Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2021 Aug 16.
Published in final edited form as: Nat Rev Rheumatol. 2021 Mar 8;17(4):213–223. doi: 10.1038/s41584-021-00584-4

TNF in the era of immune checkpoint inhibitors: friend or foe?

Allen Y Chen 1,3,4, Jedd D Wolchok 2,4, Anne R Bass 1,4,
PMCID: PMC8366509  NIHMSID: NIHMS1728635  PMID: 33686279

Abstract

Immune checkpoint inhibitors (ICIs) are effective in the treatment of patients with advanced cancer and have emerged as a pillar of standard cancer care. However, their use is complicated by adverse effects known as immune-related adverse events (irAEs), including ICI-induced inflammatory arthritis. ICI-induced inflammatory arthritis is distinguished from other irAEs by its persistence and requirement for long-term treatment. TNF inhibitors are commonly used to treat inflammatory diseases such as rheumatoid arthritis, spondyloarthopathies and inflammatory bowel disease, and have also been adopted as second-line agents to treat irAEs refractory to glucocorticoid treatment. Experiencing an irAE is associated with a better anti-tumour response after ICI treatment. However, whether TNF inhibition can be safely used to treat irAEs without promoting cancer progression, either by compromising ICI therapy efficacy or via another route, remains an open question. In this Review, we discuss clinical and preclinical studies that address the relationship between TNF, TNF inhibition and cancer. The bulk of the evidence suggests that at least short courses of TNF inhibitors is safe in the treatment of irAEs in patients with cancer undergoing ICI therapy. Data from preclinical studies hint that TNF inhibition might augment the anti-tumour effect of ICI therapy while simultaneously ameliorating irAEs.

ToC blurb

TNF inhibitors are used to treat various immune-related adverse events caused by immune checkpoint inhibitors (ICIs). However, whether TNF inhibition compromises the efficacy of ICI therapy is unknown. This Review discusses the relationship between TNF, TNF inhibition and cancer.

Introduction

Immunotherapy is now a standard approach to cancer treatment alongside surgery, radiation, chemotherapy and targeted therapies. Immune checkpoint inhibitors (ICIs) are monoclonal antibodies that augment the pre-existing host anti-tumour response by blocking down-regulators of the immune system including cytotoxic T lymphocyte antigen 4 (CTLA4), programmed cell death 1 (PD1) and programmed cell death ligand 1 (PDL1). However, in augmenting host immune responses, ICIs cause autoimmune adverse effects, termed immune-related adverse events (irAEs), in >80% of treated patients, including high grade irAEs in ~60% of patients being treated with a combination of ICIs, ~30% of patients being treated with a CTLA4 inhibitor and ~20% of patients being treated with an inhibitor of the PD1 pathway14. Almost any organ of the body can be affected by irAEs, but different ICIs tend to target different organs. For example, rash and colitis are common with anti-CTLA4, whereas arthritis and pneumonitis are more characteristic of anti-PDL1 and anti-PD1 therapy1.

Approximately 4% of patients with cancer undergoing ICI therapy develop inflammatory arthritis5, the majority of whom present with either a rheumatoid arthritis (RA) or a polymyalgia rheumatica phenotype57. Rheumatoid factor and anti-cyclic citrullinated peptide antibodies can be present in such patients but are less common than in patients with RA6,7. Guidelines for the management of ICI-induced inflammatory arthritis are based on expert consensus and borrow heavily from treatments that were developed for RA8. Most patients are initiated treated with corticosteroids at doses determined by the severity of arthritis, and steroid-sparing agents including hydroxychloroquine, sulfasalazine and methotrexate. TNF inhibitors and occasionally IL-6R blockers are used in patients with steroid-refractory or persistently steroid-dependent arthritis912.

Data from various studies show that patients undergoing ICI therapy who develop irAEs have improved progression-free survival and overall survival1315, suggesting that ICIs augment shared immune pathways that promote both irAEs and anti-tumour activity. This finding raises the logical question as to whether immunosuppressive agents used to treat irAEs also promote cancer progression, whether by interfering with the anti-tumour activity of ICIs or via another route. Although studies have documented the oncologic safety of using corticosteroids to control irAEs16, treatment of irAEs with high-dose corticosteroids, although life-saving for patients with severe irAEs such as myocarditis or colitis17,18, was noted to reduce overall survival of patients with hypophysitis19. Survival was also reduced in those patients who were receiving corticosteroid treatment at the time of initiation of ICI therapy20. This finding implies a need to identify targeted therapies that block pathways that contribute to irAE pathogenesis but that spare those pathways that contribute to cancer survival.

The question of whether a treatment for irAEs promotes cancer progression is particularly relevant to rheumatologists because ICI-induced inflammatory arthritis often persists and can require long-term treatment with DMARDs such as TNF inhibitors10. In this Review, we address this question for TNF inhibitors by drawing from literature on the link between TNF and cancer, the link between TNF inhibitors and cancer both within and outside the context of ICIs, and the role of TNF in the tumour microenvironment.

The multifaceted effects of

When TNF was first isolated in 1975 by Carswell, Old and colleagues, it was identified as the factor responsible for endotoxin-induced haemorrhagic necrosis of experimental tumours21. The line of research leading to the isolation of TNF can be traced back to William Coley’s use of bacterial extracts to treat patients with cancer starting in 189622. Although the validity of the clinical case series reported by Coley was controversial, his work motivated subsequent pre-clinical studies in animal models. Further research in 1944 showed that lipopolysaccharide (LPS) endotoxin was the active agent in bacterial extracts that induced haemorrhagic tumour necrosis in a mouse model of benzopyrene-induced skin tumours23. In 1962, researchers found that serum from endotoxin-treated animals also induced tumour necrosis, implying that bacterial endotoxin acts indirectly, inducing an intermediary ‘tumour necrosing factor’ that acted on tumours24. It was this factor that was isolated by Carswell et al in 197521. The gene encoding human TNF was cloned in 198425 and the gene encoding mouse TNF was cloned in 198526. Ascertaining the sequence of TNF led to the discovery that this protein is the same protein molecule as cachectin27, a factor found to mediate acute shock and chronic cachexia during infection. Development of anti-TNF antibodies led to the discovery that TNF has an important role in RA synovial inflammation28. The delineation of multiple physiologic roles for TNF led to further studies of TNF in the context of cancer, sepsis and inflammatory disease

In this Review, we discuss the role of TNF in cancer as is relevant to the safety of TNF inhibitors in the treatment of irAEs. We also discuss the role of TNF in inflammatory disease as is relevant to the efficacy of TNF inhibitors in the treatment of irAEs, with the caveat that irAEs are iatrogenic disease entities whose aetiology is not well-understood and might be different from that of spontaneous inflammatory diseases. Although irAEs are known to be caused by ICIs, it is not yet known which of cell types that are modulated by ICIs mediate these autoimmune toxicities, or why TNF inhibition is an effective treatment, although previous work on the mechanism of action of TNF inhibitors will be a valuable guide. The roles of TNF in cancer and inflammatory diseases are summarized in Box 1.

Key points.

Box 1: Roles of TNF in cancer and inflammatory disease.

Pro-cancer effects
  • Induces tumour angiogenesis39

  • Promotes cancer cell survival and proliferation130

  • Helps cancer cells evade immune surveillance139,141

Anti-cancer effects
  • Induces haemorrhagic tumour necrosis via pro-coagulant effect31, by inducing endothelial cell death32

  • Membrane TNF has direct cytotoxic activity against cancer cells36

Pro-inflammatory effects
  • Upregulates endothelial cell expression of leukocyte adhesion molecules51

  • Induces synovial fibroblast production of IL-6, IL-8, and prostaglandin59

  • Induces macrophage pro-inflammatory phenotype61

Anti-inflammatory effects
  • Ameliorates T cell driven experimental autoimmune encephalomyelitis53

  • Promotes regulatory T cell suppressive function62

  • Chronic TNF attenuates T cell response to antigen stimulation56

Role of TNF in cancer

Early optimism that TNF would be a useful anti-cancer therapy was tempered by the realization that it has a narrow therapeutic window. In clinical trials, systemically administered TNF caused acute shock but without the anti-tumour responses originally reported by Coley29. The physiologic serum concentration of TNF in humans is on the order of 10 pg/mL30, whereas the doses of TNF used in these clinical trials corresponded to TNF serum concentrations on the order of 10 ng/mL29. It is now thought that haemorrhagic tumour necrosis induced by high-dose TNF is largely mediated by the pro-coagulant effects of TNF that lead to thrombosis within the tumour vasculature31. TNF has been shown to induce endothelial cell apoptosis in vitro32. If this process occurs in vivo it could be another mechanism by which high-dose TNF induces haemorrhagic tumour necrosis. The current consensus is that soluble TNF alone, at levels tolerated by patients, is not directly cytotoxic to cancer cells33. However, non-soluble TNF or TNF in conjunction with a second effector molecule can be directly cytotoxic. TNF does have direct cytotoxic effects on cancer cells when used together with small molecules that oppose inhibitor of apoptosis proteins (IAPs)34. In addition, membrane-bound TNF (which serves as a ligand to TNF receptors on adjacent cells) has been shown, in vitro, to have direct cytotoxic effect on target cells, including the KYM-1D4 cancer cell line35,36.

In parallel with studies of TNF as a potential anti-tumour therapy, evidence began to emerge in the late 1980s that TNF could in fact be a tumour promoting factor. Patient-derived juvenile chronic myelogenous leukemia cells were found to produce TNF and use it as an autocrine growth factor37. Another study, in a rabbit cornea model, showed that low-dose TNF induces angiogenesis, an unexpected finding given that high-dose TNF causes destruction of tumour vascular beds38. This finding prompted studies that found that TNF stimulated tumour growth and does so in part by mediating angiogenesis39.

Work since the late 1990s has shown that TNF at physiologic levels (as opposed to the supraphysiologic levels of TNF used in anti-tumour therapy) has a major role in tumorigenesis. In the 1990s, methods to generate gene knockout mice provided powerful tools to elucidate the role of specific genes in mammalian biology. The development of TNF knockout mice led to the discovery that lack of TNF had a protective effect against skin tumours induced by the carcinogen DMBA40. Similarly, TNF receptor knockout mice were also protected from UVB-induced skin tumours41. Administration of TNF inhibitors to mice had a protective effect in urethane-induced pulmonary tumours and colonic tumours associated with chemically induced colitis42,43.

Additional evidence for a pro-tumorigenic role for TNF came from studies of gastric cancer associated with Helicobacter pylori infection. Gastric mucosal tissue samples from patients with chronic gastritis, gastric intestinal metaplasia, gastric dysplasia or gastric adenocarcinoma showed higher expression levels of TNF than samples from healthy individuals. Moreover, in the same individuals, a higher expression of TNF was associated with positivity for H. pylori, suggesting that the association of gastric cancer with H. pylori infection might in part be mediated by the induction of host TNF production by H. pylori44. Indeed, H. pylori can produce Tip-α, a protein that induces host TNF production and functions as a pro-tumorigenic factor in a manner possibly mediated by TNF signalling45.

Another line of evidence indicating that TNF is pro-tumorigenic comes from genetic studies of human populations. The -308G/A polymorphism in the promoter of TNF regulates TNF transcription; the less common A variant promotes transcription of the gene. As a result, individuals with the heterozygous G/A genotype have a two-fold increase in TNF production over individuals with the G/G genotype (as shown in a whole blood LPS stimulation assay46). Evidence from a number of case-control studies show that individuals with this promoter polymorphism have an increased risk of cancer risk. A study of 9,986 patients with gastrointestinal cancer (colorectal, oesophageal, gastric, hepatocellular or pancreatic cancer) and 15,511 healthy individuals showed that the A/A and G/A genotypes taken together confer 1.2-fold odds of gastrointestinal cancer compared with the G/G genotype (95% CI 1.1–1.4)47. Another study of 5,757 patients with prostate cancer and 6,137 healthy individuals showed that the A/A and G/A genotypes taken together confer 1.5-fold odds of prostate cancer compared with the G/G genotype (95% CI 1.1–2.1)48.

Role of TNF in inflammatory disease

The first randomized double-blind trial of a TNF inhibitor for the treatment of inflammatory disease was a study in RA in which TNF inhibition showed considerable efficacy49. This study demonstrated the therapeutic potential of TNF inhibitors for inflammatory disease and spurred further studies that expanded clinical indications to psoriatic arthritis, psoriasis, ankylosing spondylitis, juvenile RA. Crohn’s disease and ulcerative colitis50.

The clinical promise of TNF inhibitors also motivated studies into its mechanism of action and the role of TNF in inflammatory disease. TNF has a gatekeeping role in local tissue inflammation through its effects on vascular endothelial cells; it upregulates endothelial cell expression of surface adhesion molecules that recruit circulating leukocytes into the local tissue51. TNF also induces endothelial cell expression of cyclooxygenase 2 and subsequent prostaglandin release, resulting in vasodilation51.

TNF both promotes and restrains inflammatory processes through opposing functional consequences of signalling through its two surface receptors: TNF receptor 1 (TNFR1) and TNF receptor 2 (TNFR2). TNFR1 is expressed on all cells, whereas TNFR2 is expressed on a restricted subset of cell types including immune cells, endothelial cells and neurons. In general, TNF activates innate immune responses via TNFR1 signalling, while suppressing adaptive immune responses via TNFR2 signalling52. TNFR1 signalling in mesenchymal cells activate an innate immune response that promotes disease in RA, spondyloarthropathies (SpA) and inflammatory bowel disease (IBD)53. By contrast, TNFR2 signalling seems sufficient to ameliorate T cell-driven experimental autoimmune encephalomyelitis53, although the T cell subsets that mediate this effect is unknown. The suppressive effect of TNFR2 signalling on the adaptive immune response is thought to explain the unexpected aggravation of disease by TNF inhibition in a trial in patients with multiple sclerosis, as well as the sporadic occurrence of demyelinating disease in patients receiving a TNF inhibitor for other diseases52.

The duration, or chronicity, of TNF signalling helps to determine its effect on inflammatory pathology. Administration of TNF three times a week in NZB/W F1 lupus-prone mice delayed the onset of renal disease and led to improved survival at 3 months54. Similarly, TNF administered recurrently over 3 months protected non-obese diabetic mice against the development of diabetes55. In transgenic mice with T cells expressing a T cell receptor (TCR) specific for the influenza hemagglutinin antigen, repeated administration of TNF over 3 weeks yielded CD4+ T cells that had attenuated responses to TCR stimulation. Such T cells had a reduced capacity to proliferate and produce cytokines after TCR stimulation with the specific antigen56. Data from in vitro assays also showed that CD4+ T cells persistently exposed to TNF have attenuated TCR signalling. Thus, chronic TNF signalling might ameliorate autoimmune inflammation in part by reducing CD4+ T cell responsiveness to antigen stimulation.

How TNF modulates cells in inflammatory disease tissue has been most closely studied for RA and IBD. Synovial samples from patients with RA show robust immunohistochemistry staining for TNF57 and synovial fibroblasts and macrophages from patients with RA express TNF receptors58. TNF signals through TNFR1 on synovial fibroblasts to induce the production of IL-6, IL-8 and prostaglandin E259, all of which have pro-inflammatory properties. TNF has also been shown to signal through TNFR1 on CD4+ T cells to inhibit TH1 and TH17 cell differentiation and expansion in a mouse model of collagen-induced arthritis. In that model, TNF inhibitor treatment increased the numbers of TH1 and TH17 cells, which are considered pathogenic, but treatment also inhibited the accumulation of these cells in the joint60. This finding concurs with the observation in patients with RA that TNF inhibition can increase numbers of pathogenic T cells while ameliorating arthritis60. In studies of IBD, TNF inhibition induced macrophage differentiation to a regulatory phenotype that ameliorates intestinal inflammation61. TNF signalling through TNFR2 in various T cell subsets have different effects on experimental colitis. For example, TNFR2 signalling in regulatory T (Treg) cells and CD8+ T cells ameliorates colitis62,63, whereas in CD4+ effector T cells this signalling exacerbates colitis64.

ICI-induced inflammatory arthritis and ICI-induced colitis share molecular features with RA and IBD, respectively. Synovial biopsy samples from patients with ICI-induced inflammatory arthritis showed robust staining for TNF65, similar to synovial samples from patients with RA57. Similarly, colonic biopsy samples from patients with ICI-induced colitis showed evidence of increased TNF signalling in myeloid cells as compared with cells from healthy individuals66, which is in line with the finding that TNF signalling in macrophages helps to mediate IBD61. These shared molecular features provide support for borrowing TNF inhibition from the RA and IBD armamentarium to treat ICI-induced inflammatory arthritis and ICI-induced colitis.

TNF inhibitor use and cancer risk

In this section, we review data from clinical studies investigating the link between TNF inhibitor use and the risk of cancer and use this information to make inferences regarding the safety of TNF inhibitors during irAE treatment. Currently, limited data are available regarding the effect of TNF inhibitor treatment on the efficacy of ICI therapy (that is, the effect on cancer survival), but we can also draw from a large body of data on the link between TNF inhibition and cancer risk accumulated over many patient-years of experience using a TNF inhibitor for a wide range of indications.

Effects on ICI efficacy

In a study of 27 ICI-treated patients with melanoma who developed colitis and underwent TNF inhibitor treatment, the patients had a median progression free survival of 3 months, which is comparable to that reported in previous studies of ICI-treated patients with melanoma who did not undergo TNF inhibitor treatment67. Similarly, a retrospective study of patients with ICI-induced colitis found no difference in overall survival in those patients treated with corticosteroids alone (n=38) compared with those patients treated with corticosteroids plus a TNF inhibitor (n=23) (p=0.263)68. By contrast, in a study published in 2020, Verheijden and colleagues69 examined patients with grade ≥ 3 irAEs of various types (65 of whom received a TNF inhibitor and 157 of whom received corticosteroids only), the median overall survival was lower in the TNF inhibitor-treated group than in the corticosteroid-only group (17 versus 27 months; adjusted HR 1.61; 95% CI 1.03–2.51). However, the use of overall survival as the endpoint might have introduced confounders as some high-grade irAEs (such as colitis) have a higher mortality than others (such as high-grade endocrine toxicity, which can be treated with hormone replacement). In the Verheijden study, 61 of the 65 patients in the TNF inhibitor group had ICI-induce colitis, whereas the breakdown of irAE subtypes was not reported for the corticosteroid-only group. This study also failed to account for the time to irAE onset which, as a rule, is shorter for colitis than for endocrinopathies70,71 and is shorter for more severe than for less severe irAEs7072. This difference could have biased the results due to different follow-up times and treatment exposures between patients who did versus patients who did not require a TNF inhibitor73. For example, patients who did not require a TNF inhibitor might also be ones who developed later-onset irAEs; to be included in this group, patients had to have first survived long enough to acquire a later-onset irAE.

Empirically, it is notable that despite extensive efforts to identify tumour biomarkers that predict clinical response to ICI therapy, TNF has not emerged as one such biomarker7476. This finding suggests that TNF signalling in the tumour microenvironment has a neutral net effect on pathways that promote or inhibit the anti-tumour activity of ICI therapy, implying that TNF inhibition does not diminish this activity.

Effects on risk of cancer

Post-marketing surveillance of TNF inhibitor treatment in patients with RA, SpA, IBD and psoriasis provides data on the effects of decreasing levels of TNF on cancer risk. However, these studies might be biased by the fact that patients with autoimmune disease have a higher baseline risk of cancer than the general population, including a higher risk of lymphoma and lung cancer for patients with RA and a higher risk of non-melanoma skin cancer for patients with psoriasis7779. This elevated risk is thought to be caused by the presence of chronic inflammation, but shared genetic and environmental risk factors might also have a role77. Thus, disease severity (which is associated with the degree of inflammation) could confound the analysis of TNF inhibitor-associated cancer risk, at least in observational studies, if TNF inhibitor treatment is given preferentially to patients with more active disease. Risk assessment is also confounded by concomitant medications taken by patients on a TNF inhibitor, as some conventional DMARDS are also associated with risk of cancer. Examples include an elevated risk of hepatosplenic T cell lymphoma in TNF inhibitor-treated patients with IBD taking concomitant thiopurines (for example, azathioprine or 6-mercaptopurine)80 and an increased incidence of non-melanoma skin cancer in patients taking methotrexate81. Other factors that bias some studies of cancer risk in TNF inhibitor-treated patients include the use of self-reported cancer diagnoses, a lack of an active comparator group (for example, patients being treated with other DMARDs), a lack of adjustment for other treatments and comorbidities and inconsistent definitions of the time of TNF inhibitor exposure82.

Solid tumour malignancies

The above methodological limitations could, if anything, introduce spurious associations between TNF inhibition and cancer where there is none. It is therefore reassuring that most observational studies of patients with RA, SpA, IBD and psoriasis have failed to show any increased risk of cancer in patients being treated with a TNF inhibitor8386. For example, a systematic literature review and meta-analysis of 9 large RA registries (87,018 patient years in the TNF inhibitor treatment group and 50,734 patient years in the untreated control group) found no increased risk of solid tumours in the TNF inhibitor-treated patients (risk ratio 0.84; 95% CI 0.60–1.18)86. An early meta-analysis of clinical trials did show an elevated risk of cancer risk TNF inhibitor-treated patients (odds ratio 3.3; 95% CI, 1.2–9.1)87, but was criticized for not using individual level data and for using an average follow-up period, even though TNF inhibitor-treated patients had a longer follow-up period82,88. Subsequent meta-analyses of clinical trials have failed to show an increased cancer risk in patients being treated with a TNF inhibitor8992, and studies using large administrative datasets have been similarly negative93,94. One exception is the Wegener’s Granulomatosus Etanercept Trial (WGET), which compared cyclophosphamide alone to cyclophosphamide plus a TNF inhibitor (etanercept) for the treatment of granulomatosis with polyangiitis, and found a higher incidence of solid tumours in patients who received combination therapy95. This finding suggests that there might be synergistic toxicity when TNF inhibitors are used together with cytotoxic agents, similar to that observed when TNF inhibitors are combined with thiopurines80. Such synergistic effects might have relevance for patients with cancer receiving ICI therapy in combination with chemotherapy who require irAE management.

Lymphoma

A small number of studies have suggested patients being treated with a TNF inhibitor have an increased risk of developing a lymphoma. One published series described 48 cases of malignancy reported to the FDA in children on a TNF inhibitor, half of which were lymphomas96. Even though this case series did not control for confounding factors such as the risk of cancer associated with the underlying condition or concomitant medications, the FDA issued a black box warning of cancer risk in children being treated with a TNF inhibitor and later warned of an excess risk of developing especially rare hepatosplenic T cell lymphomas for children with IBD being treated with a combination of a TNF inhibitor and thiopurine97. An analysis of TNF inhibitor-treated patients with IBD in the French National Health insurance database also showed a higher rate of lymphoma (HR 2.41, 95% CI 1.60–3.64)98 compared with patients IBD who had no TNF inhibitor exposure; furthermore, a study of patients with juvenile idiopathic arthritis, IBD or psoriasis that used a Medicaid database hinted at a similar, albeit non-significant, increase in risk of lymphoma (adjusted HR 2.64, 95% CI 0.93–7.51)93. However, administrative datasets lack important information about confounders such as disease phenotype and severity, and many other prospective rheumatic disease registries have failed to show any increased risk of lymphoma in TNF inhibitor-treated patients83,99102.

Skin cancer

One study of a Swedish RA registry found an increased risk of melanoma in TNF inhibitor-treated patients with RA103. By contrast, no increased risk was found in a larger study that combined 11 European RA registries104, in a Scandinavian SpA registry study105 or in a large meta-analysis of RA clinical trials106. However, patients with psoriasis or RA undergoing TNF inhibitor treatment might have an increased risk of non-melanoma skin cancer107,108. With the possible exception of non-melanoma skin cancer109, studies have also failed to show an increased risk of cancer recurrence in patients with cancer being treated with a TNF inhibitor110,111.

In summary, although the assessment of TNF inhibitor-associated cancer risk is confounded by the excess background risk in patients with rheumatic diseases, available evidence suggests that these inhibitors do not increase the risk of solid tumours. The risk might be increased for lymphoma, particularly in patients with IBD being treated with a TNF inhibitor plus thiopurines, or for non-melanoma skin cancer in patients with psoriasis being treated with a TNF inhibitor. These data provide reassurance about the safety of TNF inhibition for the treatment of irAEs in the setting of ICI use, except perhaps in patients being treated for lymphoma.

irAE therapy: is TNF inhibition safe?

In this section, we review preclinical studies of the effects of TNF and TNF inhibition in the tumour microenvironment, and synthesize this information to make supporting inferences regarding the safety of TNF inhibitors in the treatment of irAEs. We summarize the immune and non-immune cell types present in the tumour microenvironment (Table 1), and discuss studies that address the effect of TNF, TNF inhibitors or ICIs on relevant cell types. We synthesize information from these studies into a model of interactions between cell types that also incorporates TNF inhibition and ICI therapy (Figure 1). This model enables us to systematically analyze the different paths by which TNF inhibitors and ICIs regulate cancer cell proliferation, and to make a prediction regarding the net effect of TNF inhibitors on tumour growth. We also discuss two preclinical studies that test this prediction.

Table 1:

Activities of various cell types in tumours and their effects on tumours.

Effect on tumour Cell type Mechanism Refs
Anti-tumour effects CTLs Kill cancer cells that display THE tumour-associated antigen and produce IFNγ, which has anti-tumour effects 146, 147
NK cells Kill cancer cells that over-express ligands recognized by NK cell receptors 148
TH cells Maintain adequate numbers of CTLs and promote CTL tumour infiltration 149
TH1 cells Produce IFNγ, which has anti-tumour effects, and promote the development of tumour antigen-specific CTLs 147,150
Dendritic cells Present tumour antigens to naïve antigen-specific T cells to induce their effector differentiation, including into TH1 cells and CTLs 150
Mixed effects TH2 cells Can be pro-tumorigenic or anti-tumorigenic depending on context 151
TH17 cells Can be pro-tumorigenic or anti-tumorigenic depending on context 133135,151
Endothelial cells Regulate immune cell infiltration into tumours via adhesion molecules 152
MSCs Regulate immune cell infiltration into tumours via chemokines 138
Pro-tumour effects TH cells
(non-canonical)
Follicular helper-like CD4PD1hi T cells are pro-tumorigenic 116
Treg cells Inhibit TH cell proliferation, differentiation of naïve CD4+ T cells into TH1 and TH2 cells, and CTL cytotoxicity 153, 153, 154
MDSCs Decrease the numbers of CTLs, TH cells and NK cells, increase the numbers of Treg cells and inhibit the activity of CTLs by inactivating TCRs 146,155
Tumour-associated macrophages Produce CCL22, which recruits T reg cells, and IL-10 and TGFβ, which are immunosuppressive cytokines 156

CTLs: CD8+ cytotoxic T lymphocytes. NK cells: natural killer cells. TH cells: CD4+ T helper cells. Treg cells: CD4+ regulatory T cells. MDSCs: myeloid-derived suppressor cells. MSCs: mesenchymal stromal cells.

Figure 1: Pro-tumour and anti-tumour effects of TNF inhibition and ICI therapy.

Figure 1:

A model of immune interactions in the tumour microenvironment. CTLs have direct cytotoxic effects on cancer cells, and moreover serve as a hub to integrate the indirect effects of other immune cell types, TNF inhibition and ICI therapy. Naïve CD8+ T cell differentiation replenishes the CTL pool. NK cells, TH1 cells, and tumour-associated macrophages have direct effects on cancer cell proliferation, and integrate the indirect effects of other immune cell types and TNF inhibition. TNF inhibition has a direct inhibitory effect on cancer cell proliferation. This model maps the different paths by which TNF inhibition exerts pro-tumour or anti-tumour effects. Each path starts with TNF inhibition exerting a direct effect on a cell type; *denotes that the effect of TNF inhibition is inferred from experimental data on TNF

The general pattern that emerges from this model is that TNF inhibition promotes the activity of anti-tumour immune cell types (such as CD8+ cytotoxic T lymphocytes (CTLs) and natural killer (NK) cells) while restraining the activity of immunosuppressive cell types (such as Treg cells, myeloid-derived suppressor cells (MDSCs) and mesenchymal stromal cells (MSCs), with an expected net anti-tumour effect that augments the benefits of ICIs. The effect of TNF inhibition on dendritic cells and naïve CD8+ T cells is an exception that might be pertinent in the setting of prolonged TNF inhibitor use. A further caveat is that although two preclinical studies have tested the net effect of TNF inhibition on tumour growth, studies in patients will be needed to validate this model in the clinical setting.

ICIs, TNF inhibitors and tumorigenesis

ICIs that target CTLA4, PD1, or PDL1 inhibit signalling through co-inhibitory112 pathways in tumour-infiltrating T cells113. Both inhibition of CTLA4 signalling and inhibition of PD1 signalling promote proliferation of exhausted PD1-positive CTLs, which subsequently become ‘reinvigorated’ to take on an effector-like phenotype associated with anti-tumour activity114. These CTLs continue to express PD1 but proliferate and express the activation surface markers HLA-DR and CD38115. ICIs have differing effects on different subtypes of T helper (TH) cells114,116, but overall promote the function of tumour-infiltrating T cells with a net anti-tumour effect. PD1 signalling in myeloid precursor cells promotes the development of MDSCs, which are pro-tumorigenic; thus, ICI might also exert their anti-tumour effect in part by inhibiting MDSC development117. The interactions between ICIs and tumour microenvironment immune cells including Treg cells, tumour-associated macrophages and MDSCs, have been comprehensively reviewed elsewhere74.

Effect of TNF on CTLs and NKs

TNF signals through TNFR2 on CTLs to promote activation-induced cell death118,119, thereby depleting the pool of anti-tumorigenic CTLs. TNF also signals through TNFR1 on CTLs to inhibit CTL infiltration into mouse B16K1 melanoma tumours, whereas TNF inhibition promotes CTL infiltration into tumours120. Moreover, TNF signalling in NK cells induces the expression of the co-inhibitory receptor TIM-3121 and decreases the expression of the activating cytotoxicity receptor NKp46122, thereby impairing NK cell anti-tumour activity. As TNF signalling restrains the function of these two anti-tumour immune cell types, it can be inferred that TNF inhibition could promote their function.

Effect of TNF on T cells

TNF signals through TNFR2 on Treg cells to promote Treg cell proliferation123 and thereby increases their suppressive function as a population123. TNF might also inhibit the suppressive function of Treg cells in vitro124,125, but this effect seems to be outweighed in vivo by the effect of TNF on Treg cell population expansion123,126. TNFR2-positive Treg cells (defined as CD4+ CD25+ TNFR2+ cells) suppress the proliferation of non-Treg cells (defined as CD4+ CD25 cells)127 to a greater extent than TNFR2-negative Treg cells, and are found at high density in Lewis lung carcinoma tumours127, as well as in human ovarian cancer ascites128. In a B16F10 mouse model of melanoma lung metastasis, administering TNF promotes pulmonary Treg cell proliferation and increases metastatic tumour growth129. A TNFR2 blocking antibody inhibits proliferation of Treg cells isolated from human peripheral blood, and kills Treg cells isolated from ascites of patients with ovarian cancer130, as well as from the blood of patients with cutaneous T cell lymphoma131. It can be inferred that TNF inhibition, by exerting the opposite effect as TNF, would restrain the function of immunosuppressive Treg cells.

TNF signals through TNFR2 on TH cells to promote TH cell proliferation and TH1 pro-inflammatory cytokine production64, and to promote their resistance to suppression by Treg cell132. The effect of TNF inhibitors on TH1 cells might then be an exception to the general pattern seen for other immune cell types, in that TNF inhibition would restrain the function of these anti-tumour immune cells. Nevertheless, as TNF inhibitors restrains Treg cells, which suppress TH1 cells, TNF inhibitors might indirectly promote TH1 cell function. TNF also signals through TNFR1 on TH cells to increase the relative number of TH17 cells and TH17 cytokine production133. Evidence suggests that TH17 cells can have both pro-tumour and anti-tumour effects: for example, TH17 cells recruit pro-tumour myeloid cells133, recruit anti-tumour TH1 cells134 and promote activation of anti-tumour CTLs135.

Effect of TNF on MDSCs and MSCs

TNF signals through TNFR2 on MDSCs to promote MDSC survival136 and their suppressive activity137. TNF inhibition impairs the growth of mouse FB61 fibrosarcoma tumours and simultaneously impairs peripheral accumulation of MDSCs, suggesting a correlation between an increased number of MDSCs and tumour growth in this model136. TNF also induces lymphoma-associated MSCs to express high levels of chemokine ligands for the chemokine receptor CCR2. These chemokines recruit CCR2-expressing tumour-associated macrophages into the tumour with overall pro-tumorigenic effect138. It can be inferred that TNF inhibition would then restrain the function of immunosuppressive MDSCs and MSCs.

Effect of TNF on cancer cells

Finally, TNF signalling in cancer cells helps them evade immune surveillance while promoting their survival and proliferation130, with overall pro-tumorigenic effect. In several cancer cell lines, TNF signalling increases the surface expression of PDL1, an immune checkpoint ligand that helps cancer cells evade T cell immune surveillance139. In human melanoma lesions, PDL1 gene expression positively correlates with TNF gene expression140. TNF signalling also promotes de-differentiation of melanoma cells accompanied by loss of immunogenicity, which helps cancer cells to evade T cell immune surveillance141. Hence, TNF inhibition might exert a direct inhibitory effect on cancer cells

Overall, the model of interactions depicted in Figure 1 predicts that TNF inhibitors augments ICI anti-tumour activity by promoting CTL activity, and that TNF inhibitors promote additional anti-tumour activity through ICI-independent pathways.

Prolonged use of TNF inhibitors

In contrast to the short-term effects of TNF inhibition on tumorigenesis, prolonged use of a TNF inhibitor might deplete the anti-tumour CTL pool via inhibition of naïve CD8+ T cell differentiation into CTLs, leading to decreased numbers of tumour antigen-specific CTLs relative to that of tumour antigen-specific naïve CD8+ T cells. This depletion might occur because TNF signals through TNFR2 on naïve CD8+ T cells to provide a co-stimulatory signal that promotes TCR-mediated proliferation, activation and differentiation into CTLs142,143. Moreover, TNF signalling supports the dendritic cells that promote naïve CD8+ T cell differentiation. TNF signalling through TNFR1 in immature dendritic cells induces their maturation, whereas TNF signalling through both TNFR1 and TNFR2 in dendritic cells promote their survival144.

Although a CTL depletion effect has not been investigated in preclinical models or in patients, this effect is of particular concern with regard to chronic use of TNF inhibitors and has relevance to the management of ICI-induced inflammatory arthritis, which is often persistent. It would be worthwhile to study whether CTL depletion occurs in patients receiving ICI therapy plus prolonged TNF inhibitor therapy compared with patients receiving ICI therapy alone or ICI therapy plus prolonged therapy with corticosteroids. If CTL depletion is observed, another important question would be whether this effect correlates with clinical outcomes such as progression-free survival.

Data from preclinical models

The predictions outlined in our model (Figure 1) have been tested in a mouse model of engrafted B16K1 melanoma in which the tumours are being treated with an anti-PD1 therapy140. In this model, treatment with a TNF inhibitor augmented the anti-tumour activity of ICI therapy, as assessed by the proportion of tumours that completely regress and by overall survival. Moreover, this improved ICI efficacy was associated with an increased proportion of CTLs out of the total number of cells in the tumours, and a decreased amount of cell death of these CTLs, suggesting that the improved ICI efficacy was attributable to a TNF inhibitor-mediated increase in CTL activity. The researchers also found that TNF inhibitor treatment alone did not have anti-tumour activity in this tumour model, contradicting our prediction that TNF inhibitors have ICI-independent anti-tumour activity. However, additional factors might be present that contribute to the tumour response to TNF inhibition that are not accounted for in our model.

In another preclinical study, researchers created a mouse model of ICI-induced colitis, in which the mice were given a combination of anti-CTLA4 and anti-PD1 therapy to treat engrafted MC38 tumours145 and were concomitantly given dextran sulfate sodium to induce colitis, which is exacerbated by the combination ICI treatment. In this model, TNF inhibitor treatment both ameliorated colitis and augmented ICI anti-tumour activity. The improved ICI efficacy was associated with an increase in tumour antigen-specific CTLs in the tumours, and decreased cell death of these CTLs. A limitation of both of the studies described in this section is that the duration of TNF inhibitor treatment was at most 10 days, so the studies did not address the question of what the effect of chronic TNF inhibitor treatment is on CTL activity or ICI efficacy. Preclinical and clinical studies that look at an extended duration of TNF inhibitor treatment would be valuable.

Conclusion

TNF is a pleiotropic cytokine with pro-inflammatory and immunosuppressive effects in inflammatory disease and cancer. TNF inhibitors are effective treatment for a number of inflammatory diseases including RA, IBD and ICI-induced inflammatory arthritis. Multiple clinical studies of TNF inhibitors in patients with inflammatory disease support the hypothesis that TNF inhibitor poses a relatively low risk of cancer, but limited clinical data is available regarding its risk profile in patients with cancer undergoing ICI therapy. TNF can promote or inhibit the activities of the immune cells and cancer cells within tumours. The net effect of TNF inhibition on tumorigenesis might be positive or negative depending on qualitative (that is, the presence of specific cell types) and quantitative (that is, the local concentration of TNF) factors. Moreover, acute versus chronic TNF inhibition might have opposing effects on tumour growth. Preclinical models can be extrapolated to the clinic only to the extent that the qualitative and quantitative details of the experimental model match those found in patients. Despite these caveats, the bulk of current data supports two conclusions: TNF inhibitor treatment of rheumatic diseases does not seem to increase the risk of cancer, except for non-melanoma skin cancer and possibly lymphoma, and preclinical data suggest that short-term TNF inhibitor treatment of irAEs should not diminish the anti-cancer efficacy of ICI therapy. Thus, short courses of TNF inhibitors might be safe to use in treatment of ICI-associated irAEs. Further studies in preclinical models are required to directly assess the safety of long-term TNF inhibitor use in the context of ICI cancer treatment. Clinical studies that directly assess the effect of TNF inhibitor treatment on ICI efficacy are required to draw conclusions regarding the short-term and long-term safety of TNF inhibitor treatment for irAEs. Preclinical studies provide evidence that TNF inhibitors, despite their efficacy in ameliorating irAEs, do not also restrain anti-cancer immune activity. The data from these studies suggests that different arms of the immune response are important for anti-self versus anti-cancer activities, with TNF inhibition restraining some arms of the immune response while promoting or having a neutral effect on others. The cellular and molecular details of how the pleiotropic effects of TNF signalling interact with different arms of the immune response remain to be fully delineated. A more complete map of these interactions might reveal novel drug targets for the treatment of inflammatory disease and cancer.

Acknowledgements

The work of J.D.W Is funded in part through the NIH/NCI Cancer Center Support Grant P30 CA008748. J.D.W. is also affiliated with: Ludwig Collaborative and Swim Across America Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Parker Institute for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, New York, NY, USA. The authors would like to thank L.B. Ivashkiv at the Hospital for Special Surgery for their comments on the manuscript.

Footnotes

Competing interests

A.Y. Chen declares no competing interests. J.D. Wolchok is a consultant for Adaptive Biotech, Amgen, Apricity, Arsenal, Ascentage Pharma, Astellas, AstraZeneca, Bayer, Boehriner Ingelheim, Bristol Myers Squibb, Eli Lilly, F Star, Imvaq, Kyowa Hakko Kirin, Merck, Neon Therapuetics, Psioxus, Recepta, Sellas, Serametrix, Surface Oncology, Syndax and Syntalogic, Takara Bio, Trieza and Truvax; receives research support from AstraZeneca, Bristol Myers Squibb and Sephora; and has equity in Adaptive Biotechnologies, Apricity, Arsenal, Beigene, Imvaq, Linneaus, Tizona Pharmaceuticals. A.R. Bass declares no competing interests.

References

  • 1.Arnaud-Coffin P et al. A systematic review of adverse events in randomized trials assessing immune checkpoint inhibitors. Int J Cancer 145, 639–648, doi: 10.1002/ijc.32132 (2019). [DOI] [PubMed] [Google Scholar]
  • 2.Postow MA, Sidlow R & Hellmann MD Immune-Related Adverse Events Associated with Immune Checkpoint Blockade. N Engl J Med 378, 158–168, doi: 10.1056/NEJMra1703481 (2018). [DOI] [PubMed] [Google Scholar]
  • 3.Chan KK & Bass AR Autoimmune complications of immunotherapy: pathophysiology and management. BMJ 369, m736, doi: 10.1136/bmj.m736 (2020). [DOI] [PubMed] [Google Scholar]
  • 4.Larkin J, Hodi FS & Wolchok JD Combined Nivolumab and Ipilimumab or Monotherapy in Untreated Melanoma. N Engl J Med 373, 1270–1271, doi: 10.1056/NEJMc1509660 (2015). [DOI] [PubMed] [Google Scholar]
  • 5.Kostine M et al. Rheumatic disorders associated with immune checkpoint inhibitors in patients with cancer-clinical aspects and relationship with tumour response: a single-centre prospective cohort study. Ann Rheum Dis 77, 393–398, doi: 10.1136/annrheumdis-2017-212257 (2018). [DOI] [PubMed] [Google Scholar]
  • 6.Cappelli LC et al. Clinical presentation of immune checkpoint inhibitor-induced inflammatory arthritis differs by immunotherapy regimen. Semin Arthritis Rheum 48, 553–557, doi: 10.1016/j.semarthrit.2018.02.011 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Ghosh N et al. Checkpoint Inhibitor-Associated Arthritis: A Systematic Review of Case Reports and Case Series. J Clin Rheumatol, doi: 10.1097/RHU.0000000000001370 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Thompson JA et al. NCCN Guidelines Insights: Management of Immunotherapy-Related Toxicities. Featured Updates to the NCCN Guidelines 18 (2020). [DOI] [PubMed] [Google Scholar]
  • 9.Smith MH & Bass AR Arthritis After Cancer Immunotherapy: Symptom Duration and Treatment Response. Arthritis Care Res (Hoboken) 71, 362–366, doi: 10.1002/acr.23467 (2019). [DOI] [PubMed] [Google Scholar]
  • 10.Braaten TJ et al. Immune checkpoint inhibitor-induced inflammatory arthritis persists after immunotherapy cessation. Ann Rheum Dis, doi: 10.1136/annrheumdis-2019-216109 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Kim ST et al. Successful treatment of arthritis induced by checkpoint inhibitors with tocilizumab: a case series. Ann Rheum Dis 76, 2061–2064, doi: 10.1136/annrheumdis-2017-211560 (2017). [DOI] [PubMed] [Google Scholar]
  • 12.Roberts J et al. Hydroxychloroquine is a safe and effective steroid-sparing agent for immune checkpoint inhibitor-induced inflammatory arthritis. Clin Rheumatol 38, 1513–1519, doi: 10.1007/s10067-019-04451-2 (2019). [DOI] [PubMed] [Google Scholar]
  • 13.Teulings HE et al. Vitiligo-like depigmentation in patients with stage III-IV melanoma receiving immunotherapy and its association with survival: a systematic review and meta-analysis. J Clin Oncol 33, 773–781, doi: 10.1200/JCO.2014.57.4756 (2015). [DOI] [PubMed] [Google Scholar]
  • 14.Zhou X et al. Are immune-related adverse events associated with the efficacy of immune checkpoint inhibitors in patients with cancer? A systematic review and meta-analysis. BMC Med 18, 87, doi: 10.1186/s12916-020-01549-2 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Haratani K et al. Association of Immune-Related Adverse Events With Nivolumab Efficacy in Non-Small-Cell Lung Cancer. JAMA Oncol 4, 374–378, doi: 10.1001/jamaoncol.2017.2925 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Horvat TZ et al. Immune-Related Adverse Events, Need for Systemic Immunosuppression, and Effects on Survival and Time to Treatment Failure in Patients With Melanoma Treated With Ipilimumab at Memorial Sloan Kettering Cancer Center. J Clin Oncol 33, 3193–3198, doi: 10.1200/JCO.2015.60.8448 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Mahmood SS et al. Myocarditis in Patients Treated With Immune Checkpoint Inhibitors. J Am Coll Cardiol 71, 1755–1764, doi: 10.1016/j.jacc.2018.02.037 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Marthey L et al. Cancer Immunotherapy with Anti-CTLA-4 Monoclonal Antibodies Induces an Inflammatory Bowel Disease. J Crohns Colitis 10, 395–401, doi: 10.1093/ecco-jcc/jjv227 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Faje AT et al. High-dose glucocorticoids for the treatment of ipilimumab-induced hypophysitis is associated with reduced survival in patients with melanoma. Cancer 124, 3706–3714, doi: 10.1002/cncr.31629 (2018). [DOI] [PubMed] [Google Scholar]
  • 20.Arbour KC et al. Impact of Baseline Steroids on Efficacy of Programmed Cell Death-1 and Programmed Death-Ligand 1 Blockade in Patients With Non-Small-Cell Lung Cancer. J Clin Oncol 36, 2872–2878, doi: 10.1200/JCO.2018.79.0006 (2018). [DOI] [PubMed] [Google Scholar]
  • 21.Carswell EA et al. An endotoxin-induced serum factor that causes necrosis of tumors. Proc Natl Acad Sci U S A 72, 3666–3670, doi: 10.1073/pnas.72.9.3666 (1975). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Nauts HC, Swift WE & Coley BL The treatment of malignant tumors by bacterial toxins as developed by the late William B. Coley, M.D., reviewed in the light of modern research. Cancer Res 6, 205–216 (1946). [PubMed] [Google Scholar]
  • 23.Shear MJ & Perrault A Chemical Treatment of Tumors. IX. Reactions of Mice with Primary Subcutaneous Tumors to Injection of a Hemorrhage-Producing Bacterial Polysaccharide. JNCI: Journal of the National Cancer Institute 4 (1944). [Google Scholar]
  • 24.O’Malley WE, Achinstein B & Shear MJ Journal of the National Cancer Institute, Vol. 29, 1962: Action of bacterial polysaccharide on tumors. II. Damage of sarcoma 37 by serum of mice treated with Serratia marcescens polysaccharide, and induced tolerance. Nutr Rev 46, 389–391, doi: 10.1111/j.1753-4887.1988.tb05376.x (1988). [DOI] [PubMed] [Google Scholar]
  • 25.Pennica D et al. Human tumour necrosis factor: precursor structure, expression and homology to lymphotoxin. Nature 312, 724–729, doi: 10.1038/312724a0 (1984). [DOI] [PubMed] [Google Scholar]
  • 26.Fransen L et al. Molecular cloning of mouse tumour necrosis factor cDNA and its eukaryotic expression. Nucleic Acids Res 13, 4417–4429, doi: 10.1093/nar/13.12.4417 (1985). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Beutler B et al. Identity of tumour necrosis factor and the macrophage-secreted factor cachectin. Nature 316, 552–554, doi: 10.1038/316552a0 (1985). [DOI] [PubMed] [Google Scholar]
  • 28.Brennan FM, Chantry D, Jackson A, Maini R & Feldmann M Inhibitory effect of TNF alpha antibodies on synovial cell interleukin-1 production in rheumatoid arthritis. Lancet 2, 244–247, doi: 10.1016/s0140-6736(89)90430-3 (1989). [DOI] [PubMed] [Google Scholar]
  • 29.Gamm H, Lindemann A, Mertelsmann R & Herrmann F Phase I trial of recombinant human tumour necrosis factor alpha in patients with advanced malignancy. Eur J Cancer 27, 856–863 (1991). [DOI] [PubMed] [Google Scholar]
  • 30.Arican O, Aral M, Sasmaz S & Ciragil P Serum levels of TNF-alpha, IFN-gamma, IL-6, IL-8, IL-12, IL-17, and IL-18 in patients with active psoriasis and correlation with disease severity. Mediators Inflamm 2005, 273–279, doi: 10.1155/MI.2005.273 (2005). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Waters JP, Pober JS & Bradley JR Tumour necrosis factor and cancer. J Pathol 230, 241–248, doi: 10.1002/path.4188 (2013). [DOI] [PubMed] [Google Scholar]
  • 32.Robaye B, Mosselmans R, Fiers W, Dumont JE & Galand P Tumor necrosis factor induces apoptosis (programmed cell death) in normal endothelial cells in vitro. Am J Pathol 138, 447–453 (1991). [PMC free article] [PubMed] [Google Scholar]
  • 33.Balkwill F Tumour necrosis factor and cancer. Nat Rev Cancer 9, 361–371, doi: 10.1038/nrc2628 (2009). [DOI] [PubMed] [Google Scholar]
  • 34.Wu H, Tschopp J & Lin SC Smac mimetics and TNFalpha: a dangerous liaison? Cell 131, 655–658, doi: 10.1016/j.cell.2007.10.042 (2007). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Ratner A & Clark WR Role of TNF-alpha in CD8+ cytotoxic T lymphocyte-mediated lysis. J Immunol 150, 4303–4314 (1993). [PubMed] [Google Scholar]
  • 36.Caron G et al. Human NK cells constitutively express membrane TNF-alpha (mTNFalpha) and present mTNFalpha-dependent cytotoxic activity. Eur J Immunol 29, 3588–3595, doi: (1999). [DOI] [PubMed] [Google Scholar]
  • 37.Freedman MH et al. Central role of tumour necrosis factor, GM-CSF, and interleukin 1 in the pathogenesis of juvenile chronic myelogenous leukaemia. Br J Haematol 80, 40–48, doi: 10.1111/j.1365-2141.1992.tb06398.x (1992). [DOI] [PubMed] [Google Scholar]
  • 38.Frater-Schroder M, Risau W, Hallmann R, Gautschi P & Bohlen P Tumor necrosis factor type alpha, a potent inhibitor of endothelial cell growth in vitro, is angiogenic in vivo. Proc Natl Acad Sci U S A 84, 5277–5281, doi: 10.1073/pnas.84.15.5277 (1987). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Li B et al. Low levels of tumor necrosis factor alpha increase tumor growth by inducing an endothelial phenotype of monocytes recruited to the tumor site. Cancer Res 69, 338–348, doi: 10.1158/0008-5472.CAN-08-1565 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Moore RJ et al. Mice deficient in tumor necrosis factor-alpha are resistant to skin carcinogenesis. Nat Med 5, 828–831, doi: 10.1038/10552 (1999). [DOI] [PubMed] [Google Scholar]
  • 41.Starcher B Role for tumour necrosis factor-alpha receptors in ultraviolet-induced skin tumours. Br J Dermatol 142, 1140–1147, doi: 10.1046/j.1365-2133.2000.03539.x (2000). [DOI] [PubMed] [Google Scholar]
  • 42.Karabela SP et al. Neutralization of tumor necrosis factor bioactivity ameliorates urethane-induced pulmonary oncogenesis in mice. Neoplasia 13, 1143–1151, doi: 10.1593/neo.111224 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Popivanova BK et al. Blocking TNF-alpha in mice reduces colorectal carcinogenesis associated with chronic colitis. J Clin Invest 118, 560–570, doi: 10.1172/JCI32453 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Senthilkumar C, Niranjali S, Jayanthi V, Ramesh T & Devaraj H Molecular and histological evaluation of tumor necrosis factor-alpha expression in Helicobacter pylori-mediated gastric carcinogenesis. J Cancer Res Clin Oncol 137, 577–583, doi: 10.1007/s00432-010-0921-9 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Suganuma M, Kuzuhara T, Yamaguchi K & Fujiki H Carcinogenic role of tumor necrosis factor-alpha inducing protein of Helicobacter pylori in human stomach. J Biochem Mol Biol 39, 1–8 (2006). [DOI] [PubMed] [Google Scholar]
  • 46.Louis E et al. Tumour necrosis factor (TNF) gene polymorphism influences TNF-alpha production in lipopolysaccharide (LPS)-stimulated whole blood cell culture in healthy humans. Clin Exp Immunol 113, 401–406, doi: 10.1046/j.1365-2249.1998.00662.x (1998). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Guo XF et al. TNF-alpha-308 polymorphism and risk of digestive system cancers: a meta-analysis. World J Gastroenterol 19, 9461–9471, doi: 10.3748/wjg.v19.i48.9461 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Ma L et al. Association between tumor necrosis factor-alpha gene polymorphisms and prostate cancer risk: a meta-analysis. Diagn Pathol 9, 74, doi: 10.1186/1746-1596-9-74 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Elliott MJ et al. Randomised double-blind comparison of chimeric monoclonal antibody to tumour necrosis factor alpha (cA2) versus placebo in rheumatoid arthritis. Lancet 344, 1105–1110, doi: 10.1016/s0140-6736(94)90628-9 (1994). [DOI] [PubMed] [Google Scholar]
  • 50.Monaco C, Nanchahal J, Taylor P & Feldmann M Anti-TNF therapy: past, present and future. Int Immunol 27, 55–62, doi: 10.1093/intimm/dxu102 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Bradley JR TNF-mediated inflammatory disease. J Pathol 214, 149–160, doi: 10.1002/path.2287 (2008). [DOI] [PubMed] [Google Scholar]
  • 52.Kalliolias GD & Ivashkiv LB TNF biology, pathogenic mechanisms and emerging therapeutic strategies. Nat Rev Rheumatol 12, 49–62, doi: 10.1038/nrrheum.2015.169 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Apostolaki M, Armaka M, Victoratos P & Kollias G Cellular mechanisms of TNF function in models of inflammation and autoimmunity. Curr Dir Autoimmun 11, 1–26, doi: 10.1159/000289195 (2010). [DOI] [PubMed] [Google Scholar]
  • 54.Gordon C, Ranges GE, Greenspan JS & Wofsy D Chronic therapy with recombinant tumor necrosis factor-alpha in autoimmune NZB/NZW F1 mice. Clin Immunol Immunopathol 52, 421–434, doi: 10.1016/0090-1229(89)90157-8 (1989). [DOI] [PubMed] [Google Scholar]
  • 55.Jacob CO, Aiso S, Michie SA, McDevitt HO & Acha-Orbea H Prevention of diabetes in nonobese diabetic mice by tumor necrosis factor (TNF): similarities between TNF-alpha and interleukin 1. Proc Natl Acad Sci U S A 87, 968–972, doi: 10.1073/pnas.87.3.968 (1990). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Cope AP et al. Chronic tumor necrosis factor alters T cell responses by attenuating T cell receptor signaling. J Exp Med 185, 1573–1584, doi: 10.1084/jem.185.9.1573 (1997). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Chu CQ, Field M, Feldmann M & Maini RN Localization of tumor necrosis factor alpha in synovial tissues and at the cartilage-pannus junction in patients with rheumatoid arthritis. Arthritis Rheum 34, 1125–1132, doi: 10.1002/art.1780340908 (1991). [DOI] [PubMed] [Google Scholar]
  • 58.Alsalameh S et al. Distribution of TNF-alpha, TNF-R55 and TNF-R75 in the rheumatoid synovial membrane: TNF receptors are localized preferentially in the lining layer; TNF-alpha is distributed mainly in the vicinity of TNF receptors in the deeper layers. Scand J Immunol 49, 278–285, doi: 10.1046/j.1365-3083.1999.00458.x (1999). [DOI] [PubMed] [Google Scholar]
  • 59.Kunisch E et al. Predominant activation of MAP kinases and pro-destructive/pro-inflammatory features by TNF alpha in early-passage synovial fibroblasts via TNF receptor-1: failure of p38 inhibition to suppress matrix metalloproteinase-1 in rheumatoid arthritis. Ann Rheum Dis 66, 1043–1051, doi: 10.1136/ard.2006.062521 (2007). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Notley CA et al. Blockade of tumor necrosis factor in collagen-induced arthritis reveals a novel immunoregulatory pathway for Th1 and Th17 cells. J Exp Med 205, 2491–2497, doi: 10.1084/jem.20072707 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Koelink PJ et al. Anti-TNF therapy in IBD exerts its therapeutic effect through macrophage IL-10 signalling. Gut 69, 1053–1063, doi: 10.1136/gutjnl-2019-318264 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Housley WJ et al. Natural but not inducible regulatory T cells require TNF-alpha signaling for in vivo function. J Immunol 186, 6779–6787, doi: 10.4049/jimmunol.1003868 (2011). [DOI] [PubMed] [Google Scholar]
  • 63.Punit S et al. Tumor Necrosis Factor Receptor 2 Restricts the Pathogenicity of CD8(+) T Cells in Mice With Colitis. Gastroenterology 149, 993–1005 e1002, doi: 10.1053/j.gastro.2015.06.004 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Chen X et al. TNFR2 expression by CD4 effector T cells is required to induce full-fledged experimental colitis. Sci Rep 6, 32834, doi: 10.1038/srep32834 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Murray-Brown W et al. Nivolumab-induced synovitis is characterized by florid T cell infiltration and rapid resolution with synovial biopsy-guided therapy. J Immunother Cancer 8, doi: 10.1136/jitc-2019-000281 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Luoma AM et al. Molecular Pathways of Colon Inflammation Induced by Cancer Immunotherapy. Cell, doi: 10.1016/j.cell.2020.06.001 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Lesage C et al. Incidence and Clinical Impact of Anti-TNFalpha Treatment of Severe Immune Checkpoint Inhibitor-induced Colitis in Advanced Melanoma: The Mecolit Survey. J Immunother, doi: 10.1097/CJI.0000000000000268 (2019). [DOI] [PubMed] [Google Scholar]
  • 68.Wang Y et al. Immune-checkpoint inhibitor-induced diarrhea and colitis in patients with advanced malignancies: retrospective review at MD Anderson. J Immunother Cancer 6, 37, doi: 10.1186/s40425-018-0346-6 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Verheijden RJ et al. Association of anti-TNF with decreased survival in steroid refractory ipilimumab and anti-PD1 treated patients in the Dutch Melanoma Treatment Registry. Clin Cancer Res, doi: 10.1158/1078-0432.CCR-19-3322 (2020). [DOI] [PubMed] [Google Scholar]
  • 70.Weber JS et al. Safety Profile of Nivolumab Monotherapy: A Pooled Analysis of Patients With Advanced Melanoma. J Clin Oncol 35, 785–792, doi: 10.1200/JCO.2015.66.1389 (2017). [DOI] [PubMed] [Google Scholar]
  • 71.Sznol M et al. Pooled Analysis Safety Profile of Nivolumab and Ipilimumab Combination Therapy in Patients With Advanced Melanoma. J Clin Oncol 35, 3815–3822, doi: 10.1200/JCO.2016.72.1167 (2017). [DOI] [PubMed] [Google Scholar]
  • 72.Wang DY et al. Fatal Toxic Effects Associated With Immune Checkpoint Inhibitors: A Systematic Review and Meta-analysis. JAMA Oncol 4, 1721–1728, doi: 10.1001/jamaoncol.2018.3923 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Eggermont AMM et al. Association Between Immune-Related Adverse Events and Recurrence-Free Survival Among Patients With Stage III Melanoma Randomized to Receive Pembrolizumab or Placebo: A Secondary Analysis of a Randomized Clinical Trial. JAMA Oncol, doi: 10.1001/jamaoncol.2019.5570 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Havel JJ, Chowell D & Chan TA The evolving landscape of biomarkers for checkpoint inhibitor immunotherapy. Nat Rev Cancer 19, 133–150, doi: 10.1038/s41568-019-0116-x (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Bridge JA, Lee JC, Daud A, Wells JW & Bluestone JA Cytokines, Chemokines, and Other Biomarkers of Response for Checkpoint Inhibitor Therapy in Skin Cancer. Front Med (Lausanne) 5, 351, doi: 10.3389/fmed.2018.00351 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Gibney GT, Weiner LM & Atkins MB Predictive biomarkers for checkpoint inhibitor-based immunotherapy. Lancet Oncol 17, e542–e551, doi: 10.1016/S1470-2045(16)30406-5 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Baecklund E, Smedby KE, Sutton LA, Askling J & Rosenquist R Lymphoma development in patients with autoimmune and inflammatory disorders--what are the driving forces? Seminars in cancer biology 24, 61–70, doi: 10.1016/j.semcancer.2013.12.001 (2014). [DOI] [PubMed] [Google Scholar]
  • 78.Smitten AL, Simon TA, Hochberg MC & Suissa S A meta-analysis of the incidence of malignancy in adult patients with rheumatoid arthritis. Arthritis Res Ther 10, R45, doi: 10.1186/ar2404 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Pouplard C et al. Risk of cancer in psoriasis: a systematic review and meta-analysis of epidemiological studies. J Eur Acad Dermatol Venereol 27Suppl 3, 36–46, doi: 10.1111/jdv.12165 (2013). [DOI] [PubMed] [Google Scholar]
  • 80.Deepak P et al. T-cell non-Hodgkin’s lymphomas reported to the FDA AERS with tumor necrosis factor-alpha (TNF-α) inhibitors: results of the REFURBISH study. The American journal of gastroenterology 108, 99–105, doi: 10.1038/ajg.2012.334 (2013). [DOI] [PubMed] [Google Scholar]
  • 81.Solomon DH et al. Adverse Effects of Low-Dose Methotrexate: A Randomized Trial. Ann Intern Med, doi: 10.7326/M19-3369 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Solomon DH, Mercer E & Kavanaugh A Observational studies on the risk of cancer associated with tumor necrosis factor inhibitors in rheumatoid arthritis: a review of their methodologies and results. Arthritis Rheum 64, 21–32, doi: 10.1002/art.30653 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Askling J et al. Anti-tumour necrosis factor therapy in rheumatoid arthritis and risk of malignant lymphomas: relative risks and time trends in the Swedish Biologics Register. Ann Rheum Dis 68, 648–653, doi: 10.1136/ard.2007.085852 (2009). [DOI] [PubMed] [Google Scholar]
  • 84.Nyboe Andersen N et al. Association between tumor necrosis factor-alpha antagonists and risk of cancer in patients with inflammatory bowel disease. JAMA 311, 2406–2413, doi: 10.1001/jama.2014.5613 (2014). [DOI] [PubMed] [Google Scholar]
  • 85.Haynes K et al. Tumor necrosis factor alpha inhibitor therapy and cancer risk in chronic immune-mediated diseases. Arthritis Rheum 65, 48–58, doi: 10.1002/art.37740 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.de La Forest Divonne M, Gottenberg JE & Salliot C Safety of biologic DMARDs in RA patients in real life: A systematic literature review and meta-analyses of biologic registers. Joint, bone, spine : revue du rhumatisme 84, 133–140, doi: 10.1016/j.jbspin.2016.02.028 (2017). [DOI] [PubMed] [Google Scholar]
  • 87.Bongartz T et al. Anti-TNF antibody therapy in rheumatoid arthritis and the risk of serious infections and malignancies: systematic review and meta-analysis of rare harmful effects in randomized controlled trials. JAMA 295, 2275–2285, doi: 10.1001/jama.295.19.2275 (2006). [DOI] [PubMed] [Google Scholar]
  • 88.Dixon W & Silman A Is there an association between anti-TNF monoclonal antibody therapy in rheumatoid arthritis and risk of malignancy and serious infection? Commentary on the meta-analysis by Bongartz et al. Arthritis research & therapy 8, 111, doi: 10.1186/ar2026 (2006). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Dommasch ED et al. The risk of infection and malignancy with tumor necrosis factor antagonists in adults with psoriatic disease: a systematic review and meta-analysis of randomized controlled trials. J Am Acad Dermatol 64, 1035–1050, doi: 10.1016/j.jaad.2010.09.734 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Lichtenstein GR et al. A pooled analysis of infections, malignancy, and mortality in infliximab- and immunomodulator-treated adult patients with inflammatory bowel disease. The American journal of gastroenterology 107, 1051–1063, doi: 10.1038/ajg.2012.89 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Maneiro JR, Souto A & Gomez-Reino JJ Risks of malignancies related to tofacitinib and biological drugs in rheumatoid arthritis: Systematic review, meta-analysis, and network meta-analysis. Seminars in arthritis and rheumatism 47, 149–156, doi: 10.1016/j.semarthrit.2017.02.007 (2017). [DOI] [PubMed] [Google Scholar]
  • 92.Hou LQ et al. The Comparative Safety of TNF Inhibitors in Ankylosing Spondylitis-a Meta-Analysis Update of 14 Randomized Controlled Trials. Clinical reviews in allergy & immunology 54, 234–243, doi: 10.1007/s12016-017-8623-6 (2018). [DOI] [PubMed] [Google Scholar]
  • 93.Beukelman T et al. Risk of malignancy associated with paediatric use of tumour necrosis factor inhibitors. Annals of the rheumatic diseases 77, 1012–1016, doi: 10.1136/annrheumdis-2017-212613 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Jung SM, Kwok SK, Ju JH, Park YB & Park SH Risk of malignancy in patients with rheumatoid arthritis after anti-tumor necrosis factor therapy: results from Korean National Health Insurance claims data. The Korean journal of internal medicine 34, 669–677, doi: 10.3904/kjim.2016.374 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Silva F et al. Solid malignancies among etanercept-treated patients with granulomatosis with polyangiitis (Wegener’s): long-term followup of a multicenter longitudinal cohort. Arthritis and rheumatism 63, 2495–2503, doi: 10.1002/art.30394 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Diak P et al. Tumor necrosis factor alpha blockers and malignancy in children: forty-eight cases reported to the Food and Drug Administration. Arthritis Rheum 62, 2517–2524, doi: 10.1002/art.27511 (2010). [DOI] [PubMed] [Google Scholar]
  • 97.FDA. FDA Drug Safety Communication: Safety Review update on reports of Hepatosplenic T-Cell Lymphoma in adolescents and young adults receiving tumor necrosis factor (TNF) blockers, azathioprine and/or mercaptopurine, <http://wayback.archive-it.org/7993/20170112031812/http://www.fda.gov/Drugs/DrugSafety/ucm250913.htm> (2011).
  • 98.Lemaitre M et al. Association Between Use of Thiopurines or Tumor Necrosis Factor Antagonists Alone or in Combination and Risk of Lymphoma in Patients With Inflammatory Bowel Disease. JAMA 318, 1679–1686, doi: 10.1001/jama.2017.16071 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Wolfe F & Michaud K The effect of methotrexate and anti-tumor necrosis factor therapy on the risk of lymphoma in rheumatoid arthritis in 19,562 patients during 89,710 person-years of observation. Arthritis and rheumatism 56, 1433–1439, doi: 10.1002/art.22579 (2007). [DOI] [PubMed] [Google Scholar]
  • 100.Hellgren K et al. Rheumatoid Arthritis and Risk of Malignant Lymphoma: Is the Risk Still Increased? Arthritis & rheumatology (Hoboken, N.J.) 69, 700–708, doi: 10.1002/art.40017 (2017). [DOI] [PubMed] [Google Scholar]
  • 101.Mercer LK et al. Risk of lymphoma in patients exposed to antitumour necrosis factor therapy: results from the British Society for Rheumatology Biologics Register for Rheumatoid Arthritis. Ann Rheum Dis 76, 497–503, doi: 10.1136/annrheumdis-2016-209389 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Hyams JS et al. Infliximab Is Not Associated With Increased Risk of Malignancy or Hemophagocytic Lymphohistiocytosis in Pediatric Patients With Inflammatory Bowel Disease. Gastroenterology 152, 1901–1914.e1903, doi: 10.1053/j.gastro.2017.02.004 (2017). [DOI] [PubMed] [Google Scholar]
  • 103.Raaschou P, Simard JF, Holmqvist M, Askling J & Group AS Rheumatoid arthritis, anti-tumour necrosis factor therapy, and risk of malignant melanoma: nationwide population based prospective cohort study from Sweden. BMJ 346, f1939, doi: 10.1136/bmj.f1939 (2013). [DOI] [PubMed] [Google Scholar]
  • 104.Mercer LK et al. Risk of invasive melanoma in patients with rheumatoid arthritis treated with biologics: results from a collaborative project of 11 European biologic registers. Annals of the rheumatic diseases 76, 386–391, doi: 10.1136/annrheumdis-2016-209285 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Hellgren K et al. Cancer risk in patients with spondyloarthritis treated with TNF inhibitors: a collaborative study from the ARTIS and DANBIO registers. Annals of the rheumatic diseases 76, 105–111, doi: 10.1136/annrheumdis-2016-209270 (2017). [DOI] [PubMed] [Google Scholar]
  • 106.Lopez-Olivo MA et al. Risk of malignancies in patients with rheumatoid arthritis treated with biologic therapy: a meta-analysis. JAMA 308, 898–908, doi: 10.1001/2012.jama.10857 (2012). [DOI] [PubMed] [Google Scholar]
  • 107.Peleva E et al. Risk of cancer in patients with psoriasis on biological therapies: a systematic review. The British journal of dermatology 178, 103–113, doi: 10.1111/bjd.15830 (2018). [DOI] [PubMed] [Google Scholar]
  • 108.Wang JL et al. Risk of non-melanoma skin cancer for rheumatoid arthritis patients receiving TNF antagonist: a systematic review and meta-analysis. Clinical rheumatology, doi: 10.1007/s10067-019-04865-y (2019). [DOI] [PubMed] [Google Scholar]
  • 109.Scott FI et al. Risk of Nonmelanoma Skin Cancer Associated With the Use of Immunosuppressant and Biologic Agents in Patients With a History of Autoimmune Disease and Nonmelanoma Skin Cancer. JAMA dermatology 152, 164–172, doi: 10.1001/jamadermatol.2015.3029 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Raaschou P, Söderling J, Turesson C & Askling J Tumor Necrosis Factor Inhibitors and Cancer Recurrence in Swedish Patients With Rheumatoid Arthritis: A Nationwide Population-Based Cohort Study. Annals of internal medicine 169, 291–299, doi: 10.7326/M17-2812 (2018). [DOI] [PubMed] [Google Scholar]
  • 111.Silva-Fernández L et al. The incidence of cancer in patients with rheumatoid arthritis and a prior malignancy who receive TNF inhibitors or rituximab: results from the British Society for Rheumatology Biologics Register-Rheumatoid Arthritis. Rheumatology (Oxford, England) 55, 2033–2039, doi: 10.1093/rheumatology/kew314 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Chen L & Flies DB Molecular mechanisms of T cell co-stimulation and co-inhibition. Nat Rev Immunol 13, 227–242, doi: 10.1038/nri3405 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Ribas A & Wolchok JD Cancer immunotherapy using checkpoint blockade. Science 359, 1350–1355, doi: 10.1126/science.aar4060 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Wei SC et al. Distinct Cellular Mechanisms Underlie Anti-CTLA-4 and Anti-PD-1 Checkpoint Blockade. Cell 170, 1120–1133 e1117, doi: 10.1016/j.cell.2017.07.024 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Huang AC et al. T-cell invigoration to tumour burden ratio associated with anti-PD-1 response. Nature 545, 60–65, doi: 10.1038/nature22079 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Zappasodi R et al. Non-conventional Inhibitory CD4(+)Foxp3(−)PD-1(hi) T Cells as a Biomarker of Immune Checkpoint Blockade Activity. Cancer Cell 33, 1017–1032 e1017, doi: 10.1016/j.ccell.2018.05.009 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Strauss L et al. Targeted deletion of PD-1 in myeloid cells induces antitumor immunity. Sci Immunol 5, doi: 10.1126/sciimmunol.aay1863 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Zheng L et al. Induction of apoptosis in mature T cells by tumour necrosis factor. Nature 377, 348–351, doi: 10.1038/377348a0 (1995). [DOI] [PubMed] [Google Scholar]
  • 119.Kim EY, Teh SJ, Yang J, Chow MT & Teh HS TNFR2-deficient memory CD8 T cells provide superior protection against tumor cell growth. J Immunol 183, 6051–6057, doi: 10.4049/jimmunol.0803482 (2009). [DOI] [PubMed] [Google Scholar]
  • 120.Bertrand F et al. Blocking Tumor Necrosis Factor alpha Enhances CD8 T-cell-Dependent Immunity in Experimental Melanoma. Cancer Res 75, 2619–2628, doi: 10.1158/0008-5472.CAN-14-2524 (2015). [DOI] [PubMed] [Google Scholar]
  • 121.Zheng Y et al. TNF-α-induced Tim-3 expression marks the dysfunction of infiltrating natural killer cells in human esophageal cancer. Journal of translational medicine 17, 165, doi: 10.1186/s12967-019-1917-0 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Ivagnes A et al. TNFR2/BIRC3-TRAF1 signaling pathway as a novel NK cell immune checkpoint in cancer. Oncoimmunology 7, e1386826, doi: 10.1080/2162402X.2017.1386826 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Grinberg-Bleyer Y et al. Pathogenic T cells have a paradoxical protective effect in murine autoimmune diabetes by boosting Tregs. J Clin Invest 120, 4558–4568, doi: 10.1172/JCI42945 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Zanin-Zhorov A et al. Protein kinase C-theta mediates negative feedback on regulatory T cell function. Science 328, 372–376, doi: 10.1126/science.1186068 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Zaragoza B et al. Suppressive activity of human regulatory T cells is maintained in the presence of TNF. Nat Med 22, 16–17, doi: 10.1038/nm.4019 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Bilate AM & Lafaille JJ Can TNF-alpha boost regulatory T cells? J Clin Invest 120, 4190–4192, doi: 10.1172/JCI45262 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Chen X et al. Cutting edge: expression of TNFR2 defines a maximally suppressive subset of mouse CD4+CD25+FoxP3+ T regulatory cells: applicability to tumor-infiltrating T regulatory cells. J Immunol 180, 6467–6471, doi: 10.4049/jimmunol.180.10.6467 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Govindaraj C et al. Impaired Th1 immunity in ovarian cancer patients is mediated by TNFR2+ Tregs within the tumor microenvironment. Clinical immunology (Orlando, Fla.) 149, 97–110, doi: 10.1016/j.clim.2013.07.003 (2013). [DOI] [PubMed] [Google Scholar]
  • 129.Chopra M et al. Tumor necrosis factor receptor 2-dependent homeostasis of regulatory T cells as a player in TNF-induced experimental metastasis. Carcinogenesis 34, 1296–1303, doi: 10.1093/carcin/bgt038 (2013). [DOI] [PubMed] [Google Scholar]
  • 130.Torrey H et al. Targeting TNFR2 with antagonistic antibodies inhibits proliferation of ovarian cancer cells and tumor-associated Tregs. Sci Signal 10, doi: 10.1126/scisignal.aaf8608 (2017). [DOI] [PubMed] [Google Scholar]
  • 131.Torrey H et al. Targeted killing of TNFR2-expressing tumor cells and Tregs by TNFR2 antagonistic antibodies in advanced Sezary syndrome. Leukemia 33, 1206–1218, doi: 10.1038/s41375-018-0292-9 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Chen X et al. Expression of costimulatory TNFR2 induces resistance of CD4+FoxP3- conventional T cells to suppression by CD4+FoxP3+ regulatory T cells. J Immunol 185, 174–182, doi: 10.4049/jimmunol.0903548 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Charles KA et al. The tumor-promoting actions of TNF-alpha involve TNFR1 and IL-17 in ovarian cancer in mice and humans. J Clin Invest 119, 3011–3023, doi: 10.1172/JCI39065 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Nunez S et al. T helper type 17 cells contribute to anti-tumour immunity and promote the recruitment of T helper type 1 cells to the tumour. Immunology 139, 61–71, doi: 10.1111/imm.12055 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Martin-Orozco N et al. T helper 17 cells promote cytotoxic T cell activation in tumor immunity. Immunity 31, 787–798, doi: 10.1016/j.immuni.2009.09.014 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Zhao X et al. TNF signaling drives myeloid-derived suppressor cell accumulation. J Clin Invest 122, 4094–4104, doi: 10.1172/JCI64115 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Sade-Feldman M et al. Tumor necrosis factor-alpha blocks differentiation and enhances suppressive activity of immature myeloid cells during chronic inflammation. Immunity 38, 541–554, doi: 10.1016/j.immuni.2013.02.007 (2013). [DOI] [PubMed] [Google Scholar]
  • 138.Ren G et al. CCR2-dependent recruitment of macrophages by tumor-educated mesenchymal stromal cells promotes tumor development and is mimicked by TNFα. Cell stem cell 11, 812–824, doi: 10.1016/j.stem.2012.08.013 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Lim SO et al. Deubiquitination and Stabilization of PD-L1 by CSN5. Cancer Cell 30, 925–939, doi: 10.1016/j.ccell.2016.10.010 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Bertrand F et al. TNFalpha blockade overcomes resistance to anti-PD-1 in experimental melanoma. Nat Commun 8, 2256, doi: 10.1038/s41467-017-02358-7 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Landsberg J et al. Melanomas resist T-cell therapy through inflammation-induced reversible dedifferentiation. Nature 490, 412–416, doi: 10.1038/nature11538 (2012). [DOI] [PubMed] [Google Scholar]
  • 142.Kim EY & Teh HS Critical role of TNF receptor type-2 (p75) as a costimulator for IL-2 induction and T cell survival: a functional link to CD28. J Immunol 173, 4500–4509, doi: 10.4049/jimmunol.173.7.4500 (2004). [DOI] [PubMed] [Google Scholar]
  • 143.Calzascia T et al. TNF-alpha is critical for antitumor but not antiviral T cell immunity in mice. J Clin Invest 117, 3833–3845, doi: 10.1172/JCI32567 (2007). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Maney NJ, Reynolds G, Krippner-Heidenreich A & Hilkens CMU Dendritic cell maturation and survival are differentially regulated by TNFR1 and TNFR2. J Immunol 193, 4914–4923, doi: 10.4049/jimmunol.1302929 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Perez-Ruiz E et al. Prophylactic TNF blockade uncouples efficacy and toxicity in dual CTLA-4 and PD-1 immunotherapy. Nature 569, 428–432, doi: 10.1038/s41586-019-1162-y (2019). [DOI] [PubMed] [Google Scholar]
  • 146.Ham B, Fernandez MC, D’Costa Z & Brodt P The diverse roles of the TNF axis in cancer progression and metastasis. Trends Cancer Res 11, 1–27 (2016). [PMC free article] [PubMed] [Google Scholar]
  • 147.Castro F, Cardoso AP, Goncalves RM, Serre K & Oliveira MJ Interferon-Gamma at the Crossroads of Tumor Immune Surveillance or Evasion. Front Immunol 9, 847, doi: 10.3389/fimmu.2018.00847 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Koch J, Steinle A, Watzl C & Mandelboim O Activating natural cytotoxicity receptors of natural killer cells in cancer and infection. Trends Immunol 34, 182–191, doi: 10.1016/j.it.2013.01.003 (2013). [DOI] [PubMed] [Google Scholar]
  • 149.Marzo AL et al. Tumor-specific CD4+ T cells have a major “post-licensing” role in CTL mediated anti-tumor immunity. J Immunol 165, 6047–6055, doi: 10.4049/jimmunol.165.11.6047 (2000). [DOI] [PubMed] [Google Scholar]
  • 150.Dunn GP, Old LJ & Schreiber RD The three Es of cancer immunoediting. Annu Rev Immunol 22, 329–360, doi: 10.1146/annurev.immunol.22.012703.104803 (2004). [DOI] [PubMed] [Google Scholar]
  • 151.Dobrzanski MJ Expanding roles for CD4 T cells and their subpopulations in tumor immunity and therapy. Front Oncol 3, 63, doi: 10.3389/fonc.2013.00063 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Briscoe DM, Cotran RS & Pober JS Effects of tumor necrosis factor, lipopolysaccharide, and IL-4 on the expression of vascular cell adhesion molecule-1 in vivo. Correlation with CD3+ T cell infiltration. J Immunol 149, 2954–2960 (1992). [PubMed] [Google Scholar]
  • 153.Li MO & Flavell RA TGF-beta: a master of all T cell trades. Cell 134, 392–404, doi: 10.1016/j.cell.2008.07.025 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Mempel TR et al. Regulatory T cells reversibly suppress cytotoxic T cell function independent of effector differentiation. Immunity 25, 129–141, doi: 10.1016/j.immuni.2006.04.015 (2006). [DOI] [PubMed] [Google Scholar]
  • 155.Nagaraj S et al. Altered recognition of antigen is a mechanism of CD8+ T cell tolerance in cancer. Nat Med 13, 828–835, doi: 10.1038/nm1609 (2007). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Chanmee T, Ontong P, Konno K & Itano N Tumor-associated macrophages as major players in the tumor microenvironment. Cancers (Basel) 6, 1670–1690, doi: 10.3390/cancers6031670 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES