of three phase I trials of different DR5-targeted antibodies were reported in 2010: conatumumab, drozitumab, and tigatuzumab [9294]. These trials shared similar safety profiles; however, none of these patients achieved an objective response. These results showed that although first-generation TRAIL receptor antibodies (TRAs) were safe, their clinical value was limited.

Many groups have tried to overcome the limitations of the first-generation TRA molecules, such as limited half-life, and weak agonistic activity for TRAIL-R1 and TRAIL-R2 [82]. A number of protein modifications of rTRAIL are being explored. Such modifications include adding amino acids to the N-terminal such as to the leucine zipper (LZ-TRAIL), isoleucine zipper (IZ-TRAIL), poly-histidine (His-TRAIL), or tenascin-C (TNC) oligomerization domain (TNC-TRAIL) [82]. SCB-313 is a human TRAIL-trimer fusion protein designed by in-frame fusion of the C-terminus of TRAIL and C-propeptide of α1collagen (Trimer-Tag) [95]. Liu et al. showed that TRAIL-trimer is 4–5 times superior compared to native TRAIL with pharmacokinetic and anti-tumor activity [96]. Currently, we are awaiting the results of three Chinese (NCT04051112, NCT04047771, NCT04123886) and two Australian (NCT03443674, NCT03869697) clinical trials assessing SCB-313 in malignant ascites, peritoneal carcinomatosis, malignant pleural effusions, and peritoneal malignancies. Mesenchymal stromal cells (MSCs) expressing TRIAL by a lentiviral (MSCTRAIL) is a new TRAIL-based approach for modulating TME. TACTICAL is an ongoing phase I/II trial to assess the safety and efficacy of MSCTRAIL in combination with first-line standard of care in patients with metastatic lung adenocarcinoma. For more potent signaling, TRAIL-R1/2 needs to trimerize after binding to TRAIL. Eftozanermin alfa (ABBV-621), a hexavalent agonistic fusion protein, is designed to increase receptor clustering independent of FcγR cross-linking [97]. It showed promising anti-tumor activity even in ABBV-621-resistant cells when combined with chemotherapeutics or BCL-XL inhibitors. In a phase I trial, de Jonge et al. showed that Eftozanermin alfa in combination with venetoclax is safe but with potentially limited anti-tumor activity in acute myeloid leukemia and diffuse large B-cell lymphoma [98]. The same group found similar safety, and anti-tumor activity in solid tumors [99]. The baseline frequency of myeloblasts was higher in acute myeloid leukemia patients with progressive disease compared with the only patient who achieved a complete response. On the other hand, myelomonocytes were higher in the patient with complete response. This might be due to the high DR4 and DR5 expression on both myeloblasts and myelomonocytes. At baseline testing, DR4 and DR5 expression was found to be highest in the patient with complete response. Thus, future studies need to also evaluate the expression of TNFSF/TNFRSF genes in the tumor microenvironment in addition to tumor cells to better capture the prognostic ability of these proteins. TAS266 is an agonistic multivalent nanobody based on four high-affinity single variable domains (VHH) that target DR5 [100]. However, its phase I trial was terminated quickly due to severe unexpected hepatotoxicity [100]. The authors noted the presence of pre-existing anti-TAS266 antibodies in patients who developed hepatotoxicity. INBRX-109 is a next-generation agonistic multivalent nanobody that does not bind to anti-TAS266 antibodies found in the previous trial; the response rate was 40.7. None of the patients in the chondrosarcoma had severe transaminitis. These results led to the initiation of ChonDRAgon (NCT04950075) a phase II randomized trial for using INBRX-109 in conventional chondrosarcoma. Such recent advances highlight the potential of TRAIL-based drugs in cancer treatment.

Immune co-stimulation

During the generation of a successful adaptive immune response, many molecular signals are necessary. A primary signal is the binding of cognate antigens to T and B lymphocyte antigen receptors. Secondary signals include the engagement of co-stimulatory molecules expressed by T and B lymphocytes with their respective ligands. As outlined herein, several members of the TNF receptor family act and interact, after initial T cell activation, to sustain T cell responses.

CD-40 and CD-40L

CD40 (TNFRSF5) is a member of the TNFR superfamily that is expressed on a variety of immune cells such as dendritic cells (DC), B cells, and macrophages [8]. In addition, it is found in tumor cells, such as in multiple myeloma, Hodgkin lymphoma, non-Hodgkin lymphoma, renal cell carcinoma, urothelial carcinomas, and ovarian tumors [101, 102]. Its ligand, CD40L (TNFSF5), is expressed on the surface of T cells and natural killer (NK) cells [8]. This interaction induces a variety of immune responses, including the activation of APCs, the proliferation and differentiation of T cells, and the production of cytokines such as IL-12 and IFN-γ and chemokines [102]. This led to the development of agonistic anti-CD40 antibodies to mimic multimeric CD40L on CD40, promoting the recruitment of DCs, B-cell antibody production and T-cell differentiation, and cytotoxic activity [103105].

As mentioned, CD40 expression is found on multiple tumors including bladder cancer, lung cancer, breast cancer, melanoma, colon cancer, acute lymphoblastic leukemia, and non-Hodgkin lymphoma [106]. The anti-tumor effect of CD40 agonism has been shown in many preclinical studies [107109]. For a potent and effective activation of the CD40 pathway, two methods have been developed. Higher order crosslinking of the CD40 monoclonal antibody Fc region leads to the clustering of Fc-gamma receptors IIB (FcγRIIB) [107, 110]. ADC-1013, a fully human agonistic CD40 antibody, was the first to be tested as intra-tumoral therapy in advanced solid malignancies [111]. The trial showed ADC-1013 was safe with some good tumor response of superficial lesions. The second method is based on editing the IgG2 hinge region to provide activation with the need for FcγRIIB crosslinking [112]. Selicrelumab and CDX-1140 are examples of such antibodies [113]. Vonderheide et al. found that selicrelumab, a CD40 agonist monoclonal antibody, was safe but showed limited anti-tumor activity in advanced solid tumors [114]. The authors also noticed a transient yet significant drop in CD19 + B cells in all patients. CD40 and CD40L expression were not evaluated. The authors also did not report if selicrelumab was associated with higher CD8 + T cell infiltration. Increased CD8 + T cell infiltration and activation are expected after receiving CD40-based treatments [113]. In a phase I trial, the use of CDX-1140 with pembrolizumab for patients with PD-1/PD-L1-resistant solid tumors showed one patient with complete response [115]. Sotigalimab is a CD40 monoclonal antibody with an Fc-engineered segment to increase the interaction with FcγRIIB [116]. PRINCE is a randomized phase II trial evaluating the efficacy of sotigalimab and chemotherapy with and without nivolumab in first-line metastatic pancreatic cancer [117]. Surprisingly, the use of triple therapy was inferior in terms of survival compared to nivolumab and chemotherapy and sotigalimab with chemotherapy (1-year overall survival was 41.3%, 48.1%, and 57.7%, respectively). Recently, bi-specific antibodies targeting both CD40 and other tumor antigens have been under investigation. ABBV-428 is a mesothelin-CD40 bi-specific antibody that showed promising results in a preclinical mouse model [118]. Its anti-tumor effect was present in both mesothelin+ and mesothelin tumor cells. Luke et al. reported the safety and efficacy of ABBV-428 in advanced solid cancer patients. Although safe, there were no objective responses [119]. In patients who had more than 90% CD40 receptor occupancy, no patients achieved objective responses. In addition, the baseline tumor expression of mesothelin did not correlate with progression-free survival. There was no clear pattern of CD8 + T cells, and PD-L1 expressing cell changes from baseline to after treatment. Importantly, no patient developed cytokine release syndrome with minimal changes in cytokines levels (IFN-γ, TNF-α, IL-6, IL-1β, IL-2, IL-8, IL-10, and IL-12p70) after ABBV-428 treatment. It should be noted that most patients in these trials have very advanced disease and/or are heavily pre-treated, which might drive the failure to anti-CD40. While the use of CD40-based immunotherapy has not achieved a clinical effect yet, we are still learning from previous pitfalls, improving on current strategies, and awaiting the results of ongoing trials.

OX-40 and OX-40L

OX40, also known as TNFRSF4 (CD134), is transmembrane protein type I expressed on different T cells [120]. It is the sole receptor for OX40 ligand (OX40L). OX40L (TNFSF4) binding to OX40 leads to activating signals of T cells enhancing T-cell survival and pro-inflammatory cytokine production [121]. Similar to agonistic anti-CD40 antibodies, the use of agonistic OX40 antibodies can be theoretically of potential benefit in cancer treatment. Zhang et al. demonstrated that OX40 co-stimulation inhibits FOXP3 gene expression, a marker of Tregs, by increasing the expression of the transcription factors BATF and BATF3 and activating the mTOR pathway [122]. Agonistic OX40 antibodies have shown good anti-tumor responses either as monotherapy or as part of an immunotherapy combination in preclinical tumor models of immunologically active tumors [123126]. It is worth noting that agonistic OX40 antibodies did not show similar results in tumors with poor immunogenicity [127].

Curti et al. conducted the first phase I clinical trial of agonistic OX40 antibodies. They treated 30 patients with metastatic tumors. They found that agonistic OX40 antibodies had an acceptable toxicity with lymphopenia being the most common adverse event and evidence of some nodular regression in 12 patients [128]. OX40 agonist antibodies also led to increased CD4 + FoxP3 − and CD8 + T cell proliferation with no changes to CD4 + FoxP3 − T cells. Davis et al. evaluated the safety and efficacy of INCAGN01949, an OX40 agonist [129]. INCAGN01949 had an acceptable safety profile but with a low disease control rate of 27.6%. In their trial, no predictors of progression were described although the authors noted higher OX40 receptor levels in the blood after receiving INCAGN01949. ENGAGE-1 is a phase I evaluation of the use of the OX40 agonist GSK3174998 with or without pembrolizumab in patients with advanced solid malignancies [130]. The authors found that GSK3174998 is well-tolerated with and without pembrolizumab but with unsatisfactory clinical activity. Notably, the greatest changes were observed for CD134 + cells, NK/NKT cells, and FOXP3 + Tregs. In addition, 39% of patients developed anti-GSK3174998 antibodies. This might partially explain the poor anti-tumor responses in these patients. However, GSK3174998’s ability to modify the tumor microenvironment makes it a feasible candidate to be partnered with other immune checkpoint inhibitors in future studies. Although the aforementioned results did not show promising efficacy, we are still awaiting readouts from a number of trials.

CD70 and CD27

CD27 (TNFRSF7) is constitutively expressed mostly in naive T cells. CD27 binds to CD70 (TNFSF7), which is transiently expressed on activated T cells, DC, and NK cells [10]. CD27 and CD70 binding results in immune cell survival, the expansion of T and B cells, and enhances cellular immunity [131]. CD70 is expressed in primary and metastatic tumor samples [132]. In addition, co-expression of CD27 and CD70 has been detected in different hematological malignancies such as acute myeloid leukemia, acute lymphoblastic leukemia, non-Hodgkin lymphoma, and multiple myeloma [133136]. Subsequently, the role of this duo differs between solid and hematological malignancy. In hematological malignancies, the interaction promotes proliferation and stemness while leading to immune evasion in solid tumors [132]. In addition, the presence of CD27 on solid tumor cells and microenvironment is not essential in promoting the generation and maintenance of CSCs, metastasis, and heterogeneity [137, 138]. The CD27-CD70 interaction also increases the proliferation and reduces apoptosis of Tregs, via inducing CD4 + T cells to produce IL-2 [139]. Moreover, Liu et al. found that CD70 + breast cancer cells have self-renewal and differentiation ability and can metastasize to the lung [140]. Conversely, the loss of CD70 expression promoted cancer lung metastases in melanoma in vivo models [141]. Current treatment strategies revolve around either activating the CD70-CD27 axis of tumor-infiltrating lymphocytes or killing CD70-harboring cells [142].

Antibody–drug conjugates (ADCs) targeting CD70 have been studied in clinical settings. However, the first three ADCs investigated (MDX-1203, AMG 172, and SGN-75) were not well tolerated and no objective responses were seen [132, 143]. NCT04227847 is a phase I trial investigating the safety of SGN-CD70A in myeloid malignancies, a CD70 ADC. Cusatuzumab (ARGX-110) is an afucosylated glycoengineered CD70 antibody that activates both NK cells and complement systems to kill CD70-expressing cells via antibody-dependent cellular cytotoxicity and complement-dependent cytotoxicity, respectively [144]. In a phase I trial, cusatuzumab had a good safety profile but showed limited ant-tumor activity in advanced solid and hematological cancers [145]. There were no overall trends in blood and biochemical biomarkers data over time including CD70 RNA levels and soluble CD27 (sCD27) protein levels. Interestingly, patients with relapsed/refractory cutaneous T-cell lymphoma had a 23% objective response rate [146]. There was a drop in sCD27 levels in about 62% of partial responders in Sezary syndrome patients. Thus, sCD27 level changes might serve as biological biomarkers of response for CD70-based agents in some tumors.

Perhaps, the most exciting CD70-based therapy in early-phase clinical trials is CD70-based CAR-T cell therapy [147]. Other strategies targeting CD27 are also under investigation. In theory, CD27 is less promising due to its expression on non-tumor cells and lower expression on tumor cells compared to CD70 [148]. Varlilumab was the first CD27 agnostic monoclonal antibody to enter clinical trials. In the phase I trial conducted by Burris et al., varlilumab to 10 mg/kg was well tolerated without identification of a maximum tolerated dose. Varlilumab therapy resulted in one partial response in advanced solid tumors [149]. A follow-up phase I/II trial studied the use of varlilumab with nivolumab for advanced solid tumors [150]. The combination was well-tolerated, but the objective response rate for the whole cohort was 10%. A randomized phase II trial evaluating varlilumab combined with nivolumab versus nivolumab alone in B-cell non-Hodgkin lymphoma did not show additional response benefits compared to single-agent nivolumab [151].

4-1BB and 4-1BBL

4-1BB (TNFRSF9; CD137) is expressed on activated T lymphocytes, monocytes, dendritic cells, and natural killer cells. It binds to 4-1BBL (TNFSF9) expressed on antigen-presenting cells (APCs) [12]. Binding can induce CD8 + T cell survival by increasing anti-apoptotic signals of Bcl-XL and Bfl-1 and decreasing the pro-apoptotic singles of Bim [152154]. 4-1BB plays an important role in the anti-tumor activity of the tumor microenvironment [155]. Wilcox et al. found that IL-2 and IL-15 can induce 4-1BB-expressing NK cells which in turn can promote CD8 + T cells responsiveness to IL-2 and IFN-Y secretion [156]. Interestingly, 4-1BB can also lead to Treg proliferation and their differentiation to CD8 + or CD4 + T cells [157, 158]. Thus, 4-1BB agnostics have been an attractive idea for modulating the tumor microenvironment. Melero et al. were the first group to show the feasibility of using 4-1BB antibodies in mice sarcoma models [159]. Curran et al. found that the combination of anti-CTLA-4 and 4-1BB agonists led to an increase in CD8 + and CD4 + T cells in the B16 melanoma model [160]. Similarly, Chen et al. found that anti-PD1 and 4-1BB agonist combination led to tumor regression, IFN-Y and CD8 + T cells in the B16F10 melanoma model [161]. Interestingly, the authors compared that combination with an anti-PD-1 and anti-LAG-3 combination and found that the first had a more profound tumor regression effect compared to the latter. B16 melanoma is considered a poorly immunogenic model, which may suggest that an anti-PD1 and 4-1BB combination may enhance a tumor’s “hotness.”

Urelumab was the first anti-4-1BB agonist to start clinical trials. In the phase I trial reported by Sznol and colleagues, urelumab demonstrated dose-dependent neutropenia and 11% of the patients had grade 3/4 transaminitis [162]. Based on the phase I results, a phase II randomized trial was designed in patients with metastatic melanoma. However, the study was terminated due to grade 5 hepatotoxicity [163]. New clinical trials to combine a lower dose of urelumab with nivolumab, cetuximab, rituximab, and elotuzumab have been announced. However, hepatotoxicity was still present and the trials were halted for now. Utomilumab is another anti-4-1BB agonist with a presumably better safety profile. In the first phase I trial, there was no dose-limiting toxicity or transaminitis [164]. In another phase Ib trial, utomilumab was used in combination with pembrolizumab and showed a good safety profile and a 26.1% response rate [165]. Cohen et al. reported the results of utomilumab in combination with mogamulizumab. The combination was tolerable but with a low response rate of 4.2% [166]. In addition, 4-1BB is a part of the co-stimulatory endodomain in FDA-approved CAR-T cell therapies tisagenlecleucel and isocabtagene maraleucel for patients with B-cell acute lymphoblastic leukemia and large B-cell lymphoma [167]. Thus, 4-1BB has shown previous clinical significance and is potentially will be part of the next generation of immunotherapies.

GITR and GITRL

GITR (TNFRSF18; CD357) is expressed on Tregs at a higher level while it can be found at lower levels on NK cells, B cells, and naïve and memory T cells [18, 168]. In addition, GITR expression rises 24–72 h after first activation on both Tregs and effector T cells [169, 170]. GITR binds to GITRL (TNFSF18) expressed on antigen-presenting cells [171]. As a result, GITR acts as a co-activating receptor and subsequent inflammatory response [172]. Coe et al. showed that the anti-tumor function of DTA-1, an agonistic GITR monoclonal antibody, is strongly associated with Treg depletion but not with their dysfunction [173]. Kim et al. found that tumor regression is mainly dependent on Treg depletion rather than on CD8 + activation [174]. Mitsui et al. studied combining anti-CTLA-4 with anti-GITR and showed further depletion of Treg and higher CD8 + in tumor sites in CMS5a (a murine fibrosarcoma cell line) and CT26 (a murine colon carcinoma cell line) [175]. Villarreal et al. reported the use of anti-PD-1, GITR monoclonal agonistic antibodies and a peptide vaccine led to a significant regression of tumors in about half of the mice [176]. Leyland et al. concluded the GITRL fusion protein had a good anti-tumor effect which was enhanced by anti-OX40 and also when combined with PD-L1, PD-1, or CTLA-4 [177]. AMG228 was the first GITR monoclonal agonistic antibody to enter clinical trials. In the phase I trial, AMG228 was safe and well-tolerated but no biological nor anti-tumor activity was found [178]. Interestingly, in patients with available matched paired tumor biopsies, GITR expression was no longer detectable after AMG228 treatment compared to its presence in pretreatment samples. In addition, there was a decrease in Treg infiltration and GITR expression and increased CD8 + T cells in colorectal tumor samples after AMG228 treatment. No objective responses were seen in other GITR agonistic antibody trials such as those with BMS-986156, GWN323, and MK-4166 [165, 179181]. MEDI1873, a GITRL agonistic antibody was safe but without objective responses in phase I trials of advanced solid tumors [182]. Although GITR-based strategies still did not show sufficient anti-tumor activity, they still demonstrated a level of tumor microenvironment modulation, indicating possible interactions with other immune checkpoint inhibitors.

HVEM and LIGHT

HVEM, also known as TNFR superfamily 14 (TNFRSF14), was discovered for its role in the entry of Herpes Simplex Virus into cells and viral manipulation [183]. HVEM is expressed by many human tissue elements, including naïve T cells, APCs, endothelium, lung, kidney, and liver [16]. HVEM is a receptor for multiple ligands expressed in the immune system including the TNFSF members LIGHT (TNFSF14) and LTα and immunoglobulin superfamily members BTLA and CD160 [184]. Both LIGHT and LTα play a co-stimulatory role by binding to the cysteine-rich domain-2 (CRD2) and CRD3 of HVEM-activating NF-κB signals and subsequent activating lymphocytes. On the other hand, the binding of BTLA and CD160 to CRD1 of HVEM results in inhibiting antigen receptor stimulation in T cells and B cells. Furthermore, BTLA + dendritic cells promote Treg cell differentiation and the induction of peripheral Treg cell tolerance [185].

The expression of HVEM has been found to be upregulated in many tumors including melanoma, gastric cancer, colorectal carcinoma, chronic lymphocytic leukemia (CLL), acute lymphoblastic leukemia (ALL), and multiple myeloma [186, 187]. However, the expression of HVEM and BTLA were downregulated in follicular lymphoma. In 251 follicular lymphoma patients’ samples, Cheung et al. found HVEM to be mutated in 18.3% [188]. The presence of HVEM mutation was associated with worse survival. On the other hand, activation of LTα promotes the formation of tertiary lymphoid structures and protects against lung cancer and melanoma development in mouse models [189]. Due to the dual action of HVEM in regulating the immune system, current therapeutic strategies are directed toward its ligands; LIGHT, BTLA, and CD160.

The expression of LIGHT mediates the remodeling of the TME by provoking IFNγ-mediated apoptosis and vasculature normalization and encouraging the generation of tertiary lymphoid structures [190]. LIGHT is expressed on activated T cells, tumor-sensing NK cells, and immature DC [191, 192]. Different therapeutic strategies are being studied to deliver or induce LIGHT in preclinical models. Yu et al. developed an adenovirus-expressing LIGHT (Ad-LIGHT) and showed its intra-tumoral administration to 4T1 tumor and Ag104Ld fibrosarcoma model mouse model leads to CD8 + T cells tumor infiltration and killing of tumor cells in both the injection and metastatic sites [193]. A similar approach using Myxoma Virus Expressing LIGHT was found to be effective in pancreatic ductal adenocarcinoma and lung metastatic osteosarcoma models [194, 195]. Tang et al. developed a unique fusion protein by modifying fusing multiple monomers of LIGHT to an anti-EGFR monoclonal antibody (anti-EGFR-hmLIGHT) [196]. Anti-EGFR-hmLIGHT was effective in enhancing lymphocyte infiltration and tumor cell killing in Ag104Ld fibrosarcoma and MC38 colon adenocarcinoma mice models. In addition, the combination of anti-PD-L1 and anti-EGFR-hmLIGHT was associated with robust and deep anti-tumor activity in PD-L1-resistant models. However, the anti-tumor effect was less substantial in non-overexpressing EGFR cells. Although we are unaware of any early-phase clinical trials, we are predicting there will be some trials to be initiated soon due to the success of LIGHT-based therapy in preclinical models.

Other strategies include targeting either BTLA or CD160. Lasaro et al. showed that the BTLA/CD160-HVEM pathway blockade induces an anti-tumor effect on large tumors in a genetically modified mouse thyroid cancer model [197]. Sordo-Bahamonde et al. showed that the BTLA blockade increased NK cell-mediated cytotoxicity and depleted leukemic cell numbers in cocultured peripheral blood mononuclear cells from CLL patients [198]. Tifcemalimab, also known as icatolimab, is a first-in-class anti-BTLA monoclonal antibody. In a phase Ia dose escalation study by Schilder et al. (NCT04137900), 25 patients with advanced solid tumors were safely treated with no observed dose-limiting toxicity and with an ORR of 36.8% [199]. Importantly, the co-expression of CD8 and HVEM was associated with better response. Preliminary results of phase I/II trial (NCT05000684) (reported at the ASCO annual meeting in 2023) of tifcemalimab and toripalimab in patients with refractory extensive-stage small-cell lung cancer showed that a combination was well tolerated with an ORR of 26.3% [200]. The ORR was 40.0% in immunotherapy-naïve patients and 8.3% in immunotherapy-treated patients. Preliminary results of a phase I trial of tifcemalimab with or without toripalimab (anti-PD1) in relapsed/refractory lymphomas (NCT04477772) reported a response rate of 42%. Notably, these patients were heavily pre-treated with 66.7% of the patients progressing upon prior anti-PD-1/L1 therapy [201]. Regarding CD160-based therapies, we are only aware of one phase I trial of an anti-CD160 in melanoma (NCT04477876) with no results published so far.

APRIL and BAFF systems

The B-cell activating factor (BAFF) (TNFSF13B; CD257) and a proliferation-inducing ligand (APRIL (TNFSF13)) are unique members of the TNFSF. Both proteins are type II transmembrane proteins with the latter being cleaved by proteases to be secreted in their soluble forms. Both signaling pathways are involved in B-cell survival, maturation, and differentiation [202]. BAFF can bind to BAFF-R, TACI, and weakly to BCMA. APRIL binds strongly to BCMA and moderately to TACI, whereas BAFF binds weakly to BCMA and strongly to TACI. The activation of these receptors on B and T cells enhances survival, co-stimulation of the adaptive immune system of cells, differentiation and maintenance of plasma cells, and proliferation linking it pathogenesis of different malignancies [15]. BAFF-BAFF-R interaction can also drive CD4 + T cells and Treg survival and proliferation via PI3K-Akt pathways [203].

BAFF, APRIL, and their receptors are overexpressed in different hematological malignancies [204]. The role of APRIL differs in solid malignancies as its presence in tumor microenvironment inhibits tumor cell apoptosis, and promotes metastasis [205]. APRIL was overexpressed in different solid tumors including breast cancer, glioma, cervical cancer, bladder cancer, and ovarian cancer [204, 206]. Moreover, the overexpression of APRIL, BCMA, and TACI participates in carcinogenesis in human non-small cell lung cancer and breast cancer by promoting survival signals of p38, ERK1/2, and JNK1/2 [205]. Targeting the BAFF and APRIL pathways and their receptors have been employed in different hematological malignancies. Different treatment strategies have been investigated such as anti-BAFF, anti-APRIL, anti-BAFF-R, and anti-BCMA antibodies, BAFF, APRIL, and BCMA CAR-T cell therapy, and bi-specific antibodies (reviewed in Ref. [207]). Belantamab mafodotin (anti-BCMA) is an antibody–drug conjugate that showed responses in about a third of multiple myeloma patients in phase II randomized, open-label trial, which led to its FDA approval [208]. In 2021, the FDA approved idecabtagene vicleucel, BCMA-based CAR-T therapy for relapsed/refractory multiple myeloma based on the results of the KarMMa trial [209]. In 2022, the FDA approved ciltacabtagene autoleucel, another BCMA-based CAR-T therapy based on the CARTITUDE-1 trial [202]. Ongoing trials of high interest of TNFSF/TNFRSF-based agents are summarized in Table 3.

Table 3.

Selected ongoing clinical trials targeting different TNFSF/TNFRSF members

NCT ID Drug of interest Diagnosis Target Trial design Setting Rationale No. of participants Estimated completion year
NCT03298763 MSCTRAIL Non-small cell lung cancer Mesenchymal stromal cells expressing TRAIL Phase I/II multicenter, randomized double-blind placebo-controlled Combination therapy (pemetrexed/cisplatin chemotherapy) Delivering TRAIL-expressing cells to selectively induce apoptosis 46 2025
NCT04570631 Eftozanermin alfa Relapsed/refractory multiple myeloma TRAIL receptor agonist Phase I non-randomized trial Combination with IV or subcutaneous (SC) bortezomib and oral dexamethasone tablet Fusion protein based on TRAIL to selectively induce apoptosis 40 2024
NCT04950075 INBRX-109 Conventional chondrosarcoma Death receptor 5 (DR5) antibody Phase II randomized, blinded, placebo-controlled trial Single-agent INBRX-109 DR5 activation to induce cell death 201 2024
NCT04807972 ABBV-927 Pancreatic cancer CD40 agonistic antibodies Phase 1b/2, randomized, controlled, open-label study ABBV-927 administered in combination with modified FOLFIRINOX (mFFX) with or without budigalimab Promote immune responses using the CD40 axis 40 2024
NCT03502330 APX005M Melanoma, non-small cell lung cancer, and renal cell carcinoma CD40 agonistic antibodies A Phase I/Ib single-arm trial APX005M in combination with nivolumab and cabiralizumab Promote immune responses using the CD40 axis 42 2027
NCT03739931 mRNA-2752 Relapsed/refractory solid tumor malignancies or lymphoma Lipid nanoparticle encapsulating mRNAs encoding human OX40L, IL-23, and IL-36γ A Phase 1, open-label, multicenter, dose-escalation study Intra-tumoral Injection alone and in combination with immune checkpoint blockade OX40L, IL-23, and IL-36γ pro-inflammatory cytokines to promote immune responses 264 2025
NCT03092856 PF-04518600 Renal cell carcinoma OX40 agonistic antibodies Phase II randomized trial Axitinib with or without anti-OX40 antibody Promote immune responses using the OX40 axis 62 2025
NCT02830724 Anti-hCD70 CAR transduced T cells CD70-expressing cancers Anti-CD70 CAR-T Phase I/II non-randomized trial Administering peripheral blood lymphocytes transduced with CD70-CAR-T cells CD70-specific CAR-T to target CD-70-expressing cancer cells 124 2028
NCT04696731 ALLO-316 Renal cell carcinoma Anti-CD70 CAR-T Phase I single-arm trial Lymphodepletion regimen of fludarabine, cyclophosphamide, and ALLO-647 followed by CD70 CAR-T therapy CD70-specific CAR-T to target CD-70-expressing cancer cells 120 2025
NCT04903873 EU101 Advanced solid tumors 4-1BB agonistic antibodies Phase I/II single-arm trial Single-agent EU101 Promote immune responses using the 4-1BB axis 110 2025
NCT02845323 Urelumab Muscle-invasive urothelial carcinoma of the bladder 4-1BB agonistic antibodies Phase II randomized, blinded, controlled trial Neoadjuvant nivolumab with or without urelumab (4-1BB agonistic antibodies) Promote immune responses using the 4-1BB axis 15 2024
NCT05301764 LVGN6051 Soft tissue sarcoma 4-1BB agonistic antibodies Phase I/II single-arm trial LVGN6051 combined with anlotinib Promote immune responses using the 4-1BB axis 65 2025
NCT05117242 Acasunlimab Non-small cell lung cancer 4-1BB agonistic antibodies Phase II randomized, open-label, controlled trial Acasunlimab with or without pembrolizumab Promote immune responses using the 4-1BB axis 160 2024
NCT05192486 GNC-038 Diffuse large B-cell lymphoma CD19xCD3 × 4-1BBxPD-L1 tetra-specific antibody Phase Ib/II, open-label, multi-center study Single-agent GNC-038 Tetra-specific antibody to activate and enhance immune response and decrease immune system suppuration 20 2024
NCT04225039 INCAGN01876 Glioblastoma GITR agonistic antibodies Phase II non-randomized trial Anti-GITR agonist INCAGN1876 and the PD-1 inhibitor INCMGA00012 in combination with stereotactic radiosurgery followed with/without surgery Promote immune responses using the GITR axis 39 2025
NCT04021043 BMS-986156 Advanced solid tumors GITR agonistic antibodies Phase I/II non-randomized trial Ipilimumab or nivolumab with BMS-986156 and hypofractionated stereotactic radiation therapy Promote immune responses using the GITR axis 68 2024
NCT05891080 JS004 Non-small cell lung cancer BTLA antagonist antibodies Phase II randomized trial Neoadjuvant toripalimab and JS004 combined with platinum-based doublet chemotherapy Promote immune responses by inhibiting the BTLA axis 124 2030
NCT05427396 JS004 Advanced solid tumors BTLA antagonist antibodies Phase I single-arm trial Recombinant humanized anti-btla monoclonal antibody (JS004) injection combined with oripalimab Promote immune responses by inhibiting the BTLA axis 198 2025
NCT05664971 JS004 Non-small cell lung cancer BTLA antagonist antibodies Phase Ib/II single-arm trial Recombinant humanized anti-BTLA monoclonal antibody (JS004) injection combined with toripalimab and with standard chemotherapy Promote immune responses by inhibiting the BTLA axis 240 2024
NCT05069051 Belimumab Chronic lymphocytic leukemia BAFF antagonist antibodies Phase II randomized trial Rituximab/venetoclax with or without belimumab Promote sensitivity to rituximab/venetoclax by inhibiting the BTLA axis 120 2027
NCT05546723 LMY-920 Relapsed/refractory multiple myeloma BAFF-based CAR-T Phase I single-arm trial Single-agent LMY-920 BAFF-specific CAR-T to target BAFF receptors-expressing cancer cells 30 2024
NCT04879043 HDP-101 Relapsed/refractory multiple myeloma BCMA antibody–drug conjugates Phase I/II Single-agent HDP-101 Antibody–drug conjugates targeting BCMA-expressing cells with amanitin (RNA polymerase II inhibitor) as the payload 78 2025

Immunotherapy and treatment of side effects using TNF inhibitors

The frequency of immune-related adverse events (irAEs) varies, depending on the agents used, the number of cycles, and patients’ genetic makeup [210]. Several guidelines were published to guide physicians on irAE management, including those provided by the American Society of Clinical Oncology (ASCO) [211], the European Society for Medical Oncology (ESMO) [212], the National Comprehensive Cancer Network (NCCN) [213], and the Society for Immunotherapy of Cancer (SITC) [214]. Corticosteroids are the first-line therapy against most irAEs. However, prolonged use of corticosteroids can result in a number of adverse effects such as hypertension, hyperglycemia, osteoporosis, and infections [215]. In addition, some patients may not respond to steroid treatment. Notably, the usual second-line option involves TNF inhibitors. Drugs such as infliximab, adalimumab, golimumab, etanercept, and certolizumab are all considered good options in cases of colitis, myocarditis, arthritis, and pneumonitis [216218]. It is still largely unknown if using other TNFSF/TNFRSF inhibitors would result in clinically successful management of irAEs.

Verheijden et al. explored the outcome of patients with severe irAEs and found that the overall survival was lower in patients treated with TNF inhibitors compared to the corticosteroid-only group [219]. However, since this was not a prospective, randomized trial, it is unclear if the worse survival is due to the treatment with TNF inhibitors or due to patients with worse problems receiving the treatment. Wang et al. retrospectively compared the use of corticosteroids alone and corticosteroids with TNF inhibitors in patients with immune-related colitis and found no difference in outcome [220]. The difference between the two studies may be attributed to different follow-up times between patients who required a TNF inhibitor and those who did not as well as the fact that different cancer types were included and neither study was a randomized control trial [221]. Ogusu et al. showed that second-line immunosuppressants including infliximab for steroid-refractory irAEs in patients with lung cancer are effective in 72.2% of the cases [222]. Badran et al. reported patients who had immune-related enterocolitis, re-initiation of immunotherapy with concurrent selective immune inhibitors including anti-TNF agents is safe, reduces severe immune-related even recurrence, and has no negative impact on survival outcomes. [45]. Overall, these results suggest that TNF inhibitors are effective in managing irAEs and unlikely to affect overall survival, though the latter still remains incompletely adjudicated.

TNF-related molecules and their clinical impact beyond cancer

The TNFRSF and TNFSF play important roles in immune function and hence in autoimmune diseases, as well as in the body’s response to a variety of infectious agents.

Autoimmune disease

The role of the TNFSF and TNFRSF molecules has been explored in autoimmune conditions [223]. The TNF-TNFR2 interaction induces T cell activation, expression of adhesion molecules on endothelial cells, and secretion of inflammatory cytokines such as IFNγ and IL-6 in inflammatory bowel disease [224]. TNF also plays a role in rheumatoid arthritis, psoriatic arthritis, and autoimmune uveitis [225]. Interestingly and similar to its double-edge role in cancer, TNF family members may also act to stabilize the disease [226]. However, anti-TNF drugs are still the mainstay of treatment of many autoimmune conditions due to their high efficacy and better safety profile compared to corticosteroids [227]. For example, infliximab is approved for the treatment of inflammatory bowel disease, rheumatoid arthritis, ankylosing spondylitis, psoriasis, and psoriatic arthritis. Several TNF inhibitors are also authorized for skin autoimmune disorders such as psoriasis (infliximab, etanercept, and adalimumab) and for hidradenitis suppurativa (adalimumab), a severe long-term skin condition that produces skin abscesses and scarring [228]. Lastly, the FDA has authorized adalimumab, a TNFα inhibitor, as the sole systemic non-corticosteroid medication for the treatment of non-infectious uveitis [229]. These agents’ FDA-approved indications in autoimmune diseases and their mechanisms of action are summarized in Table 4 [230].

Table 4.

Examples of FDA-approved indications for commonly used anti-TNF drugs in managing immune-related conditions [230]

Drug Indications Structure and target
Infliximab Ankylosing spondylitis, Crohn’s disease, psoriasis, psoriatic arthritis, rheumatoid arthritis, ulcerative colitis Chimeric monoclonal antibody against TNF-α
Etanercept Ankylosing spondylitis, psoriasis, polyarticular juvenile idiopathic arthritis, psoriatic arthritis, rheumatoid arthritis A dimeric fusion protein of TNFR2 and a human IgG1 Fc domain
Adalimumab Ankylosing spondylitis, Crohn’s disease, hidradenitis suppurativa, juvenile idiopathic arthritis, psoriasis, psoriatic arthritis, rheumatoid arthritis, ulcerative colitis, and uveitis Fully humanized monoclonal antibody against TNF-α
Golimumab Ankylosing spondylitis, psoriatic arthritis, rheumatoid arthritis, ulcerative colitis Fully humanized monoclonal antibody against TNF-α
Certolizumab pegol Ankylosing spondylitis, Crohn’s disease, psoriatic arthritis, rheumatoid arthritis Fully humanized PEGylated anti-TNF-α monoclonal antibody

In addition to TNF, OX40, 4-1BB, HVEM, LTβR, DcR3, BAFF, and GITR have been implicated in several autoimmune diseases (further reviewed in [231]). Activation of these co-stimulatory molecules can further recruit and activate lymphocytes and propagate the inflammatory effect of local tissues. Several preclinical and clinical studies have investigated targeting TNFSF/TNFRSF in autoimmune diseases [232]. Belimumab, a BAFF antagonist, was approved for the treatment of systemic lupus erythematosus in 2011 ending a 50-year drought of no new medications in that field [233]. The use of CD40L antagonists has been halted due to the increased risk of thromboembolic events [234]. Blocking GITR has been shown to be an effective strategy in a preclinical arthritis model, but we are not aware of any clinical trials of such agents [235]. Targeting 4-1BB in autoimmune diseases represents a unique situation as an unexpected finding is that 4-1BB agonism can shut off or limit autoimmune and other inflammatory reactions. [236]. This was partially attributed to its up-regulatory effect on Treg cells and subsequently less inflammation [237]. Still, no clinical trial results are currently available. Overall, the success of TNF inhibitors has not been replicated yet with other TNFSF/TNFRSF family members in autoimmune diseases, but ongoing investigations are being carried out to find the silver bullet.

Infectious diseases

There may be an increased risk of opportunistic infections such as tuberculosis and fungal infection in patients treated with TNF blockers, which may in turn be related to the role of TNF in both host defense and immune response [238]. Moreover, there may be an increased risk of herpes zoster in patients receiving TNF blockers for the treatment of rheumatological diseases [238]. HIV, varicella-zoster virus, Epstein–Barr virus, cytomegalovirus, and human papillomavirus may also occur in patients receiving TNF-blocking therapy for chronic inflammatory conditions [239].

Concerning COVID-19, TNF-α is among the early cytokines produced to mediate pro-inflammatory responses and enhance immune cell infiltration after SARS-CoV-2 infections. Yet, TNF-α-mediated inflammation can also cause detrimental tissue damage and promote lung fibrosis, which later results in pneumonia, pulmonary edema, and acute respiratory distress syndrome [240].

Conclusions and future directions

TNF-related molecules have a variety of paradoxical (inhibitory and stimulatory) effects on cancer cells [241]. The TNF proteins form a vast superfamily of receptors and ligands, which act as a communication module for cancer and immunity. These molecules exert a complex array of actions that affect oncogenesis as well as autoimmune diseases and infectious immunity. As a pro-inflammatory cytokine, TNF is secreted by inflammatory cells, which may be involved in inflammation-associated carcinogenesis. TNF exerts its biological functions through activating distinct signaling pathways such as NF-κB and JNK. NF-κB is a major cell survival signal that is anti-apoptotic, while sustained JNK activation contributes to cell death.

Regarding cancer, TNF is therefore two-faced. TNF can act as a tumor promoter because TNF stimulates cancer cells’ growth, proliferation, invasion and metastasis, and tumor angiogenesis. TNF can also induce cancer cell death. The actions of the multiple TNFRSF and TNFSF molecules are similarly complex. This paradoxical effect might be driven by multiple mechanisms. First, the type of tumor may be important with a possible dichotomy between solid and hematological malignancies. The proliferation and activation of immune cells typically drive more anti-tumor activity in the first while, in hematological malignancies, there is exploitation of these mechanisms for the benefit of the malignant cells. Additionally, some TNFRSF members promote different activities. For example, HVEM promotes co-stimulatory signals when bound to LIGHT and LTα, while it promotes inhibitory signals when bound to CD160 and BTLA. Finally, whether the pro- or anti-tumor effect predominates also depends on the cell type involved, including tumor cells, CD8 + cells, CD4 + cells, Treg cells, and other tumor microenvironment cells.

Targeting immune checkpoints CTLA-4, PD-1, PD-L1, or LAG-3 has transformed cancer treatment. However, many tumors do not respond to targeting these checkpoints and patients may suffer from severe immune-related adverse events. In addition to exploring novel co-inhibitory targets, agonistic antibodies to stimulatory molecules such as TNF/TNF receptor family members may offer new treatment strategies for cancer patients. Most trials test next-generation immunotherapy in patients with very aggressive diseases and expected poor prognosis. Testing these drugs in earlier lines, fitter patients, and with ICI combination might be a feasible approach in future studies. Although many of the discussed approaches failed to demonstrate anti-tumor activity, they still showed a favorable TME modulation effect. Thus, it might be reasonable to use some of these drugs to mold the TME into a “hot” microenvironment followed by using ICI.

There is also a need to explore biomarkers for such novel treatments. We found that most trials failed to elucidate biomarker associations with response. As described previously, co-expression of CD8 and HVEM might predict anti-BTLA monoclonal antibody response and drop in sCD27 levels in Sezary syndrome [146], 199. Moreover, beyond the classically FDA-approved biomarkers (PD-L1 level, tumor mutational burden, and microsatellite stability), TNFSF and TNFRSF might be usable as predictive biomarkers for ICI response and survival[242]. Finally, combination approaches may theoretically have synergistic effects, improving the overall anti-tumor impact.

Targeting TNFSF/TNFRSF family members using both agonist and antagonist drugs is being explored in the clinic for patients with cancer. To date, anti-TNF agents have proven effective for treating multiple autoimmune disorders as well as for immune-related toxicities after checkpoint blockade for cancer. The use of specific TNF family member agonists and antagonists for cancer therapy has been less successful to date and may require biomarker selection of patients [243] as has been successfully applied in the precision genomics oncology field [244248].

Author contributions

All of the authors had participated in the preparation of this article. R.K. and S.K. identified the topic and supervised the project. O.A., D.N., and R.K. conceived the structure of the article; O.A. drafted the original draft; D.N., S.K., and R.K. revised the article. All the authors have approved the final version of the manuscript.

Funding

RK is funded in part by 5U01CA180888-08 and 5UG1CA233198-05.

Data availability

No datasets were generated or analysed during the current study.

Declarations

Competing interests

The authors declare no competing interests.

Footnotes

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Shumei Kato and Razelle Kurzrock contributed equally to this study.

Contributor Information

Obada Ababneh, Email: oeababneh185@med.just.edu.jo.

Razelle Kurzrock, Email: rkurzrock@mcw.edu.

References

  • 1.Fountzilas, E., Kurzrock, R., Vo, H. H., & Tsimberidou, A. M. (2021). Wedding of molecular alterations and immune checkpoint blockade: genomics as a matchmaker. JNCI: Journal of the National Cancer Institute,113(12), 1634–1647. 10.1093/JNCI/DJAB067 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Adashek, J. J., Kato, S., Nishizaki, D., Miyashita, H., De, P., Lee, S., … Kurzrock, R. (2023). LAG-3 transcriptomic expression patterns across malignancies: Implications for precision immunotherapeutics. Cancer Medicine, 12(12), 13155–13166. 10.1002/CAM4.6000 [DOI] [PMC free article] [PubMed]
  • 3.Miyashita, H., Kurzrock, R., Bevins, N. J., Thangathurai, K., Lee, S., Pabla, S., … Kato, S. (2023). T-cell priming transcriptomic markers: Implications of immunome heterogeneity for precision immunotherapy. NPJ Genomic Medicine, 8(1). 10.1038/S41525-023-00359-8 [DOI] [PMC free article] [PubMed]
  • 4.Fujiwara, Y., Kato, S., Nesline, M. K., Conroy, J. M., DePietro, P., Pabla, S., & Kurzrock, R. (2022). Indoleamine 2,3-dioxygenase (IDO) inhibitors and cancer immunotherapy. Cancer Treatment Reviews,110, 102461. 10.1016/J.CTRV.2022.102461 [DOI] [PubMed] [Google Scholar]
  • 5.Müller, D. (2023). Targeting co-stimulatory receptors of the TNF superfamily for cancer immunotherapy. BioDrugs,37(1), 21–33. 10.1007/S40259-022-00573-3/TABLES/4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Takahashi, H., Yoshimatsu, G., & Faustman, D. L. (2022). The roles of TNFR2 signaling in cancer cells and the tumor microenvironment and the potency of TNFR2 targeted therapy. Cells,11(12), 1952. 10.3390/CELLS11121952 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Alves Costa Silva, C., Facchinetti, F., Routy, B., & Derosa, L. (2020). New pathways in immune stimulation: Targeting OX40. ESMO Open,5(1), e000573. 10.1136/ESMOOPEN-2019-000573 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Elgueta, R., Benson, M. J., De Vries, V. C., Wasiuk, A., Guo, Y., & Noelle, R. J. (2009). Molecular mechanism and function of CD40/CD40L engagement in the immune system. Immunological Reviews,229(1), 152–172. 10.1111/J.1600-065X.2009.00782.X [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Peter, M. E., Hadji, A., Murmann, A. E., Brockway, S., Putzbach, W., Pattanayak, A., & Ceppi, P. (2015). The role of CD95 and CD95 ligand in cancer. Cell Death and Differentiation,22(4), 549–559. 10.1038/cdd.2015.3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Borst, J., Hendriks, J., & Xiao, Y. (2005). CD27 and CD70 in T cell and B cell activation. Current Opinion in Immunology,17(3), 275–281. 10.1016/J.COI.2005.04.004 [DOI] [PubMed] [Google Scholar]
  • 11.Kennedy, M. K., Willis, C. R., & Armitage, R. J. (2006). Deciphering CD30 ligand biology and its role in humoral immunity. Immunology,118(2), 143. 10.1111/J.1365-2567.2006.02354.X [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Kim, A. M. J., Nemeth, M. R., & Lim, S. O. (2022). 4–1BB: A promising target for cancer immunotherapy. Frontiers in Oncology,12, 968360. 10.3389/FONC.2022.968360/PDF [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Wajant, H. (2019). Molecular mode of action of TRAIL receptor agonists-common principles and their translational exploitation. Cancers,11(7), 954. 10.3390/CANCERS11070954 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Burkly, L. C., Michaelson, J. S., & Zheng, T. S. (2011). TWEAK/Fn14 pathway: An immunological switch for shaping tissue responses. Immunological Reviews,244(1), 99–114. 10.1111/J.1600-065X.2011.01054.X [DOI] [PubMed] [Google Scholar]
  • 15.Kampa, M., Notas, G., Stathopoulos, E. N., Tsapis, A., & Castanas, E. (2020). The TNFSF members APRIL and BAFF and their receptors TACI, BCMA, and BAFFR in oncology, with a special focus in breast cancer. Frontiers in Oncology,10, 827. 10.3389/FONC.2020.00827/PDF [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Cai, G., & Freeman, G. J. (2009). The CD160, BTLA, LIGHT/HVEM pathway: A bidirectional switch regulating T-cell activation. Immunological Reviews,229(1), 244–258. 10.1111/J.1600-065X.2009.00783.X [DOI] [PubMed] [Google Scholar]
  • 17.Valatas, V., Kolios, G., & Bamias, G. (2019). TL1A (TNFSF15) and DR3 (TNFRSF25): A co-stimulatory system of cytokines with diverse functions in gut mucosal immunity. Frontiers in Immunology,10(22), 421466. 10.3389/FIMMU.2019.00583/BIBTEX [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Clouthier, D. L., & Watts, T. H. (2014). Cell-specific and context-dependent effects of GITR in cancer, autoimmunity, and infection. Cytokine and Growth Factor Reviews,25(2), 91–106. 10.1016/J.CYTOGFR.2013.12.003 [DOI] [PubMed] [Google Scholar]
  • 19.Dostert, C., Grusdat, M., Letellier, E., & Brenner, D. (2019). The TNF family of ligands and receptors: Communication modules in the immune system and beyond. Physiological Reviews,99(1), 115–160. 10.1152/PHYSREV.00045.2017/ASSET/IMAGES/LARGE/Z9J0041828770007.JPEG [DOI] [PubMed] [Google Scholar]
  • 20.So, T., & Ishii, N. (2019). The TNF-TNFR family of co-signal molecules. Advances in Experimental Medicine and Biology,1189, 53–84. 10.1007/978-981-32-9717-3_3 [DOI] [PubMed] [Google Scholar]
  • 21.Clement, M. V., & Stamenkovic, I. (1994). Fas and tumor necrosis factor receptor-mediated cell death: similarities and distinctions. The Journal of Experimental Medicine,180(2), 557–567. 10.1084/JEM.180.2.557 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Carswell, E. A., Old, L. J., Kassel, R. L., Green, S., Fiore, N., & Williamson, B. (1975). An endotoxin-induced serum factor that causes necrosis of tumors. Proceedings of the National Academy of Sciences,72(9), 3666–3670. 10.1073/PNAS.72.9.3666 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Balkwill, F. (2009). Tumour necrosis factor and cancer. Nature Reviews Cancer,9(5), 361–371. 10.1038/NRC2628 [DOI] [PubMed] [Google Scholar]
  • 24.Wajant, H., Pfizenmaier, K., & Scheurich, P. (2003). Tumor necrosis factor signaling. Cell Death and Differentiation,10(1), 45–65. 10.1038/SJ.CDD.4401189 [DOI] [PubMed] [Google Scholar]
  • 25.Wajant, H. (2015). Principles of antibody-mediated TNF receptor activation. Cell Death and Differentiation,22(11), 1727–1741. 10.1038/cdd.2015.109 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Vanamee, É. S., & Faustman, D. L. (2018). Structural principles of tumor necrosis factor superfamily signaling. Science Signaling,11(511), eaao4910. 10.1126/SCISIGNAL.AAO4910 [DOI] [PubMed] [Google Scholar]
  • 27.Shi, G., & Hu, Y. (2023). TNFR1 and TNFR2, which link NF-κB activation, drive lung cancer progression, cell dedifferentiation, and metastasis. Cancers,15(17), 4299. 10.3390/CANCERS15174299 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Kartikasari, A. E. R., Cassar, E., Razqan, M. A. M., Szydzik, C., Huertas, C. S., Mitchell, A., & Plebanski, M. (2022). Elevation of circulating TNF receptor 2 in cancer: A systematic meta-analysis for its potential as a diagnostic cancer biomarker. Frontiers in Immunology,13, 918254. 10.3389/FIMMU.2022.918254/BIBTEX [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Li, M., Zhang, X., Bai, X., & Liang, T. (2022). Targeting TNFR2: A novel breakthrough in the treatment of cancer. Frontiers in Oncology,12, 862154. 10.3389/FONC.2022.862154/BIBTEX [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Yang, Y., Islam, M. S., Hu, Y., & Chen, X. (2021). TNFR2: Role in cancer immunology and immunotherapy. ImmunoTargets and Therapy,10, 103–122. 10.2147/ITT.S255224 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Kulbe, H., Thompson, R., Wilson, J. L., Robinson, S., Hagemann, T., Fatah, R., … Balkwill, F. (2007). The inflammatory cytokine tumor necrosis factor-alpha generates an autocrine tumor-promoting network in epithelial ovarian cancer cells. Cancer Research, 67(2), 585–592. 10.1158/0008-5472.CAN-06-2941 [DOI] [PMC free article] [PubMed]
  • 32.Kim, E. Y., & Teh, H.-S. (2001). TNF type 2 receptor (p75) lowers the threshold of T cell activation. Journal of Immunology (Baltimore, Md. : 1950),167(12), 6812–6820. 10.4049/JIMMUNOL.167.12.6812 [DOI] [PubMed] [Google Scholar]
  • 33.Twu, Y. C., Gold, M. R., & Teh, H. S. (2011). TNFR1 delivers pro-survival signals that are required for limiting TNFR2-dependent activation-induced cell death (AICD) in CD8+ T cells. European Journal of Immunology,41(2), 335–344. 10.1002/EJI.201040639 [DOI] [PubMed] [Google Scholar]
  • 34.Chakraborty, S., Panda, A. K., Bose, S., Roy, D., Kajal, K., Guha, D., & Sa, G. (2017). Transcriptional regulation of FOXP3 requires integrated activation of both promoter and CNS regions in tumor-induced CD8+ Treg cells. Scientific Reports,7(1), 1628. 10.1038/S41598-017-01788-Z [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Chen, X., Wu, X., Zhou, Q., Howard, O. M. Z., Netea, M. G., & Oppenheim, J. J. (2013). TNFR2 is critical for the stabilization of the CD4+Foxp3+ regulatory T cell phenotype in the inflammatory environment. Journal of Immunology (Baltimore, Md. : 1950),190(3), 1076–1084. 10.4049/JIMMUNOL.1202659 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.He, L., Bhat, K., Duhacheck-Muggy, S., Ioannidis, A., Zhang, L., Nguyen, N. T., … Pajonk, F. (2021). Tumor necrosis factor receptor signaling modulates carcinogenesis in a mouse model of breast cancer. Neoplasia (New York, N.Y.), 23(2), 197–209. 10.1016/J.NEO.2020.12.007 [DOI] [PMC free article] [PubMed]
  • 37.Ham, B., Wang, N., D’Costa, Z., Fernandez, M. C., Bourdeau, F., Auguste, P., … Brodt, P. (2015). TNF receptor-2 facilitates an immunosuppressive microenvironment in the liver to promote the colonization and growth of hepatic metastases. Cancer Research, 75(24), 5235–5245. 10.1158/0008-5472.CAN-14-3173 [DOI] [PubMed]
  • 38.Chopra, M., Riedel, S. S., Biehl, M., Krieger, S., von Krosigk, V., Bäuerlein, C. A., … Beilhack, A. (2013). Tumor necrosis factor receptor 2-dependent homeostasis of regulatory T cells as a player in TNF-induced experimental metastasis. Carcinogenesis, 34(6), 1296–1303. 10.1093/CARCIN/BGT038 [DOI] [PubMed]
  • 39.Deroose, J. P., Grünhagen, D. J., Van Geel, A. N., De Wilt, J. H. W., Eggermont, A. M. M., & Verhoef, C. (2011). Long-term outcome of isolated limb perfusion with tumour necrosis factor-α for patients with melanoma in-transit metastases. British Journal of Surgery,98(11), 1573–1580. 10.1002/BJS.7621 [DOI] [PubMed] [Google Scholar]
  • 40.Noorda, E. M., Vrouenraets, B. C., Nieweg, O. E., Van Coevorden, F., Van Slooten, G. W., & Kroon, B. B. R. (2003). Isolated limb perfusion with tumor necrosis factor-alpha and melphalan for patients with unresectable soft tissue sarcoma of the extremities. Cancer,98(7), 1483–1490. 10.1002/CNCR.11648 [DOI] [PubMed] [Google Scholar]
  • 41.Elia, A. R., Grioni, M., Basso, V., Curnis, F., Freschi, M., Corti, A., … Bellone, M. (2018). Targeting tumor vasculature with TNF leads effector t cells to the tumor and enhances therapeutic efficacy of immune checkpoint blockers in combination with adoptive cell therapy. Clinical Cancer Research : An Official Journal of the American Association for Cancer Research, 24(9), 2171–2181. 10.1158/1078-0432.CCR-17-2210 [DOI] [PubMed]
  • 42.Montfort, A., Filleron, T., Virazels, M., Dufau, C., Milhes, J., Pages, C., … Meyer, N. (2021). Combining nivolumab and ipilimumab with infliximab or certolizumab in patients with advanced melanoma: First results of a phase Ib clinical trial. Clinical Cancer Research : An Official Journal of the American Association for Cancer Research, 27(4), 1037–1047. 10.1158/1078-0432.CCR-20-3449 [DOI] [PubMed]
  • 43.Meyer, N., Lusque, A., Virazels, M., Filleron, T., Colacios, C., Montfort, A., & Ségui, B. (2022). 846P Triple combination of ipilimumab + nivolumab + anti-TNF in treatment naive melanoma patients: Final analysis of TICIMEL, a phase Ib prospective clinical trial. Annals of Oncology,33, S936–S937. 10.1016/J.ANNONC.2022.07.972 [Google Scholar]
  • 44.Tsimberidou, A. M., Giles, F. J., Duvic, M., & Kurzrock, R. (2004). Pilot study of etanercept in patients with relapsed cutaneous T-cell lymphomas. Journal of the American Academy of Dermatology,51(2), 200–204. 10.1016/J.JAAD.2003.05.009 [DOI] [PubMed] [Google Scholar]
  • 45.Badran, Y. R., Zou, F., Durbin, S. M., Dutra, B. E., Abu-Sbeih, H., Thomas, A. S., … Dougan, M. (2023). Concurrent immune checkpoint inhibition and selective immunosuppressive therapy in patients with immune-related enterocolitis. Journal for ImmunoTherapy of Cancer, 11(6), e007195. 10.1136/JITC-2023-007195 [DOI] [PMC free article] [PubMed]
  • 46.Roberts, N. J., Zhou, S., Diaz, L. A., Holdhoff, M., Roberts, N. J., Zhou, S., … Holdhoff, M. (2011). Systemic use of tumor necrosis factor alpha as an anticancer agent. Oncotarget, 2(10), 739–751. 10.18632/ONCOTARGET.344 [DOI] [PMC free article] [PubMed]
  • 47.Shen, J., Xiao, Z., Zhao, Q., Li, M., Wu, X., Zhang, L., … Cho, C. H. (2018). Anti-cancer therapy with TNFα and IFNγ: A comprehensive review. Cell Proliferation, 51(4), e12441. 10.1111/CPR.12441 [DOI] [PMC free article] [PubMed]
  • 48.Kali, A. (2015). TNFerade, an innovative cancer immunotherapeutic. Indian Journal of Pharmacology,47(5), 479–483. 10.4103/0253-7613.165190 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Feinberg, B., Kurzrock, R., Talpaz, M., Blick, M., Saks, S., & Gutterman, J. U. (1988). A phase I trial of intravenously-administered recombinant tumor necrosis factor-alpha in cancer patients. Journal of Clinical Oncology : Official Journal of the American Society of Clinical Oncology,6(8), 1328–1334. 10.1200/JCO.1988.6.8.1328 [DOI] [PubMed] [Google Scholar]
  • 50.Govindaraj, C., Tan, P., Walker, P., Wei, A., Spencer, A., & Plebanski, M. (2014). Reducing TNF receptor 2+ regulatory T cells via the combined action of azacitidine and the HDAC inhibitor, panobinostat for clinical benefit in acute myeloid leukemia patients. Clinical Cancer Research : An Official Journal of the American Association for Cancer Research,20(3), 724–735. 10.1158/1078-0432.CCR-13-1576 [DOI] [PubMed] [Google Scholar]
  • 51.Torrey, H., Butterworth, J., Mera, T., Okubo, Y., Wang, L., Baum, D., … Faustman, D. L. (2017). Targeting TNFR2 with antagonistic antibodies inhibits proliferation of ovarian cancer cells and tumor-associated Tregs. Science Signaling, 10(462). 10.1126/SCISIGNAL.AAF8608/SUPPL_FILE/AAF8608_SM.PDF [DOI] [PubMed]
  • 52.Nie, Y., He, J., Shirota, H., Trivett, A. L., Yang, D., Klinman, D. M., … Chen, X. (2018). Blockade of TNFR2 signaling enhances the immunotherapeutic effect of CpG ODN in a mouse model of colon cancer. Science Signaling, 11(511). 10.1126/SCISIGNAL.AAN0790 [DOI] [PMC free article] [PubMed]
  • 53.Case, K., Tran, L., Yang, M., Zheng, H., Kuhtreiber, W. M., & Faustman, D. L. (2020). TNFR2 blockade alone or in combination with PD-1 blockade shows therapeutic efficacy in murine cancer models. Journal of Leukocyte Biology,107(6), 981–991. 10.1002/JLB.5MA0420-375RRRRR [DOI] [PubMed] [Google Scholar]
  • 54.O’Brien, D. I., Nally, K., Kelly, R. G., O’Connor, T. M., Shanahan, F., & O’Connell, J. (2005). Targeting the Fas/Fas ligand pathway in cancer. Expert Opinion on Therapeutic Targets,9(5), 1031–1044. 10.1517/14728222.9.5.1031 [DOI] [PubMed] [Google Scholar]
  • 55.Abrahams, V. M., Kamsteeg, M., & Mor, G. (2003). The Fas/Fas ligand system and cancer: Immune privilege and apoptosis. Molecular Biotechnology,25(1), 19–30. 10.1385/MB:25:1:19 [DOI] [PubMed] [Google Scholar]
  • 56.Li, Q., Peng, J., Li, X. H., Liu, T., Liang, Q. C., & Zhang, G. Y. (2010). Clinical significance of Fas and FasL protein expression in gastric carcinoma and local lymph node tissues. World Journal of Gastroenterology,16(10), 1274–1278. 10.3748/WJG.V16.I10.1274 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Cheng, J., Zhou, T., Liu, C., Shapiro, J. P., Brauer, M. J., Kiefer, M. C., … Mountz, J. D. (1994). Protection from Fas-mediated apoptosis by a soluble form of the Fas molecule. Science (New York, N.Y.), 263(5154), 1759–1762. 10.1126/SCIENCE.7510905 [DOI] [PubMed]
  • 58.Owen-Schaub, L. B., Angelo, L. S., Radinsky, R., Ware, C. F., Gesner, T. G., & Bartos, D. P. (1995). Soluble Fas/APO-1 in tumor cells: A potential regulator of apoptosis? Cancer Letters,94(1), 1–8. 10.1016/0304-3835(95)03834-J [DOI] [PubMed] [Google Scholar]
  • 59.Pitti, R. M., Marsters, S. A., Lawrence, D. A., Roy, M., Kischkel, F. C., Dowd, P., … Ashkenazi, A. (1998). Genomic amplification of a decoy receptor for Fas ligand in lung and colon cancer. Nature, 396(6712), 699–703. 10.1038/25387 [DOI] [PubMed]
  • 60.Hahne, M., Rimoldi, D., Schröter, M., Romero, P., Schreier, M., French, L. E., … Tschopp, J. (1996). Melanoma cell expression of Fas(Apo-1/CD95) ligand: Implications for tumor immune escape. Science (New York, N.Y.), 274(5291), 1363–1366. 10.1126/SCIENCE.274.5291.1363 [DOI] [PubMed]
  • 61.Strand, S., Hofmann, W. J., Hug, H., Müller, M., Otto, G., Strand, D., … Galle, P. R. (1996). Lymphocyte apoptosis induced by CD95 (APO-1/Fas) ligand-expressing tumor cells--A mechanism of immune evasion? Nature Medicine, 2(12), 1361–1366. 10.1038/NM1296-1361 [DOI] [PubMed]
  • 62.O’Connell, J., O’Sullivan, G. C., Collins, J. K., & Shanahan, F. (1996). The Fas counterattack: Fas-mediated T cell killing by colon cancer cells expressing Fas ligand. The Journal of Experimental Medicine,184(3), 1075–1082. 10.1084/JEM.184.3.1075 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Igney, F. H., & Krammer, P. H. (2005). Tumor counterattack: Fact or fiction? Cancer Immunology, Immunotherapy : CII,54(11), 1127–1136. 10.1007/S00262-005-0680-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Motz, G. T., Santoro, S. P., Wang, L. P., Garrabrant, T., Lastra, R. R., Hagemann, I. S., … Coukos, G. (2014). Tumor endothelium FasL establishes a selective immune barrier promoting tolerance in tumors. Nature Medicine, 20(6), 607–615. 10.1038/NM.3541 [DOI] [PMC free article] [PubMed]
  • 65.Ceppi, P., Hadji, A., Kohlhapp, F. J., Pattanayak, A., Hau, A., Liu, X., … Peter, M. E. (2014). CD95 and CD95L promote and protect cancer stem cells. Nature Communications, 5. 10.1038/NCOMMS6238 [DOI] [PMC free article] [PubMed]
  • 66.Teodorczyk, M., Kleber, S., Wollny, D., Sefrin, J. P., Aykut, B., Mateos, A., … Martin-Villalba, A. (2015). CD95 promotes metastatic spread via Sck in pancreatic ductal adenocarcinoma. Cell Death and Differentiation, 22(7), 1192–1202. 10.1038/CDD.2014.217 [DOI] [PMC free article] [PubMed]
  • 67.Zheng, H., Li, W., Wang, Y., Liu, Z., Cai, Y., Xie, T., … Jiang, B. (2013). Glycogen synthase kinase-3 beta regulates snail and β-catenin expression during Fas-induced epithelial-mesenchymal transition in gastrointestinal cancer. European Journal of Cancer (Oxford, England : 1990), 49(12), 2734–2746. 10.1016/J.EJCA.2013.03.014 [DOI] [PubMed]
  • 68.Zheng, H. X., Cai, Y. D., Wang, Y. D., Cui, X. B., Xie, T. T., Li, W. J., … Jiang, B. (2013). Fas signaling promotes motility and metastasis through epithelial-mesenchymal transition in gastrointestinal cancer. Oncogene, 32(9), 1183–1192. 10.1038/ONC.2012.126 [DOI] [PubMed]
  • 69.Ogasawara, J., Watanabe-Fukunaga, R., Adachi, M., Matsuzawa, A., Kasugai, T., Kitamura, Y., … Nagata, S. (1993). Lethal effect of the anti-Fas antibody in mice. Nature, 364(6440), 806–809. 10.1038/364806A0 [DOI] [PubMed]
  • 70.Greaney, P., Nahimana, A., Lagopoulos, L., Etter, A. L., Aubry, D., Attinger, A., … Duchosal, M. A. (2006). A Fas agonist induces high levels of apoptosis in haematological malignancies. Leukemia Research, 30(4), 415–426. 10.1016/J.LEUKRES.2005.08.006 [DOI] [PubMed]
  • 71.Rikhof, B., Van Der Graaf, W. T. A., Meijer, C., Le, P. T. K., Meersma, G. J., De Jong, S., … Suurmeijer, A. J. H. (2008). Abundant Fas expression by gastrointestinal stromal tumours may serve as a therapeutic target for MegaFasL. British Journal of Cancer, 99(10), 1600–1606. 10.1038/SJ.BJC.6604736 [DOI] [PMC free article] [PubMed]
  • 72.Orbach, A., Rachmilewitz, J., Shani, N., Isenberg, Y., Parnas, M., Huang, J. H., … Dranitzki-Elhalel, M. (2010). CD40·FasL and CTLA-4·FasL fusion proteins induce apoptosis in malignant cell lines by dual signaling. The American Journal of Pathology, 177(6), 3159–3168. 10.2353/AJPATH.2010.100301 [DOI] [PMC free article] [PubMed]
  • 73.Wiley, S. R., Schooley, K., Smolak, P. J., Din, W. S., Huang, C. P., Nicholl, J. K., … Goodwin, R. G. (1995). Identification and characterization of a new member of the TNF family that induces apoptosis. Immunity, 3(6), 673–682. 10.1016/1074-7613(95)90057-8 [DOI] [PubMed]
  • 74.Yuan, X., Gajan, A., Chu, Q., Xiong, H., Wu, K., & Wu, G. S. (2018). Developing TRAIL/TRAIL death receptor-based cancer therapies. Cancer Metastasis Reviews,37(4), 733–748. 10.1007/S10555-018-9728-Y [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Lamoureux, F., Moriceau, G., Picarda, G., Rousseau, J., Trichet, V., & Rédini, F. (2010). Regulation of osteoprotegerin pro- or anti-tumoral activity by bone tumor microenvironment. Biochimica et Biophysica Acta,1805(1), 17–24. 10.1016/J.BBCAN.2009.08.004 [DOI] [PubMed] [Google Scholar]
  • 76.Pimentel, J. M., Zhou, J. Y., & Wu, G. S. (2023). The role of TRAIL in apoptosis and immunosurveillance in cancer. Cancers,15(10), 2752. 10.3390/CANCERS15102752 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Mirandola, P., Ponti, C., Gobbi, G., Sponzilli, I., Vaccarezza, M., Cocco, L., … Vitale, M. (2004). Activated human NK and CD8+ T cells express both TNF-related apoptosis-inducing ligand (TRAIL) and TRAIL receptors but are resistant to TRAIL-mediated cytotoxicity. Blood, 104(8), 2418–2424. 10.1182/BLOOD-2004-04-1294 [DOI] [PubMed]
  • 78.Sag, D., Ayyildiz, Z. O., Gunalp, S., & Wingender, G. (2019). The role of TRAIL/DRs in the modulation of immune cells and responses. Cancers,11(10), 1469. 10.3390/CANCERS11101469 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Zerafa, N., Westwood, J. A., Cretney, E., Mitchell, S., Waring, P., Iezzi, M., & Smyth, M. J. (2005). Cutting edge: TRAIL deficiency accelerates hematological malignancies. Journal of Immunology (Baltimore, Md. : 1950),175(9), 5586–5590. 10.4049/JIMMUNOL.175.9.5586 [DOI] [PubMed] [Google Scholar]
  • 80.Cretney, E., Takeda, K., Yagita, H., Glaccum, M., Peschon, J. J., & Smyth, M. J. (2002). Increased susceptibility to tumor initiation and metastasis in TNF-related apoptosis-inducing ligand-deficient mice. Journal of Immunology (Baltimore, Md. : 1950),168(3), 1356–1361. 10.4049/JIMMUNOL.168.3.1356 [DOI] [PubMed] [Google Scholar]
  • 81.Takeda, K., Hayakawa, Y., Smyth, M. J., Kayagaki, N., Yamaguchi, N., Kakuta, S., … Okumura, K. (2001). Involvement of tumor necrosis factor-related apoptosis-inducing ligand in surveillance of tumor metastasis by liver natural killer cells. Nature Medicine, 7(1), 94–100. 10.1038/83416 [DOI] [PubMed]
  • 82.Di Cristofano, F., George, A., Tajiknia, V., Ghandali, M., Wu, L., Zhang, Y., … El-Deiry, W. S. (2023). Therapeutic targeting of TRAIL death receptors. Biochemical Society Transactions, 51(1), 57–70. 10.1042/BST20220098 [DOI] [PMC free article] [PubMed]
  • 83.Ashkenazi, A., Pai, R. C., Fong, S., Leung, S., Lawrence, D. A., Marsters, S. A., … Schwall, R. H. (1999). Safety and antitumor activity of recombinant soluble Apo2 ligand. The Journal of Clinical Investigation, 104(2), 155–162. 10.1172/JCI6926 [DOI] [PMC free article] [PubMed]
  • 84.Schneider, P. (2000). Production of recombinant TRAIL and TRAIL receptor: Fc chimeric proteins. Methods in Enzymology,322, 325–345. 10.1016/S0076-6879(00)22031-4 [DOI] [PubMed] [Google Scholar]
  • 85.Walczak, H., Miller, R. E., Ariail, K., Gliniak, B., Griffith, T. S., Kubin, M., … Lynch, D. H. (1999). Tumoricidal activity of tumor necrosis factor-related apoptosis-inducing ligand in vivo. Nature Medicine, 5(2), 157–163. 10.1038/5517 [DOI] [PubMed]
  • 86.Herbst, R. S., Eckhardt, S. G., Kurzrock, R., Ebbinghaus, S., O’Dwyer, P. J., Gordon, M. S., … Mendelson, D. S. (2010). Phase I dose-escalation study of recombinant human Apo2L/TRAIL, a dual proapoptotic receptor agonist, in patients with advanced cancer. Journal of Clinical Oncology, 28(17), 2839–2846. 10.1200/JCO.2009.25.1991 [DOI] [PubMed]
  • 87.Subbiah, V., Brown, R. E., Buryanek, J., Trent, J., Ashkenazi, A., Herbst, R., & Kurzrock, R. (2012). Targeting the apoptotic pathway in chondrosarcoma using recombinant human Apo2L/TRAIL (dulanermin), a dual proapoptotic receptor (DR4/DR5) agonist. Molecular Cancer Therapeutics,11(11), 2541–2546. 10.1158/1535-7163.MCT-12-0358 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Horak, P., Pils, D., Haller, G., Pribill, I., Roessler, M., Tomek, S., … Krainer, M. (2005). Contribution of epigenetic silencing of tumor necrosis factor-related apoptosis inducing ligand receptor 1 (DR4) to TRAIL resistance and ovarian cancer. Molecular Cancer Research : MCR, 3(6), 335–343. 10.1158/1541-7786.MCR-04-0136 [DOI] [PubMed]
  • 89.Zhang, L., Zhang, X., Barrisford, G. W., & Olumi, A. F. (2007). Lexatumumab (TRAIL-receptor 2 mAb) induces expression of DR5 and promotes apoptosis in primary and metastatic renal cell carcinoma in a mouse orthotopic model. Cancer Letters,251(1), 146–157. 10.1016/J.CANLET.2006.11.013 [DOI] [PubMed] [Google Scholar]
  • 90.Plummer, R., Attard, G., Pacey, S., Li, L., Razak, A., Perrett, R., … De Bono, J. (2007). Phase 1 and pharmacokinetic study of lexatumumab in patients with advanced cancers. Clinical Cancer Research : An Official Journal of the American Association for Cancer Research, 13(20), 6187–6194. 10.1158/1078-0432.CCR-07-0950 [DOI] [PubMed]
  • 91.Hotte, S. J., Hirte, H. W., Chen, E. X., Siu, L. L., Le, L. H., Corey, A., … Oza, A. M. (2008). A phase 1 study of mapatumumab (fully human monoclonal antibody to TRAIL-R1) in patients with advanced solid malignancies. Clinical Cancer Research : An Official Journal of the American Association for Cancer Research, 14(11), 3450–3455. 10.1158/1078-0432.CCR-07-1416 [DOI] [PubMed]
  • 92.Herbst, R. S., Kurzrock, R., Hong, D. S., Valdivieso, M., Hsu, C. P., Goyal, L., … LoRusso, P. M. (2010). A first-in-human study of conatumumab in adult patients with advanced solid tumors. Clinical Cancer Research : An Official Journal of the American Association for Cancer Research, 16(23), 5883–5891. 10.1158/1078-0432.CCR-10-0631 [DOI] [PubMed]
  • 93.Camidge, D. R., Herbst, R. S., Gordon, M. S., Eckhardt, S. G., Kurzrock, R., Durbin, B., … Mendelson, D. (2010). A phase I safety and pharmacokinetic study of the death receptor 5 agonistic antibody PRO95780 in patients with advanced malignancies. Clinical Cancer Research : An Official Journal of the American Association for Cancer Research, 16(4), 1256–1263. 10.1158/1078-0432.CCR-09-1267 [DOI] [PubMed]
  • 94.Forero-Torres, A., Shah, J., Wood, T., Posey, J., Carlisle, R., Copigneaux, C., … Saleh, M. (2010). Phase I trial of weekly tigatuzumab, an agonistic humanized monoclonal antibody targeting death receptor 5 (DR5). Cancer Biotherapy and Radiopharmaceuticals, 25(1), 13–19. 10.1089/CBR.2009.0673 [DOI] [PMC free article] [PubMed]
  • 95.Milligan, K. L., & Barenkamp, S. J. (2013). Neonatal meningitis due to Morganella morganii. Clinical Pediatrics,52(5), 462–464. 10.1177/0009922811435166 [DOI] [PubMed] [Google Scholar]
  • 96.Liu, H., Su, D., Zhang, J., Ge, S., Li, Y., Wang, F., … Liang, P. (2017). Improvement of pharmacokinetic profile of TRAIL via trimer-tag enhances its antitumor activity in vivo. Scientific Reports, 7(1). 10.1038/S41598-017-09518-1 [DOI] [PMC free article] [PubMed]
  • 97.Phillips, D. C., Buchanan, F. G., Cheng, D., Solomon, L. R., Xiao, Y., Xue, J., … Morgan-Lappe, S. E. (2021). Hexavalent TRAIL fusion protein eftozanermin alfa optimally clusters apoptosis-inducing TRAIL receptors to induce on-target antitumor activity in solid tumors. Cancer Research, 81(12), 3402–3414. 10.1158/0008-5472.CAN-20-2178 [DOI] [PubMed]
  • 98.de Jonge, M. J. A., Carneiro, B. A., Devriese, L., Doi, T., Penugonda, S., Petrich, A. M., … Moreno, V. (2019). First-in-human study of Abbv-621, a TRAIL receptor agonist fusion protein, in patients (Pts) with relapsed/refractory (RR) acute myeloid leukemia (AML) and diffuse large B-cell lymphoma (DLBCL). Blood, 134(Supplement_1), 3924–3924. 10.1182/BLOOD-2019-129783
  • 99.LoRusso, P., Ratain, M. J., Doi, T., Rasco, D. W., de Jonge, M. J. A., Moreno, V., … Calvo, E. (2022). Eftozanermin alfa (ABBV-621) monotherapy in patients with previously treated solid tumors: Findings of a phase 1, first-in-human study. Investigational New Drugs, 40(4), 762–772. 10.1007/S10637-022-01247-1 [DOI] [PMC free article] [PubMed]
  • 100.Papadopoulos, K. P., Isaacs, R., Bilic, S., Kentsch, K., Huet, H. A., Hofmann, M., … Mahipal, A. (2015). Unexpected hepatotoxicity in a phase I study of TAS266, a novel tetravalent agonistic Nanobody® targeting the DR5 receptor. Cancer Chemotherapy and Pharmacology, 75(5), 887–895. 10.1007/S00280-015-2712-0 [DOI] [PubMed]
  • 101.Eliopoulos, A. G., & Young, L. S. (2004). The role of the CD40 pathway in the pathogenesis and treatment of cancer. Current Opinion in Pharmacology,4(4), 360–367. 10.1016/j.coph.2004.02.008 [DOI] [PubMed] [Google Scholar]
  • 102.Enell Smith, K., Deronic, A., Hägerbrand, K., Norlén, P., & Ellmark, P. (2021). Rationale and clinical development of CD40 agonistic antibodies for cancer immunotherapy. Expert Opinion on Biological Therapy,21(12), 1635–1646. 10.1080/14712598.2021.1934446 [DOI] [PubMed] [Google Scholar]
  • 103.Vonderheide, R. H., Dutcher, J. P., Anderson, J. E., Eckhardt, S. G., Stephans, K. F., Razvillas, B., … Gribben, J. G. (2001). Phase I study of recombinant human CD40 ligand in cancer patients. Journal of Clinical Oncology : Official Journal of the American Society of Clinical Oncology, 19(13), 3280–3287. 10.1200/JCO.2001.19.13.3280 [DOI] [PubMed]
  • 104.Vonderheide, R. H. (2020). CD40 agonist antibodies in cancer immunotherapy. Annual Review of Medicine,71, 47–58. 10.1146/ANNUREV-MED-062518-045435 [DOI] [PubMed] [Google Scholar]
  • 105.Vonderheide, R. H. (2018). The immune revolution: A case for priming, not checkpoint. Cancer Cell,33(4), 563–569. 10.1016/J.CCELL.2018.03.008 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Piechutta, M., & Berghoff, A. S. (2019). New emerging targets in cancer immunotherapy: The role of cluster of differentiation 40 (CD40/TNFR5). ESMO Open,4(Suppl 3), e000510. 10.1136/ESMOOPEN-2019-000510 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Dahan, R., Barnhart, B. C., Li, F., Yamniuk, A. P., Korman, A. J., & Ravetch, J. V. (2016). Therapeutic activity of agonistic, human anti-CD40 monoclonal antibodies requires selective FcγR engagement. Cancer Cell,29(6), 820–831. 10.1016/J.CCELL.2016.05.001 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Byrne, K. T., & Vonderheide, R. H. (2016). CD40 stimulation obviates innate sensors and drives T cell immunity in cancer. Cell Reports,15(12), 2719–2732. 10.1016/J.CELREP.2016.05.058 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Li, F., & Ravetch, J. V. (2013). Antitumor activities of agonistic anti-TNFR antibodies require differential FcγRIIB coengagement in vivo. Proceedings of the National Academy of Sciences of the United States of America,110(48), 19501–19506. 10.1073/PNAS.1319502110 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.White, A. L., Chan, H. T. C., Roghanian, A., French, R. R., Mockridge, C. I., Tutt, A. L., … Glennie, M. J. (2011). Interaction with FcγRIIB is critical for the agonistic activity of anti-CD40 monoclonal antibody. Journal of Immunology (Baltimore, Md. : 1950), 187(4), 1754–1763. 10.4049/JIMMUNOL.1101135 [DOI] [PubMed]
  • 111.Irenaeus, S. M. M., Nielsen, D., Ellmark, P., Yachnin, J., Deronic, A., Nilsson, A., … Ullenhag, G. J. (2019). First-in-human study with intratumoral administration of a CD40 agonistic antibody, ADC-1013, in advanced solid malignancies. International Journal of Cancer, 145(5), 1189–1199. 10.1002/IJC.32141 [DOI] [PubMed]
  • 112.White, A. L., Chan, H. T. C., French, R. R., Willoughby, J., Mockridge, C. I., Roghanian, A., … Glennie, M. J. (2015). Conformation of the human immunoglobulin G2 hinge imparts superagonistic properties to immunostimulatory anticancer antibodies. Cancer Cell, 27(1), 138–148. 10.1016/J.CCELL.2014.11.001 [DOI] [PMC free article] [PubMed]
  • 113.Djureinovic, D., Wang, M., & Kluger, H. M. (2021). Agonistic CD40 antibodies in cancer treatment. Cancers,13(6), 1–18. 10.3390/CANCERS13061302 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Vonderheide, R. H., Flaherty, K. T., Khalil, M., Stumacher, M. S., Bajor, D. L., Hutnick, N. A., … Antonia, S. J. (2007). Clinical activity and immune modulation in cancer patients treated with CP-870,893, a novel CD40 agonist monoclonal antibody. Journal of Clinical Oncology : Official Journal of the American Society of Clinical Oncology, 25(7), 876–883. 10.1200/JCO.2006.08.3311 [DOI] [PubMed]
  • 115.Sanborn, R., Gabrail, N., Carneiro, B., O’Hara, M., Bordoni, R., Gordon, M., … Yellin, M. (2022). 596 Results from a phase 1 study of CDX-1140, a fully human anti-CD40 agonist monoclonal antibody (mAb), in combination with pembrolizumab. Journal for ImmunoTherapy of Cancer, 10(Suppl 2), A623–A623. 10.1136/JITC-2022-SITC2022.0596
  • 116.Salomon, R., & Dahan, R. (2022). Next generation CD40 agonistic antibodies for cancer immunotherapy. Frontiers in Immunology,13, 940674. 10.3389/FIMMU.2022.940674/PDF [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Padrón, L. J., Maurer, D. M., O’Hara, M. H., O’Reilly, E. M., Wolff, R. A., Wainberg, Z. A., … Vonderheide, R. H. (2022). Sotigalimab and/or nivolumab with chemotherapy in first-line metastatic pancreatic cancer: Clinical and immunologic analyses from the randomized phase 2 PRINCE trial. Nature Medicine, 28(6), 1167–1177. 10.1038/S41591-022-01829-9 [DOI] [PMC free article] [PubMed]
  • 118.Ye, S., Cohen, D., Belmar, N. A., Choi, D., Tan, S. S., Sho, M., … Chao, D. T. (2019). A bispecific molecule targeting CD40 and tumor antigen mesothelin enhances tumor-specific immunity. Cancer Immunology Research, 7(11), 1864–1875. 10.1158/2326-6066.CIR-18-0805 [DOI] [PubMed]
  • 119.Luke, J. J., Barlesi, F., Chung, K., Tolcher, A. W., Kelly, K., Hollebecque, A., … Fong, L. (2021). Phase I study of ABBV-428, a mesothelin-CD40 bispecific, in patients with advanced solid tumors. Journal for Immunotherapy of Cancer, 9(2). 10.1136/JITC-2020-002015 [DOI] [PMC free article] [PubMed]
  • 120.Willoughby, J., Griffiths, J., Tews, I., & Cragg, M. S. (2017). OX40: Structure and function-What questions remain? Molecular Immunology,83, 13–22. 10.1016/J.MOLIMM.2017.01.006 [DOI] [PubMed] [Google Scholar]
  • 121.Soroosh, P., Ine, S., Sugamura, K., & Ishii, N. (2006). OX40-OX40 ligand interaction through T cell-T cell contact contributes to CD4 T cell longevity. Journal of Immunology (Baltimore, Md. : 1950),176(10), 5975–5987. 10.4049/JIMMUNOL.176.10.5975 [DOI] [PubMed] [Google Scholar]
  • 122.Zhang, X., Xiao, X., Lan, P., Li, J., Dou, Y., Chen, W., … Li, X. C. (2018). OX40 costimulation inhibits Foxp3 expression and treg induction via BATF3-dependent and independent mechanisms. Cell Reports, 24(3), 607–618. 10.1016/J.CELREP.2018.06.052 [DOI] [PMC free article] [PubMed]
  • 123.Oberst, M. D., Auge, C., Morris, C., Kentner, S., Mulgrew, K., McGlinchey, K., … Hammond, S. A. (2018). Potent immune modulation by MEDI6383, an engineered human OX40 ligand IgG4P Fc fusion protein. Molecular Cancer Therapeutics, 17(5), 1024–1038. 10.1158/1535-7163.MCT-17-0200 [DOI] [PMC free article] [PubMed]
  • 124.Messenheimer, D. J., Jensen, S. M., Afentoulis, M. E., Wegmann, K. W., Feng, Z., Friedman, D. J., … Fox, B. A. (2017). Timing of PD-1 blockade is critical to effective combination immunotherapy with anti-OX40. Clinical Cancer Research : An Official Journal of the American Association for Cancer Research, 23(20), 6165–6177. 10.1158/1078-0432.CCR-16-2677 [DOI] [PMC free article] [PubMed]
  • 125.Malamas, A. S., Hammond, S. A., Schlom, J., & Hodge, J. W. (2017). Combination therapy with an OX40L fusion protein and a vaccine targeting the transcription factor twist inhibits metastasis in a murine model of breast cancer. Oncotarget,8(53), 90825–90841. 10.18632/ONCOTARGET.19967 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Redmond, W. L., Linch, S. N., & Kasiewicz, M. J. (2014). Combined targeting of costimulatory (OX40) and coinhibitory (CTLA-4) pathways elicits potent effector T cells capable of driving robust antitumor immunity. Cancer Immunology Research,2(2), 142–153. 10.1158/2326-6066.CIR-13-0031-T [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Pourakbari, R., Hajizadeh, F., Parhizkar, F., Aghebati-Maleki, A., Mansouri, S., & Aghebati-Maleki, L. (2021). Co-stimulatory agonists: An insight into the immunotherapy of cancer. EXCLI Journal,20, 1055–1085. 10.17179/EXCLI2021-3522 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Curti, B. D., Kovacsovics-Bankowski, M., Morris, N., Walker, E., Chisholm, L., Floyd, K., … Weinberg, A. D. (2013). OX40 is a potent immune-stimulating target in late-stage cancer patients. Cancer Research, 73(24), 7189–7198. 10.1158/0008-5472.CAN-12-4174 [DOI] [PMC free article] [PubMed]
  • 129.Davis, E. J., Martin-Liberal, J., Kristeleit, R., Cho, D. C., Blagden, S. P., Berthold, D., … Mehnert, J. M. (2022). First-in-human phase I/II, open-label study of the anti-OX40 agonist INCAGN01949 in patients with advanced solid tumors. Journal for Immunotherapy Of Cancer, 10(10). 10.1136/JITC-2021-004235 [DOI] [PMC free article] [PubMed]
  • 130.Postel-Vinay, S., Lam, V. K., Ros, W., Bauer, T. M., Hansen, A. R., Cho, D. C., … Heymach, J. V. (2023). First-in-human phase I study of the OX40 agonist GSK3174998 with or without pembrolizumab in patients with selected advanced solid tumors (ENGAGE-1). Journal for Immunotherapy of Cancer, 11(3). 10.1136/JITC-2022-005301 [DOI] [PMC free article] [PubMed]
  • 131.Yang, Z. Z., Novak, A. J., Ziesmer, S. C., Witzig, T. E., & Ansell, S. M. (2007). CD70+ non-Hodgkin lymphoma B cells induce Foxp3 expression and regulatory function in intratumoral CD4+CD25- T cells. Blood,110(7), 2537–2544. 10.1182/BLOOD-2007-03-082578 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Flieswasser, T., Van den Eynde, A., Van Audenaerde, J., De Waele, J., Lardon, F., Riether, C., … Jacobs, J. (2022). The CD70-CD27 axis in oncology: The new kids on the block. Journal of Experimental and Clinical Cancer Research, 41(1). 10.1186/S13046-021-02215-Y [DOI] [PMC free article] [PubMed]
  • 133.Riether, C., Schürch, C. M., Bührer, E. D., Hinterbrandner, M., Huguenin, A. L., Hoepner, S., … Ochsenbein, A. F. (2017). CD70/CD27 signaling promotes blast stemness and is a viable therapeutic target in acute myeloid leukemia. The Journal of Experimental Medicine, 214(2), 359–380. 10.1084/JEM.20152008 [DOI] [PMC free article] [PubMed]
  • 134.Nilsson, A., De Milito, A., Mowafi, F., Winberg, G., Björk, O., Wolpert, E. Z., & Chiodi, F. (2005). Expression of CD27-CD70 on early B cell progenitors in the bone marrow: Implication for diagnosis and therapy of childhood ALL. Experimental Hematology,33(12), 1500–1507. 10.1016/J.EXPHEM.2005.10.005 [DOI] [PubMed] [Google Scholar]
  • 135.Flieswasser, T., Camara-Clayette, V., Danu, A., Bosq, J., Ribrag, V., Zabrocki, P., … Jacobs, J. (2019). Screening a broad range of solid and haematological tumour types for CD70 expression using a uniform IHC methodology as potential patient stratification method. Cancers, 11(10). 10.3390/cancers11101611 [DOI] [PMC free article] [PubMed]
  • 136.Katayama, Y., Sakai, A., Oue, N., Asaoku, H., Otsuki, T., Shiomomura, T., … Kimura, A. (2003). A possible role for the loss of CD27-CD70 interaction in myelomagenesis. British Journal of Haematology, 120(2), 223–234. 10.1046/j.1365-2141.2003.04069.x [DOI] [PubMed]
  • 137.Liu, L., Yin, B., Yi, Z., Liu, X. J., Hu, Z. Q., Gao, W. C., … Li, Q. Q. (2018). Breast cancer stem cells characterized by CD70 expression preferentially metastasize to the lungs. Breast Cancer, 25(6), 706–716. 10.1007/S12282-018-0880-6 [DOI] [PubMed]
  • 138.Jacobs, J., Deschoolmeester, V., Rolfo, C., Zwaenepoel, K., Van den Bossche, J., Deben, C., … Pauwels, P. (2017). Preclinical data on the combination of cisplatin and anti-CD70 therapy in non-small cell lung cancer as an excellent match in the era of combination therapy. Oncotarget, 8(43), 74058–74067. 10.18632/ONCOTARGET.18202 [DOI] [PMC free article] [PubMed]
  • 139.Claus, C., Riether, C., Schürch, C., Matter, M. S., Hilmenyuk, T., & Ochsenbein, A. F. (2012). CD27 signaling increases the frequency of regulatory T cells and promotes tumor growth. Cancer Research,72(14), 3664–3676. 10.1158/0008-5472.CAN-11-2791 [DOI] [PubMed] [Google Scholar]
  • 140.Liu, L., Yin, B., Yi, Z., Liu, X. J., Hu, Z. Q., Gao, W. C., … Li, Q. Q. (2018). Breast cancer stem cells characterized by CD70 expression preferentially metastasize to the lungs. Breast Cancer (Tokyo, Japan), 25(6), 706–716. 10.1007/S12282-018-0880-6 [DOI] [PubMed]
  • 141.Pich, C., Sarrabayrouse, G., Teiti, I., Mariamé, B., Rochaix, P., Lamant, L., … Tilkin-Mariamé, A. F. (2016). Melanoma-expressed CD70 is involved in invasion and metastasis. British Journal of Cancer, 114(1), 63–70. 10.1038/BJC.2015.412 [DOI] [PMC free article] [PubMed]
  • 142.Starzer, A. M., & Berghoff, A. S. (2020). New emerging targets in cancer immunotherapy: CD27 (TNFRSF7). ESMO Open,4(Suppl 3), e000629. 10.1136/ESMOOPEN-2019-000629 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Owonikoko, T. K., Hussain, A., Stadler, W. M., Smith, D. C., Kluger, H., Molina, A. M., … Cohen, L. J. (2016). First-in-human multicenter phase i study of BMS-936561 (MDX-1203), an antibody-drug conjugate targeting CD70. Cancer Chemotherapy and Pharmacology, 77(1), 155–162. 10.1007/S00280-015-2909-2 [DOI] [PubMed]
  • 144.Silence, K., Dreier, T., Moshir, M., Ulrichts, P., Gabriels, S. M. E., Saunders, M., … De Haard, H. J. (2014). ARGX-110, a highly potent antibody targeting CD70, eliminates tumors via both enhanced ADCC and immune checkpoint blockade. mAbs, 6(2), 523–532. 10.4161/MABS.27398 [DOI] [PMC free article] [PubMed]
  • 145.Aftimos, P., Rolfo, C., Rottey, S., Offner, F., Bron, D., Maerevoet, M., … Awada, A. (2017). Phase I dose-escalation study of the anti-CD70 antibody ARGX-110 in advanced malignancies. Clinical Cancer Research, 23(21), 6411–6420. 10.1158/1078-0432.CCR-17-0613 [DOI] [PubMed]
  • 146.Leupin, N., Zinzani, P. L., Morschhauser, F., Dalle, S., Maerevoet, M., Michot, J. M., … Bagot, M. (2022). Cusatuzumab for treatment of CD70-positive relapsed or refractory cutaneous T-cell lymphoma. Cancer, 128(5), 1004–1014. 10.1002/CNCR.34005 [DOI] [PubMed]
  • 147.Vanegas, Y. M., Mohty, R., Gadd, M. E., Luo, Y., Aljurf, M., Qin, H., & Kharfan-Dabaja, M. A. (2022). CAR-T cell therapies for B-cell lymphoid malignancies: Identifying targets beyond CD19. Hematology/Oncology and Stem Cell Therapy,15(3), 8. 10.56875/2589-0646.1026 [DOI] [PubMed] [Google Scholar]
  • 148.Lutfi, F., Wu, L., Sunshine, S., & Cao, X. (2021). Targeting the CD27-CD70 pathway to improve outcomes in both checkpoint immunotherapy and allogeneic hematopoietic cell transplantation. Frontiers in Immunology,12, 715909. 10.3389/fimmu.2021.715909 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Burris, H. A., Infante, J. R., Ansell, S. M., Nemunaitis, J. J., Weiss, G. R., Villalobos, V. M., … Bullock, T. (2017). Safety and activity of varlilumab, a novel and first-in-class agonist anti-CD27 antibody, in patients with advanced solid tumors. Journal of Clinical Oncology : Official Journal of the American Society of Clinical Oncology, 35(18), 2028–2036. 10.1200/JCO.2016.70.1508 [DOI] [PubMed]
  • 150.Sanborn, R. E., Pishvaian, M. J., Callahan, M. K., Weise, A., Sikic, B. I., Rahma, O., … Keler, T. (2022). Safety, tolerability and efficacy of agonist anti-CD27 antibody (varlilumab) administered in combination with anti-PD-1 (nivolumab) in advanced solid tumors. Journal for Immunotherapy of Cancer, 10(8). 10.1136/JITC-2022-005147 [DOI] [PMC free article] [PubMed]
  • 151.Villasboas, J. C., Kline, J. P., Lazaryan, A., Bartlett, N. L., Hernandez-Ilizaliturri, F. J., Awan, F. T., … Ansell, S. M. (2022). Results of the DIAL study (NCI 10089), a randomized phase 2 trial of varlilumab combined with nivolumab in patients with relapsed/refractory aggressive B-cell non-Hodgkin lymphoma (r/r B-NHL), 40(17_suppl), LBA7564–LBA7564. 10.1200/JCO.2022.40.17_SUPPL.LBA7564
  • 152.Lee, H.-W., Park, S.-J., Choi, B. K., Kim, H. H., Nam, K.-O., & Kwon, B. S. (2002). 4–1BB promotes the survival of CD8+ T lymphocytes by increasing expression of Bcl-xL and Bfl-1. Journal of Immunology (Baltimore, Md. : 1950),169(9), 4882–4888. 10.4049/JIMMUNOL.169.9.4882 [DOI] [PubMed] [Google Scholar]
  • 153.Wang, C., Lin, G. H. Y., McPherson, A. J., & Watts, T. H. (2009). Immune regulation by 4–1BB and 4–1BBL: Complexities and challenges. Immunological Reviews,229(1), 192–215. 10.1111/J.1600-065X.2009.00765.X [DOI] [PubMed] [Google Scholar]
  • 154.Sabbagh, L., Pulle, G., Liu, Y., Tsitsikov, E. N., & Watts, T. H. (2008). ERK-dependent Bim modulation downstream of the 4–1BB-TRAF1 signaling axis is a critical mediator of CD8 T cell survival in vivo. Journal of Immunology (Baltimore, Md. : 1950),180(12), 8093–8101. 10.4049/JIMMUNOL.180.12.8093 [DOI] [PubMed] [Google Scholar]
  • 155.Qi, X., Li, F., Wu, Y., Cheng, C., Han, P., Wang, J., & Yang, X. (2019). Optimization of 4–1BB antibody for cancer immunotherapy by balancing agonistic strength with FcγR affinity. Nature Communications,10(1), 2141. 10.1038/S41467-019-10088-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Wilcox, R. A., Tamada, K., Strome, S. E., & Chen, L. (2002). Signaling through NK cell-associated CD137 promotes both helper function for CD8+ cytolytic T cells and responsiveness to IL-2 but not cytolytic activity. The Journal of Immunology,169(8), 4230–4236. 10.4049/JIMMUNOL.169.8.4230 [DOI] [PubMed] [Google Scholar]
  • 157.Futagawa, T., Akiba, H., Kodama, T., Takeda, K., Hosoda, Y., Yagita, H., & Okumura, K. (2002). Expression and function of 4–1BB and 4–1BB ligand on murine dendritic cells. International Immunology,14(3), 275–286. 10.1093/INTIMM/14.3.275 [DOI] [PubMed] [Google Scholar]
  • 158.Barsoumian, H. B., Yolcu, E. S., & Shirwan, H. (2016). 4–1BB signaling in conventional T cells drives IL-2 production that overcomes CD4+CD25+FoxP3+ T regulatory cell suppression. PloS One,11(4), e0153088. 10.1371/JOURNAL.PONE.0153088 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Melero, I., Shuford, W. W., Newby, S. A., Aruffo, A., Ledbetter, J. A., Hellström, K. E., … Chen, L. (1997). Monoclonal antibodies against the 4–1BB T-cell activation molecule eradicate established tumors. Nature Medicine, 3(6), 682–685. 10.1038/NM0697-682 [DOI] [PubMed]
  • 160.Curran, M. A., Kim, M., Montalvo, W., Al-Shamkhani, A., & Allison, J. P. (2011). Combination CTLA-4 blockade and 4–1BB activation enhances tumor rejection by increasing T-cell infiltration, proliferation, and cytokine production. PloS one,6(4), e19499. 10.1371/JOURNAL.PONE.0019499 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161.Chen, S., Lee, L. F., Fisher, T. S., Jessen, B., Elliott, M., Evering, W., … Lin, J. C. (2015). Combination of 4–1BB agonist and PD-1 antagonist promotes antitumor effector/memory CD8 T cells in a poorly immunogenic tumor model. Cancer Immunology Research, 3(2), 149–160. 10.1158/2326-6066.CIR-14-0118 [DOI] [PubMed]
  • 162.Sznol, M., Hodi, F. S., Margolin, K., McDermott, D. F., Ernstoff, M. S., Kirkwood, J. M., … Logan, T. (2008). Phase I study of BMS-663513, a fully human anti-CD137 agonist monoclonal antibody, in patients (pts) with advanced cancer (CA), 26(15_suppl), 3007–3007. 10.1200/JCO.2008.26.15_SUPPL.3007
  • 163.Chester, C., Sanmamed, M. F., Wang, J., & Melero, I. (2018). Immunotherapy targeting 4–1BB: Mechanistic rationale, clinical results, and future strategies. Blood,131(1), 49–57. 10.1182/BLOOD-2017-06-741041 [DOI] [PubMed] [Google Scholar]
  • 164.Segal, N. H., He, A. R., Doi, T., Levy, R., Bhatia, S., Pishvaian, M. J., … Gopal, A. K. (2018). Phase i study of single-agent utomilumab (PF-05082566), a 4–1bb/cd137 agonist, in patients with advanced cancer. Clinical Cancer Research, 24(8), 1816–1823. 10.1158/1078-0432.CCR-17-1922 [DOI] [PubMed]
  • 165.Papadopoulos, K. P., Autio, K., Golan, T., Dobrenkov, K., Chartash, E., Chen, Q., … Long, G. V. (2021). Phase I study of MK-4166, an anti-human glucocorticoid-induced TNF receptor antibody, alone or with pembrolizumab in advanced solid tumors. Clinical Cancer Research : An Official Journal of the American Association for Cancer Research, 27(7), 1904–1911. 10.1158/1078-0432.CCR-20-2886 [DOI] [PMC free article] [PubMed]
  • 166.Cohen, E. E. W., Pishvaian, M. J., Shepard, D. R., Wang, D., Weiss, J., Johnson, M. L., … Powell, S. F. (2019). A phase Ib study of utomilumab (PF-05082566) in combination with mogamulizumab in patients with advanced solid tumors. Journal for Immunotherapy of Cancer, 7(1). 10.1186/S40425-019-0815-6 [DOI] [PMC free article] [PubMed]
  • 167.Asmamaw Dejenie, T., Tiruneh G/Medhin, M., Dessie Terefe, G., Tadele Admasu, F., Wale Tesega, W., & Chekol Abebe, E. (2022). Current updates on generations, approvals, and clinical trials of CAR T-cell therapy. Human Vaccines and Immunotherapeutics, 18(6). 10.1080/21645515.2022.2114254 [DOI] [PMC free article] [PubMed]
  • 168.Shimizu, J., Yamazaki, S., Takahashi, T., Ishida, Y., & Sakaguchi, S. (2002). Stimulation of CD25(+)CD4(+) regulatory T cells through GITR breaks immunological self-tolerance. Nature Immunology,3(2), 135–142. 10.1038/NI759 [DOI] [PubMed] [Google Scholar]
  • 169.Ronchetti, S., Nocentini, G., Bianchini, R., Krausz, L. T., Migliorati, G., & Riccardi, C. (2007). Glucocorticoid-induced TNFR-related protein lowers the threshold of CD28 costimulation in CD8+ T Cells. The Journal of Immunology,179(9), 5916–5926. 10.4049/jimmunol.179.9.5916 [DOI] [PubMed] [Google Scholar]
  • 170.Buzzatti, G., Dellepiane, C., & Del Mastro, L. (2020). New emerging targets in cancer immunotherapy: The role of GITR. ESMO Open,4(Suppl 3), e000738. 10.1136/ESMOOPEN-2020-000738 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171.Riccardi, C., Ronchetti, S., & Nocentini, G. (2018). Glucocorticoid-induced TNFR-related gene (GITR) as a therapeutic target for immunotherapy. Expert Opinion on Therapeutic Targets,22(9), 783–797. 10.1080/14728222.2018.1512588 [DOI] [PubMed] [Google Scholar]
  • 172.Knee, D. A., Hewes, B., & Brogdon, J. L. (2016). Rationale for anti-GITR cancer immunotherapy. European Journal Of Cancer (Oxford, England : 1990),67, 1–10. 10.1016/J.EJCA.2016.06.028 [DOI] [PubMed] [Google Scholar]
  • 173.Coe, D., Begom, S., Addey, C., White, M., Dyson, J., & Chai, J. G. (2010). Depletion of regulatory T cells by anti-GITR mAb as a novel mechanism for cancer immunotherapy. Cancer Immunology, Immunotherapy : CII,59(9), 1367–1377. 10.1007/S00262-010-0866-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174.Kim, Y. H., Shin, S. M., Choi, B. K., Oh, H. S., Kim, C. H., Lee, S. J., … Kwon, B. S. (2015). Authentic GITR signaling fails to induce tumor regression unless Foxp3+ regulatory T cells are depleted. The Journal of Immunology, 195(10), 4721–4729. 10.4049/jimmunol.1403076 [DOI] [PubMed]
  • 175.Mitsui, J., Nishikawa, H., Muraoka, D., Wang, L., Noguchi, T., Sato, E., … Shiku, H. (2010). Two distinct mechanisms of augmented antitumor activity by modulation of immunostimulatory/inhibitory signals. Clinical Cancer Research : An Official Journal of the American Association for Cancer Research, 16(10), 2781–2791. 10.1158/1078-0432.CCR-09-3243 [DOI] [PubMed]
  • 176.Villarreal, D. O., Chin, D., Smith, M. A., Luistro, L. L., & Snyder, L. A. (2017). Combination GITR targeting/PD-1 blockade with vaccination drives robust antigen-specific antitumor immunity. Oncotarget,8(24), 39117–39130. 10.18632/ONCOTARGET.16605 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177.Leyland, R., Watkins, A., Mulgrew, K. A., Holoweckyj, N., Bamber, L., Tigue, N. J., … Stewart, R. (2017). A novel murine GITR ligand fusion protein induces antitumor activity as a monotherapy that is further enhanced in combination with an OX40 agonist. Clinical Cancer Research, 23(13), 3416–3427. 10.1158/1078-0432.CCR-16-2000 [DOI] [PubMed]
  • 178.Tran, B., Carvajal, R. D., Marabelle, A., Patel, S. P., Lorusso, P. M., Rasmussen, E., … Schöffski, P. (2018). Dose escalation results from a first-in-human, phase 1 study of glucocorticoid-induced TNF receptor-related protein agonist AMG 228 in patients with advanced solid tumors. Journal for ImmunoTherapy of Cancer, 6(1). 10.1186/S40425-018-0407-X [DOI] [PMC free article] [PubMed]
  • 179.Heinhuis, K. M., Carlino, M., Joerger, M., Di Nicola, M., Meniawy, T., Rottey, S., … Siu, L. L. (2020). Safety, tolerability, and potential clinical activity of a glucocorticoid-induced TNF receptor-related protein agonist alone or in combination with nivolumab for patients with advanced solid tumors: A phase 1/2a dose-escalation and cohort-expansion clinical trial. JAMA Oncology, 6(1), 100–107. 10.1001/JAMAONCOL.2019.3848 [DOI] [PMC free article] [PubMed]
  • 180.Piha-Paul, S. A., Geva, R., Tan, T. J., Lim, D. W. T., Hierro, C., Doi, T., … Bedard, P. L. (2021). First-in-human phase I/Ib open-label dose-escalation study of GWN323 (anti-GITR) as a single agent and in combination with spartalizumab (anti-PD-1) in patients with advanced solid tumors and lymphomas. Journal for Immunotherapy Of Cancer, 9(8). 10.1136/JITC-2021-002863 [DOI] [PMC free article] [PubMed]
  • 181.Davar, D., & Zappasodi, R. (2023). Targeting GITR in cancer immunotherapy–There is no perfect knowledge. Oncotarget,14(1), 614–621. 10.18632/ONCOTARGET.28461 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 182.Balmanoukian, A. S., Infante, J. R., Aljumaily, R., Naing, A., Chintakuntlawar, A. V., Rizvi, N. A., … Denlinger, C. S. (2020). Safety and clinical activity of MEDI1873, a novel GITR agonist, in advanced solid tumors. Clinical Cancer Research : An Official Journal of the American Association for Cancer Research, 26(23), 6196–6203. 10.1158/1078-0432.CCR-20-0452 [DOI] [PubMed]
  • 183.Montgomery, R. I., Warner, M. S., Lum, B. J., & Spear, P. G. (1996). Herpes simplex virus-1 entry into cells mediated by a novel member of the TNF/NGF receptor family. Cell,87(3), 427–436. 10.1016/S0092-8674(00)81363-X [DOI] [PubMed] [Google Scholar]
  • 184.Cheung, T. C., & Ware, C. F. (2011). The canonical and unconventional ligands of the herpesvirus entry mediator. Advances in Experimental Medicine and Biology,691, 353. 10.1007/978-1-4419-6612-4_36 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 185.Jones, A., Bourque, J., Kuehm, L., Opejin, A., Teague, R. M., Gross, C., & Hawiger, D. (2016). Immunomodulatory functions of BTLA and HVEM govern induction of extrathymic regulatory T cells and tolerance by dendritic cells. Immunity,45(5), 1066–1077. 10.1016/J.IMMUNI.2016.10.008 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 186.Mohamed, A. H., Obeid, R. A., Fadhil, A. A., Amir, A. A., Adhab, Z. H., Jabouri, E. A., … Alshahrani, M. Y. (2023). BTLA and HVEM: Emerging players in the tumor microenvironment and cancer progression. Cytokine, 172, 156412. 10.1016/J.CYTO.2023.156412 [DOI] [PubMed]
  • 187.Pasero, C., Speiser, D. E., Derré, L., & Olive, D. (2012). The HVEM network: New directions in targeting novel costimulatory/co-inhibitory molecules for cancer therapy. Current Opinion in Pharmacology,12(4), 478–485. 10.1016/J.COPH.2012.03.001 [DOI] [PubMed] [Google Scholar]
  • 188.Cheung, K. J. J., Johnson, N. A., Affleck, J. G., Severson, T., Steidl, C., Ben-Neriah, S., … Horsman, D. E. (2010). Acquired TNFRSF14 mutations in follicular lymphoma are associated with worse prognosis. Cancer Research, 70(22), 9166–9174. 10.1158/0008-5472.CAN-10-2460 [DOI] [PubMed]
  • 189.Gubernatorova, E. O., Polinova, A. I., Petropavlovskiy, M. M., Namakanova, O. A., Medvedovskaya, A. D., Zvartsev, R. V., … Nedospasov, S. A. (2021). Dual role of TNF and LTα in carcinogenesis as implicated by studies in mice. Cancers, 13(8). 10.3390/CANCERS13081775 [DOI] [PMC free article] [PubMed]
  • 190.Skeate, J. G., Otsmaa, M. E., Prins, R., Fernandez, D. J., Da Silva, D. M., & Kast, W. M. (2020). TNFSF14: LIGHTing the way for effective cancer immunotherapy. Frontiers in Immunology,11, 537705. 10.3389/FIMMU.2020.00922/BIBTEX [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 191.Mauri, D. N., Ebner, R., Montgomery, R. I., Kochel, K. D., Cheung, T. C., Yu, G. L., … Ware, C. F. (1998). LIGHT, a new member of the TNF superfamily, and lymphotoxin alpha are ligands for herpesvirus entry mediator. Immunity, 8(1), 21–30. 10.1016/S1074-7613(00)80455-0 [DOI] [PubMed]
  • 192.Holmes, T. D., Wilson, E. B., Black, E. V. I., Benest, A. V., Vaz, C., Tan, B., … Cook, G. P. (2014). Licensed human natural killer cells aid dendritic cell maturation via TNFSF14/LIGHT. Proceedings of the National Academy of Sciences of the United States of America, 111(52), E5688–E5696. 10.1073/PNAS.1411072112 [DOI] [PMC free article] [PubMed]
  • 193.Yu, P., Lee, Y., Wang, Y., Liu, X., Auh, S., Gajewski, T. F., … Fu, Y.-X. (2007). Targeting the primary tumor to generate CTL for the effective eradication of spontaneous metastases. Journal of Immunology (Baltimore, Md. : 1950), 179(3), 1960–1968. 10.4049/JIMMUNOL.179.3.1960 [DOI] [PMC free article] [PubMed]
  • 194.Jazowiecka-Rakus, J., Hadrys, A., Rahman, M. M., McFadden, G., Fidyk, W., Chmielik, E., … Sochanik, A. (2021). Myxoma virus expressing LIGHT (TNFSF14) pre-loaded into adipose-derived mesenchymal stem cells is effective treatment for murine pancreatic adenocarcinoma. Cancers, 13(6), 1–23. 10.3390/CANCERS13061394 [DOI] [PMC free article] [PubMed]
  • 195.Christie, J. D., Appel, N., Zhang, L., Lowe, K., Kilbourne, J., Daggett-Vondras, J., … McFadden, G. (2022). Systemic delivery of mLIGHT-armed myxoma virus is therapeutic for later-stage syngeneic murine lung metastatic osteosarcoma. Cancers, 14(2). 10.3390/CANCERS14020337 [DOI] [PMC free article] [PubMed]
  • 196.Tang, H., Wang, Y., Chlewicki, L. K., Zhang, Y., Guo, J., Liang, W., … Fu, Y. X. (2016). Facilitating T cell infiltration in tumor microenvironment overcomes resistance to PD-L1 blockade. Cancer Cell, 29(3), 285–296. 10.1016/J.CCELL.2016.02.004 [DOI] [PMC free article] [PubMed]
  • 197.Lasaro, M. O., Sazanovich, M., Giles-Davis, W., Mrass, P., Bunte, R. M., Sewell, D. A., … Ertl, H. C. J. (2011). Active immunotherapy combined with blockade of a coinhibitory pathway achieves regression of large tumor masses in cancer-prone mice. Molecular Therapy, 19(9), 1727–1736. 10.1038/MT.2011.88 [DOI] [PMC free article] [PubMed]
  • 198.Sordo-Bahamonde, C., Lorenzo-Herrero, S., Gonzalez-Rodriguez, A. P., Payer, Á. R., González-García, E., López-Soto, A., & Gonzalez, S. (2021). Btla/hvem axis induces nk cell immunosuppression and poor outcome in chronic lymphocytic leukemia. Cancers,13(8), 1766. 10.3390/CANCERS13081766/S1 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 199.Schilder, R. J., Powderly, J. D., Park, H., Bilen, M. A., McKean, M., May, R., … Naing, A. (2022). Phase Ia dose-escalation study of the anti-BTLA antibody icatolimab as a monotherapy in patients with advanced solid tumor, 40(16_suppl), 2643–2643. 10.1200/JCO.2022.40.16_SUPPL.2643
  • 200.Cheng, Y., Wang, J., Yu, Y., Wang, Q., Yang, R., Lv, D., … Wang, W. (2023). Phase I/II combination study of tifcemalimab with toripalimab in patients with refractory extensive stage small cell lung cancer (ES-SCLC), 41(16_suppl), 8579–8579. 10.1200/JCO.2023.41.16_SUPPL.8579
  • 201.Ma, J., Song, Y., Xie, Y., Zhang, H.-L., Shuang, Y., Peng, Z., … Zhu, J. (2022). Phase I study of the anti-Btla antibody tifcemalimab as a single agent or in combination with toripalimab in relapsed/refractory lymphomas. Blood, 140(Supplement 1), 3716–3717. 10.1182/BLOOD-2022-157497
  • 202.Berdeja, J. G., Madduri, D., Usmani, S. Z., Jakubowiak, A., Agha, M., Cohen, A. D., … Jagannath, S. (2021). Ciltacabtagene autoleucel, a B-cell maturation antigen-directed chimeric antigen receptor T-cell therapy in patients with relapsed or refractory multiple myeloma (CARTITUDE-1): A phase 1b/2 open-label study. Lancet (London, England), 398(10297), 314–324. 10.1016/S0140-6736(21)00933-8 [DOI] [PubMed]
  • 203.Hu, S., Wang, R., Zhang, M., Liu, K., Tao, J., Tai, Y., … Wei, W. (2019). BAFF promotes T cell activation through the BAFF-BAFF-R-PI3K-Akt signaling pathway. Biomedicine and Pharmacotherapy = Biomedecine and Pharmacotherapie, 114. 10.1016/J.BIOPHA.2019.108796 [DOI] [PubMed]
  • 204.Moreaux, J., Veyrune, J. L., De Vos, J., & Klein, B. (2009). APRIL is overexpressed in cancer: Link with tumor progression. BMC Cancer,9, 1–9. 10.1186/1471-2407-9-83 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 205.Nowacka, K. H., & Jabłońska, E. (2021). Role of the APRIL molecule in solid tumors. Cytokine and Growth Factor Reviews,61, 38–44. 10.1016/J.CYTOGFR.2021.08.001 [DOI] [PubMed] [Google Scholar]
  • 206.Mhawech-Fauceglia, P., Allal, A., Odunsi, K., Andrews, C., Herrmann, F. R., & Huard, B. (2008). Role of the tumour necrosis family ligand APRIL in solid tumour development: Retrospective studies in bladder, ovarian and head and neck carcinomas. European Journal of Cancer (Oxford, England : 1990),44(15), 2097–2100. 10.1016/J.EJCA.2008.07.007 [DOI] [PubMed] [Google Scholar]
  • 207.Ullah, M. A., & Mackay, F. (2023). The BAFF-APRIL system in cancer. Cancers,15(6), 1791. 10.3390/CANCERS15061791 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 208.Lonial, S., Lee, H. C., Badros, A., Trudel, S., Nooka, A. K., Chari, A., … Cohen, A. D. (2020). Belantamab mafodotin for relapsed or refractory multiple myeloma (DREAMM-2): A two-arm, randomised, open-label, phase 2 study. The Lancet Oncology, 21(2), 207–221. 10.1016/S1470-2045(19)30788-0 [DOI] [PubMed]
  • 209.Munshi, N. C., Anderson, L. D., Shah, N., Madduri, D., Berdeja, J., Lonial, S., … San-Miguel, J. (2021). Idecabtagene vicleucel in relapsed and refractory multiple myeloma. The New England Journal of Medicine, 384(8), 705–716. 10.1056/NEJMOA2024850 [DOI] [PubMed]
  • 210.Martins, F., Sofiya, L., Sykiotis, G. P., Lamine, F., Maillard, M., Fraga, M., … Obeid, M. (2019). Adverse effects of immune-checkpoint inhibitors: Epidemiology, management and surveillance. Nature Reviews Clinical Oncology, 16(9), 563–580. 10.1038/S41571-019-0218-0 [DOI] [PubMed]
  • 211.Schneider, B. J., Naidoo, J., Santomasso, B. D., Lacchetti, C., Adkins, S., Anadkat, M., … Bollin, K. (2021). Management of immune-related adverse events in patients treated with immune checkpoint inhibitor therapy: ASCO guideline update. Journal of Clinical Oncology : Official Journal of the American Society of Clinical Oncology, 39(36), 4073–4126. 10.1200/JCO.21.01440 [DOI] [PubMed]
  • 212.Haanen, J., Obeid, M., Spain, L., Carbonnel, F., Wang, Y., Robert, C., … Larkin, J. (2022). Management of toxicities from immunotherapy: ESMO clinical practice guideline for diagnosis, treatment and follow-up. Annals of Oncology : Official Journal of the European Society for Medical Oncology, 33(12), 1217–1238. 10.1016/J.ANNONC.2022.10.001 [DOI] [PubMed]
  • 213.Thompson, J. A., Schneider, B. J., Brahmer, J., Andrews, S., Armand, P., Bhatia, S., … Engh, A. (2020). NCCN guidelines insights: Management of immunotherapy-related toxicities, version 1.2020. Journal of the National Comprehensive Cancer Network : JNCCN, 18(3), 231–241. 10.6004/JNCCN.2020.0012 [DOI] [PubMed]
  • 214.Brahmer, J. R., Abu-Sbeih, H., Ascierto, P. A., Brufsky, J., Cappelli, L. C., Cortazar, F. B., … Ernstoff, M. S. (2021). Society for Immunotherapy of Cancer (SITC) clinical practice guideline on immune checkpoint inhibitor-related adverse events. Journal for Immunotherapy of Cancer, 9(6). 10.1136/JITC-2021-002435 [DOI] [PMC free article] [PubMed]
  • 215.Morgado, M., Plácido, A., Morgado, S., & Roque, F. (2020). Management of the adverse effects of immune checkpoint inhibitors. Vaccines,8(4), 1–15. 10.3390/VACCINES8040575 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 216.Johnson, D. H., Zobniw, C. M., Trinh, V. A., Ma, J., Bassett, R. L., Abdel-Wahab, N., … Diab, A. (2018). Infliximab associated with faster symptom resolution compared with corticosteroids alone for the management of immune-related enterocolitis. Journal for ImmunoTherapy of Cancer, 6(1). 10.1186/S40425-018-0412-0 [DOI] [PMC free article] [PubMed]
  • 217.Suresh, K., Voong, K. R., Shankar, B., Forde, P. M., Ettinger, D. S., Marrone, K. A., … Naidoo, J. (2018). Pneumonitis in non-small cell lung cancer patients receiving immune checkpoint immunotherapy: Incidence and risk factors. Journal of Thoracic Oncology : Official Publication of the International Association for the Study of Lung Cancer, 13(12), 1930–1939. 10.1016/J.JTHO.2018.08.2035 [DOI] [PubMed]
  • 218.Mukherjee, E. M., & Phillips, E. J. (2022). Where to place etanercept and combination treatment for Stevens-Johnson syndrome and toxic epidermal necrolysis? Annals of Allergy, Asthma and Immunology,129(3), 269–270. 10.1016/j.anai.2022.06.025 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 219.Verheijden, R. J., May, A. M., Blank, C. U., Aarts, M. J. B., Berkmortel, F. W. P. J. V. Den, Eertwegh, A. J. M. V. Den, … Suijkerbuijk, K. P. M. (2020). Association of anti-TNF with decreased survival in steroid refractory ipilimumab and anti-PD1-treated patients in the Dutch melanoma treatment registry. Clinical Cancer Research : An Official Journal of the American Association for Cancer Research, 26(9), 2268–2274. 10.1158/1078-0432.CCR-19-3322 [DOI] [PubMed]
  • 220.Wang, Y., Abu-Sbeih, H., Mao, E., Ali, N., Ali, F. S., Qiao, W., … Diab, A. (2018). Immune-checkpoint inhibitor-induced diarrhea and colitis in patients with advanced malignancies: Retrospective review at MD Anderson. Journal for ImmunoTherapy of Cancer, 6(1). 10.1186/S40425-018-0346-6 [DOI] [PMC free article] [PubMed]
  • 221.Chen, A. Y., Wolchok, J. D., & Bass, A. R. (2021). TNF in the era of immune checkpoint inhibitors: Friend or foe? Nature Reviews Rheumatology,17(4), 213–223. 10.1038/S41584-021-00584-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 222.Ogusu, S., Harutani, Y., Tozuka, T., Saito, R., Koyama, J., Sakamoto, H., … Ariyasu, R. (2023). Second-line immunosuppressant administration for steroid-refractory immune-related adverse events in patients with lung cancer. Cancer Immunology, Immunotherapy. 10.1007/S00262-023-03528-X [DOI] [PMC free article] [PubMed]
  • 223.Croft, M., Duan, W., Choi, H., Eun, S. Y., Madireddi, S., & Mehta, A. (2012). TNF superfamily in inflammatory disease: Translating basic insights. Trends in Immunology,33(3), 144–152. 10.1016/J.IT.2011.10.004 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 224.Pagnini, C., & Cominelli, F. (2021). Tumor necrosis factor’s pathway in Crohn’s disease: Potential for intervention. International Journal of Molecular Sciences,22(19), 10273. 10.3390/IJMS221910273 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 225.Jang, D. I., Lee, A. H., Shin, H. Y., Song, H. R., Park, J. H., Kang, T. B., … Yang, S. H. (2021). The role of tumor necrosis factor alpha (TNF-α) in autoimmune disease and current TNF-α inhibitors in therapeutics. International Journal of Molecular Sciences, 22(5), 1–16. 10.3390/IJMS22052719 [DOI] [PMC free article] [PubMed]
  • 226.Chen, X., Wu, X., Zhou, Q., Howard, O. M. Z., Netea, M. G., & Oppenheim, J. J. (2013). TNFR2 is critical for the stabilization of the CD4+Foxp3+ regulatory T. cell phenotype in the inflammatory environment. Journal of Immunology (Baltimore, Md. : 1950),190(3), 1076–1084. 10.4049/JIMMUNOL.1202659 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 227.Peyrin-Biroulet, L., Sandborn, W. J., Panaccione, R., Domènech, E., Pouillon, L., Siegmund, B., … Ghosh, S. (2021). Tumour necrosis factor inhibitors in inflammatory bowel disease: The story continues. Therapeutic Advances in Gastroenterology, 14. 10.1177/17562848211059954/ASSET/IMAGES/LARGE/10.1177_17562848211059954-FIG2.JPEG [DOI] [PMC free article] [PubMed]
  • 228.Lopetuso, L. R., Cuomo, C., Mignini, I., Gasbarrini, A., & Papa, A. (2023). Focus on anti-tumour necrosis factor (TNF)-α-related autoimmune diseases. International Journal of Molecular Sciences,24(9), 8187. 10.3390/IJMS24098187 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 229.LaMattina, K. C., & Goldstein, D. A. (2017). Adalimumab for the treatment of uveitis. Expert Review of Clinical Immunology,13(3), 181–188. 10.1080/1744666X.2017.1288097 [DOI] [PubMed] [Google Scholar]
  • 230.Ghilardi, N., Pappu, R., Arron, J. R., & Chan, A. C. (2020). 30 Years of biotherapeutics development-What have we learned? Annual Review of Immunology,38, 249–287. 10.1146/ANNUREV-IMMUNOL-101619-031510 [DOI] [PubMed] [Google Scholar]
  • 231.Ward-Kavanagh, L. K., Lin, W. W., Šedý, J. R., & Ware, C. F. (2016). The TNF receptor superfamily in co-stimulating and co-inhibitory responses. Immunity,44(5), 1005–1019. 10.1016/J.IMMUNI.2016.04.019 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 232.Croft, M., Benedict, C. A., & Ware, C. F. (2013). Clinical targeting of the TNF and TNFR superfamilies. Nature Reviews Drug Discovery,12(2), 147. 10.1038/NRD3930 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 233.Stohl, W., & Hilbert, D. M. (2012). The discovery and development of belimumab: The anti-BLyS–lupus connection. Nature Biotechnology,30(1), 69. 10.1038/NBT.2076 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 234.Liossis, S. N. C., & Sfikakis, P. P. (2004). Costimulation blockade in the treatment of rheumatic diseases. BioDrugs,18(2), 95–102. 10.2165/00063030-200418020-00003/METRICS [DOI] [PubMed] [Google Scholar]
  • 235.Tian, J., Zhang, B., Rui, K., & Wang, S. (2020). The role of GITR/GITRL interaction in autoimmune diseases. Frontiers in Immunology,11, 588682. 10.3389/FIMMU.2020.588682/BIBTEX [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 236.Salek-Ardakani, S., Zajonc, D. M., & Croft, M. (2023). Agonism of 4–1BB for immune therapy: A perspective on possibilities and complications. Frontiers in Immunology,14, 1228486. 10.3389/FIMMU.2023.1228486/BIBTEX [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 237.Lee, J., Lee, E. N., Kim, E. Y., Park, H. J., Chang, C. Y., Jung, D. Y., … Kim, S. J. (2005). Administration of agonistic anti-4–1BB monoclonal antibody leads to the amelioration of inflammatory bowel disease. Immunology Letters, 101(2), 210–216. 10.1016/J.IMLET.2005.06.001 [DOI] [PubMed]
  • 238.Harris, J., & Keane, J. (2010). How tumour necrosis factor blockers interfere with tuberculosis immunity. Clinical and Experimental Immunology,161(1), 1. 10.1111/J.1365-2249.2010.04146.X [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 239.Strangfeld, A., Listing, J., Herzer, P., Liebhaber, A., Rockwitz, K., Richter, C., & Zink, A. (2009). Risk of herpes zoster in patients with rheumatoid arthritis treated with anti–TNF-α agents. JAMA,301(7), 737–744. 10.1001/JAMA.2009.146 [DOI] [PubMed] [Google Scholar]
  • 240.Mohd Zawawi, Z., Kalyanasundram, J., Mohd Zain, R., Thayan, R., Basri, D. F., & Yap, W. B. (2023). Prospective roles of tumor necrosis factor-alpha (TNF-α) in COVID-19: Prognosis, therapeutic and management. International Journal of Molecular Sciences,24(7), 6142. 10.3390/IJMS24076142 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 241.Wang, X., & Lin, Y. (2008). Tumor necrosis factor and cancer, buddies or foes? Acta Pharmacologica Sinica,29(11), 1275. 10.1111/J.1745-7254.2008.00889.X [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 242.Ababneh, O. E., Kato, S., Nishizaki, D., Miyashita, H., Lee, S., Nesline, M., … Kurzrock, R. (2024). Abstract 6393: The predictive role of TNF-related genes in patients receiving immune checkpoint inhibitors. Cancer Research, 84(6_Supplement), 6393–6393. 10.1158/1538-7445.AM2024-6393
  • 243.Kato, S., Kim, K. H., Lim, H. J., Boichard, A., Nikanjam, M., Weihe, E., … Kurzrock, R. (2020). Real-world data from a molecular tumor board demonstrates improved outcomes with a precision N-of-One strategy. Nature Communications, 11(1). 10.1038/S41467-020-18613-3 [DOI] [PMC free article] [PubMed]
  • 244.Nikanjam, M., Kato, S., Sicklick, J. K., & Kurzrock, R. (2023). At the right dose: Personalised (N-of-1) dosing for precision oncology. European Journal of Cancer,194, 113359. 10.1016/j.ejca.2023.113359 [DOI] [PubMed] [Google Scholar]
  • 245.Sicklick, J. K., Kato, S., Okamura, R., Schwaederle, M., Hahn, M. E., Williams, C. B., … Kurzrock, R. (2019). Molecular profiling of cancer patients enables personalized combination therapy: The I-PREDICT study. Nature Medicine, 25(5), 744–750. 10.1038/S41591-019-0407-5 [DOI] [PMC free article] [PubMed]
  • 246.Rodon, J., Soria, J. C., Berger, R., Miller, W. H., Rubin, E., Kugel, A., … Kurzrock, R. (2019). Genomic and transcriptomic profiling expands precision cancer medicine: The WINTHER trial. Nature Medicine 2019 25:5, 25(5), 751–758. 10.1038/s41591-019-0424-4 [DOI] [PMC free article] [PubMed]
  • 247.Sicklick, J. K., Kato, S., Okamura, R., Patel, H., Nikanjam, M., Fanta, P. T., … Kurzrock, R. (2021). Molecular profiling of advanced malignancies guides first-line N-of-1 treatments in the I-PREDICT treatment-naïve study. Genome Medicine, 13(1), 1–14. 10.1186/S13073-021-00969-W/FIGURES/3 [DOI] [PMC free article] [PubMed]
  • 248.Herman, J. M., Wild, A. T., Wang, H., Tran, P. T., Chang, K. J., Taylor, G. E., … Laheru, D. A. (2013). Randomized phase III multi-institutional study of TNFerade biologic with fluorouracil and radiotherapy for locally advanced pancreatic cancer: Final results. Journal of Clinical Oncology : Official Journal of the American Society of Clinical Oncology, 31(7), 886–894. 10.1200/JCO.2012.44.7516 [DOI] [PMC free article] [PubMed]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Data Availability Statement

No datasets were generated or analysed during the current study.


Articles from Cancer Metastasis Reviews are provided here courtesy of Springer

RESOURCES