Next Article in Journal
The APSANTICO Study: A Prospective Observational Study to Evaluate Antiphospholipid Antibody Profiles in Patients with Thromboembolic Antiphospholipid Syndrome (APS) after COVID-19 Infection and/or Vaccination
Next Article in Special Issue
Inappropriate Expression of PD-1 and CTLA-4 Checkpoints in Myeloma Patients Is More Pronounced at Diagnosis: Implications for Time to Progression and Response to Therapeutic Checkpoint Inhibitors
Previous Article in Journal
Role of Low-Risk HPV PCR Monoinfection in Screening for HSIL and Anal Cancer in Men Who Have Sex with Men Living with HIV
Previous Article in Special Issue
A New Signature That Predicts Progression-Free Survival of Clear Cell Renal Cell Carcinoma with Anti-PD-1 Therapy
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Rheumatic Immune-Related Adverse Events due to Immune Checkpoint Inhibitors—A 2023 Update

1
Department of Clinical Immunology, Osaka Metropolitan University Graduate School of Medicine, Osaka 565-0871, Japan
2
Preemptive Medicine and Lifestyle Related Disease Research Center, Kyoto University Hospital, Kyoto 606-8397, Japan
3
Department of Orthopaedic Surgery, Osaka Metropolitan University Graduate School of Medicine, Osaka 565-0871, Japan
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Int. J. Mol. Sci. 2023, 24(6), 5643; https://doi.org/10.3390/ijms24065643
Submission received: 6 February 2023 / Revised: 5 March 2023 / Accepted: 14 March 2023 / Published: 15 March 2023
(This article belongs to the Special Issue Immune Checkpoint Inhibitors and Immune Checkpoint Resistance)

Abstract

:
With the aging of the population, malignancies are becoming common complications in patients with rheumatoid arthritis (RA), particularly in elderly patients. Such malignancies often interfere with RA treatment. Among several therapeutic agents, immune checkpoint inhibitors (ICIs) which antagonize immunological brakes on T lymphocytes have emerged as a promising treatment option for a variety of malignancies. In parallel, evidence has accumulated that ICIs are associated with numerous immune-related adverse events (irAEs), such as hypophysitis, myocarditis, pneumonitis, and colitis. Moreover, ICIs not only exacerbate pre-existing autoimmune diseases, but also cause de novo rheumatic disease–like symptoms, such as arthritis, myositis, and vasculitis, which are currently termed rheumatic irAEs. Rheumatic irAEs differ from classical rheumatic diseases in multiple aspects, and treatment should be individualized based on the severity. Close collaboration with oncologists is critical for preventing irreversible organ damage. This review summarizes the current evidence regarding the mechanisms and management of rheumatic irAEs with focus on arthritis, myositis, and vasculitis. Based on these findings, potential therapeutic strategies against rheumatic irAEs are discussed.

1. Introduction

Rheumatoid arthritis (RA) is an autoimmune disorder associated with progressive joint destruction and reduced physical function [1]. Over the past few decades, treatment options, such as biological agents and molecularly targeted antirheumatic drugs, have dramatically expanded, making it a feasible therapeutic goal for many patients to achieve clinical remission [2]. RA used to be common in women in their 40s–50s; however, as the population ages, the number of patients with elderly onset RA (EORA), which occurs after 60 years of age, has been increasing [3]. Compared to young-onset RA (YORA), EORA patients are more likely to be male and seronegative for rheumatoid factor (RF) and anti-cyclic citrullinated protein (CCP) antibodies, and have markedly elevated C-reactive protein (CRP) levels and erythrocyte sedimentation rates, suggesting that EORA and YORA have a different pathophysiology [4,5,6]. In addition, patients with RA are at higher risk of developing malignancies, such as lymphomas, melanomas, and lung cancers, than healthy individuals [7,8]. Elderly patients with RA in particular are at high risk and often have a past or concurrent medical history of malignancy [9].
In recent years, immune checkpoint inhibitors (ICIs), such as anti-cytotoxic T-lymphocyte-associated protein 4 (CTLA-4), anti-programmed cell death protein-1 (PD-1), and anti-programmed cell death ligand-1 (PD-L1) antibodies, have emerged as new treatment options for malignancies [10,11,12]. These cancer immunotherapies activate T cells by inhibiting negative signals on T lymphocytes, thereby exerting an anti-tumor effect [13]. Initially tested for the treatment of malignant melanoma [14], these agents have been increasingly approved for a variety of cancer types, including renal, gastric, esophageal, colorectal, lung, and breast cancers [15]. Moreover, there are many ongoing clinical trials using checkpoint inhibitors that target novel costimulatory or coinhibitory molecules [16].
In parallel, there is increasing evidence that ICIs sometimes cause side effects called immune-related adverse events (irAEs), including hypophysitis, thyroiditis, myocarditis, and others [17]. Thus, irAEs cause diverse pathologies in nearly every organ [18,19]. Among them, conditions that resemble rheumatic diseases, such as arthritis, myositis, sicca symptoms, and vasculitis, are now termed rheumatic irAEs and are often treated with similar therapeutic agents as those for rheumatic diseases. However, rheumatic irAEs differ from idiopathic rheumatic diseases in multiple aspects, necessitating the elucidation of their pathophysiology.
This review summarizes the current evidence on the underlying mechanisms of irAEs. Then, the recent topics of rheumatic irAEs, particularly arthritis, myositis, and vasculitis, are updated. Finally, potential therapeutic strategies for rheumatic irAEs are discussed.

2. Immune-Related Adverse Events (irAEs)

2.1. Mechanisms of Action of Immune Checkpoint Inhibitors (ICIs)

Monoclonal antibodies targeting immunological checkpoints, currently termed ICIs, represent a growing class of agents effective against multiple tumor types and all disease stages [20]. Immune checkpoints are receptors expressed by immune cells that are dynamically involved in immune homeostasis and self-tolerance and particularly relevant to T cell function [21]. The development of ICIs has introduced a new era in cancer treatment, enabling the possible long-term survival of patients with metastatic disease and providing new therapeutic options in patients at an early stage.
ICIs target different sets of regulatory interactions, the most common of which is the CTLA-4, PD-1, and PD-L1 blockade [15]. Upon T cell activation, multiple mechanisms initiate to control the level of activation. The two main regulatory interactions that control T cell activation are CTLA-4 (also called CD152) binding to CD80/86 (also called B7-1 and B7-2) on antigen-presenting cells and PD-1 binding to PD-L1 (also known as B7-H1) or programmed cell death ligand 2 (also known as B7-H2) [22]. Compared to PD-1, CTLA-4 has a more proximal role, acting as a higher-affinity competitor to CD28, thus limiting T cell activation at the priming stage [22]. These regulatory systems are targets for cancer immunotherapy, and the blockade of these immunoinhibitory receptors unleashes activated T cells and exerts anti-tumor effects. Combining anti-CTLA-4 antibodies with anti-PD-1 or anti-PD-L1 antibodies further enhances the anti-tumor effect [23].
However, ICIs are effective in only a small proportion of patients. Multiple attempts are ongoing to enhance their anti-tumor effects [24,25] or determine their efficacy at an early stage with biomarkers using peripheral blood, tissue samples, and imaging modalities [26,27,28,29,30,31]. In 2018, Matson et al. reported that the patients’ gut microbiome was associated with their response to ICIs [32]. However, the results differ among cohorts and their reproducibility has not been demonstrated [33].

2.2. Mechanisms of irAEs

In parallel with their success against malignancies, ICIs induce potentially severe and even lethal side effects called irAEs, which are considered off-target tissue damage and involve essentially every organ system [17]. A wide range of irAEs have been described [34,35], including hypophysitis, thyroiditis, myocarditis, pneumonia, pancreatitis, hepatitis, nephritis, adrenal insufficiency, enteritis, and skin rash (Figure 1). Although the underlying mechanisms of irAEs are not fully understood, (1) aberrant cytotoxic T cell activation, (2) increased autoantibody production, (3) direct molecular mimicry, (4) inflammatory cytokine production, (5) complement-mediated inflammation, (6) imparted regulatory T cell function, and other mechanisms contribute to the immunopathology of irAEs [17,19,36,37,38].
These irAEs can be provoked by both the CTLA-4 and PD-1 blockades, but their frequencies vary. For instance, colitis, hypophysitis, and rash occur more frequently with CTLA-4 inhibitors, whereas pneumonitis, hypothyroidism, and vitiligo are more common with PD-1/PD-L1 inhibitors [39,40,41]. Thus, the irAE profiles are distinct between the two groups. The molecular basis requires elucidation; however, recent studies demonstrated that anti-CTLA-4 and anti-PD-1 have different effects on T cell subsets [42]. CTLA-4 blockade primarily induces CD4+ T cell activation, particularly Th1 cells and follicular helper T cells, while counteracting regulatory T cell function [43,44,45,46]. PD-1 blockade mainly reinvigorates CD8+ T cells, including exhausted and cytotoxic T cells, while expanding CD4+ regulatory T cells [47,48,49]. These molecular differences may account for the clinical differences between the two.
Recent technical advances have provided interesting insights into irAEs. For example, myocarditis due to ICIs has long been reported [50]; however, using multiomics single-cell technology, Zhu et al. found an expansion of cytotoxic CD8+ T effector cells re-expressing CD45RA in patients with ICI-induced myocarditis [51]. These cells exhibit high levels of activation and cytotoxicity as well as chemokine receptor expression, suggesting that these cells have a pathogenic role and could be a therapeutic target in ICI-induced myocarditis [51]. In addition, genome-wide association studies have identified germline variants of IL7 associated with all-grade irAEs [52,53]. Patients who carried the IL7 germline variant showed increased lymphocyte stability after ICI initiation, high risk of all-grade irAEs, and improved survival [52,53]. As interleukin (IL)-7 is a critical regulator of T cell homeostasis [54,55], these results suggest that T lymphocytes are critically involved in the efficacy and safety of ICIs.

3. Rheumatic irAEs

3.1. Autoimmune-Disease-like Symptoms Due to ICIs

Since immunological checkpoints are critical in immune homeostasis and self-tolerance, the blockade of these molecules leads to immune system overactivation and the development of autoimmune-disease-like symptoms [56]. These are now called rheumatic irAEs and have been described in a growing number of case series and reports. The abrogation of immune checkpoints, such as CTLA-4 or PD-1, has similar consequences in animal models. The deletion of CTLA4 in mice results in premature death due to lymphocyte proliferation and multiorgan failure [57,58]. In contrast, the deletion of Pd1 or Pdl1 results in less severe conditions but shows spontaneous arthritis and lupus-like glomerulonephritis [59,60]. Similar to irAEs, rheumatic irAEs are associated with diverse pathologies. The most common clinical manifestation is arthritis, followed by myositis, myalgia, vasculitis, Sjögren-syndrome-like sicca symptoms, a psoriasis-like rash, systemic-sclerosis-like skin sclerosis, and lupus-like nephritis (Figure 2).
Rheumatic irAEs resemble classical rheumatological diseases, suggesting a possible overlapping mechanism. Thus, elucidation of the disease mechanisms underlying rheumatic irAEs may lead to novel therapeutic approaches for rheumatic diseases [61]. Although the precise mechanism remains unclear, ICIs block negative signals on activated T cells, leading to increased autoreactive T cells and inflammatory cytokines such as tumor necrosis factor-α (TNF-α), IL-6, and IL-17 [62,63,64]. This provides the rationale for targeted inhibition of these cytokines to treat rheumatic irAEs [65,66]. ICIs can also induce epitope spreading, diversifying the epitope specificity of T cells [62]. Moreover, ICIs prevent the apoptosis of B cells, allowing for longer cell survival and increased autoantibody loads [62].
Rheumatic irAEs are often treated with therapeutic agents used for classical rheumatic diseases, such as glucocorticoids, conventional synthetic disease-modifying antirheumatic drugs (DMARDs), and biological DMARDs (bDMARDs) [67,68,69,70,71,72] (Figure 3). Among them, abatacept, a fusion protein of the extracellular domain of CTLA-4 and the Fc portion of immunoglobulin G, blocks the T-cell-activating interaction between CD28 and CD80/86 and is an effective bDMARD for RA [73,74]. Its mechanism is the conversion of the anti-CTLA-4 antibody, which blocks the T-cell-inhibitory interaction between CTLA-4 and CD80/86 [75,76]. For these reasons, abatacept should be avoided for the treatment of irAEs [77]; however, in cases of life-threatening conditions such as myocarditis, its use can be considered [78,79,80]. Hereafter, rheumatic irAEs will be discussed in each section. In particular, we will focus on arthritis, myositis, and vasculitis.

3.2. Arthritis

In recent years, reports of inflammatory arthritis after ICI administration have been increasing [81,82,83,84,85,86]. Arthritis is more common with PD-1/PD-L1 blockade than with CTLA-4 blockade [39], but the combination carries the highest risk [87]. In a retrospective cohort study, the prevalence of ICI-induced arthritis was estimated as 2% among patients undergoing cancer immunotherapy [87]. In this study, the mean age of 34 patients with ICI-induced arthritis was 59.2 years; 65% of them had the polyarticular type and 35% had the oligoarticular type, and the mean CRP level was 5.14 mg/dL. These characteristics were not distinguishable from RA. However, notably, RF and CCP antibodies were negative in all patients, suggesting that the pathophysiology differs between ICI-induced arthritis and RA and that autoantibodies are not directly involved in ICI-induced arthritis [87].
In contrast, according to a study by Cappelli, anti-RA33 antibodies were detected in 11.4% of patients with ICI-induced arthritis, whereas none of the ICI-treated controls without arthritis tested positive [88]. The antibodies were also present in 7.7% of patients with RA and 2% of healthy controls, suggesting that autoantibodies may play a role in ICI-induced arthritis or that individuals with pre-existing autoantibodies are at risk of ICI-induced arthritis.
According to the European Alliance of Associations for Rheumatology (EULAR), mild to moderate arthritis can be treated with nonsteroidal anti-inflammatory drugs, analgesics, and/or intra-articular glucocorticoids. If inflammation is evident despite these medications, systemic glucocorticoids should be considered [77]. Some may require high doses of glucocorticoids. However, we must consider that glucocorticoids may negatively impact the efficacy of cancer immunotherapy [89,90]. In animal models, both endogenous and exogenous glucocorticoids abolished anti-tumor immune responses [91]. Therefore, in patients with mild joint involvement, if feasible, systemic glucocorticoid administration should be avoided [90]. However, in the abovementioned retrospective study, 62% of patients were treated with systemic glucocorticoids [87], suggesting their high efficacy.
Conventional synthetic DMARDs, such as methotrexate, hydroxychloroquine, and sulfasalazine, and bDMARDs, such as TNF-α and IL-6 inhibitors, may be necessary in patients with moderate to severe arthritis that is refractory to glucocorticoid therapy [77]. Consultation with oncologists is necessary to decide whether to continue or stop ICI therapy and use bDMARDs. As mentioned above, the use of abatacept should be avoided because of the risk of antagonizing the anti-tumor effects of ICIs [77]. There is no evidence in the literature to suggest a preference for one biologic DMARD over another: infliximab, adalimumab, etanercept, tocilizumab, and sarilumab have controlled symptoms [56,66,92].
Some studies have suggested that this order of treatment should be reversed [56]. In other words, bDMARDs should be considered initially. The reasons for this are as follows: (1) ICI-induced arthritis also progresses to joint destruction from an early stage [93]; and (2) bDMARDs may not interfere with the anti-tumor response of ICI [82,94,95]. For instance, an in vitro study compared the effects of dexamethasone and infliximab on the anti-tumor activity of tumor-infiltrating lymphocytes. Even low doses of dexamethasone significantly attenuated anti-tumor activity, whereas a standard dose of infliximab had only minor effects on tumor killing. The activity of tumor-infiltrating lymphocytes was restored after the discontinuation of dexamethasone [96]. Therefore, bDMARDs have been administered in an increasing number of cases of ICI-induced arthritis [97].
A recent molecular study demonstrated that CD8+ T cells from ICI-treated patients who developed arthritis had distinct effector functions and metabolic profiles from those from ICI-treated patients who remained arthritis-free and that TNF-α inhibitors or Janus kinase (JAK) inhibitors were unable to adequately correct such dysfunction in vitro [98]. This suggests that bDMARDs and JAK inhibitors may effectively reduce symptoms but may not fundamentally restore T cell function, although because of the risk of malignancy [99], JAK inhibitors are normally avoided in ICI-induced arthritis.
We have so far discussed newly onset arthritis or “de novo” arthritis after ICI initiation, but what happens when ICIs are given in patients with pre-existing arthritis? Most of the clinical trials have excluded patients with pre-existing autoimmune disease because of the concern of disease flare; however, real world evidence is accumulating that ICIs can be administered to such patients with an acceptable safety profile, but the likelihood of disease flares is high [87,100,101]. For instance, the relapse rates for RA and psoriatic arthritis have been reported to be greater than 50%, while those for systemic lupus erythematosus, systemic sclerosis, myositis, and vasculitis range from 20 to 50%, indicating that disease flare varies depending on the type of pre-existing autoimmune disease. Even when RA flares, symptoms are relatively mild and ICIs can often be resumed [100].
More recent evidence has shown that patients with pre-existing autoimmune disease are at greater risk of all-grade, severe, and multiple irAEs, but have a better survival than controls [102]. These results indicate that having an autoimmune disease is not a contraindication to ICI and that T cells from patients with autoimmune disease may be more susceptible to ICI treatment than those from patients without autoimmune disease.

3.3. Myositis

Compared to arthritis, ICI-induced myositis is rare, with an estimated incidence of less than 1% but a high mortality rate of up to 22% [87,103,104,105]. The meta-analysis demonstrated no significant difference in the incidence of anti-PD-1/PD-L1 and anti-CTLA-4 antibodies (odds ratio, 1.07; 95% confidence interval, 0.27–9.41) [106]; however, anti-CTLA-4 antibodies have been reported less frequently than anti-PD-1/PD-L1 antibody treatment. The average age at onset of myositis is approximately 70 years [87,106,107], and the mean time from ICI initiation to myositis onset is approximately 4 weeks [106,107]. ICI-induced arthritis often occurs several months after the initiation of ICI [29], whereas myositis often develops at a relatively early treatment phase.
Classical inflammatory myositis includes dermatomyositis (DM) and polymyositis (PM), and ICI-induced myositis is indistinguishable from these in many aspects. Creatine kinase (CK) levels often increase to >1000 units/L, electromyography shows myogenic changes, and high-intensity signals can be detected in affected lesions on T2-weighted magnetic resonance imaging [87,108]. A skin rash characteristic of DM is found in 20% of patients [106]. However, ICI-induced myositis includes clinical features that are distinct from those of idiopathic inflammatory myositis. Unlike PM/DM, ICI-induced myositis often presents with ptosis and ocular motility disorders as initial symptoms [106]. Interstitial lung disease is detected in only a small proportion of patients, approximately 2%, while up to 40% of patients develop myocarditis. Respiratory muscle involvement is also common in ICI-induced myositis. Ptosis and respiratory muscle involvement are predictive of myocarditis development [106].
Most cases of ICI-induced myositis test negative for myositis-specific/-associated antibodies [106], whereas some patients show multiple myositis-specific/-associated antibodies [109]. Moreover, anti-striated muscle antibodies have often been detected in ICI-induced myositis. Anti-striated muscle antibodies include anti-titin antibodies and anti-Kv1.4 antibodies, and are often found in patients with thymoma-associated myasthenia gravis [110], suggesting an overlap in the pathophysiology between ICI-induced myositis and myasthenia gravis [87]. Thus, a fatal triad of myasthenia gravis, myositis, and myocarditis can occur in patients with ICI-induced myositis [111]. However, marked improvement in the edrophonium test, which is observed in myasthenia gravis, is rare in ICI-induced myositis [107].
Muscle biopsy shows myofiber necrosis with regenerative findings and infiltration of CD4+ T cells, CD8+ T cells, CD20+ B cells, and CD68+ macrophages. High expression of major histocompatibility complex class I is also observed. These findings are slightly different from those in anti-signal recognition particles and anti-aminoacyl-tRNA synthetase antibody-positive myositis [107]. Human leucocyte antigen (HLA) class I haplotypes (HLA-A*24:02/B*52:01/C*12:02) can be frequently detected in ICI-induced myositis [107].
According to the clinical practice guidelines for the management of irAEs [112], mild to moderate myositis can be treated with prednisolone from 10 mg to 1 mg/kg, depending on severity. In severe cases, it is recommended to start with prednisolone 1 mg/kg and consider methylprednisolone 1–2 mg/kg or a high-dose bolus. Additionally, plasma exchange therapy and/or intravenous immunoglobulin therapy should be considered [77]. If symptoms and CK levels do not improve or worsen despite treatment, other immunosuppressive agents, such as methotrexate, azathioprine, or mycophenolate mofetil, should be considered [112]. Resumption of ICIs is usually avoided when myositis, particularly in association with myocarditis, develops, although there are some reports with successful resumption of ICIs in patients with mild disease [107].

3.4. Vasculitis

Cancer immunotherapy is a potential trigger for vasculitis [72,113,114,115]. Vasculitis is classified into large-, medium-, and small-vessel types depending on blood vessel size [116], but ICIs can cause any type of vasculitis. In fact, giant cell arteritis (GCA) [117,118], aortitis and large-vessel involvement [119,120,121], IgA vasculitis [122,123], granulomatosis with polyangiitis [124,125], eosinophilic granulomatosis with polyangiitis [126], and cutaneous small-vessel vasculitis [127,128] have been reported after ICI administration. A retrospective pharmacovigilance study evaluated the association between ICIs and cardiovascular adverse events and demonstrated that ICI treatment was associated with a higher risk of vasculitis when compared with the database obtained from individual case safety reports [129]. This signal was primarily driven by GCA, with a reporting odds ratio of 12.99. Anti-PD-1/PD-L1 antibodies have a relatively higher risk of developing vasculitis than anti-CTLA-4 antibodies, but the combination of these have the highest risk [129].
Although the mechanisms underlying the development of GCA by ICIs are unclear, we have previously demonstrated that blockade of PD-1/PD-L1 signaling exacerbated vascular inflammation using a preclinical mouse model of large-vessel vasculitis [13,130]. In this mouse model, human medium- and large-sized arteries are embedded into NOD scid immunodeficient mouse, and then peripheral blood mononuclear cells from patients with GCA are transferred into this mouse, causing vasculitis by alloreaction [131]. The administration of anti-PD-1 antibody into this mouse not only increased the infiltration of activated T cells in the vasculature and inflammatory cytokines, such as IL-6, IL-17, TNF-α, and interferon gamma (IFN-γ), but also promoted hyperplasia in the intima and neovessel formation in the adventitia, suggesting that the PD-1/PD-L1 signal is critically involved in vascular remodeling [130].
These experiments prompted us to examine the expression of PD-L1 on antigen-presenting cells in the vasculature. We found that PD-L1 expression was markedly diminished in dendritic cells (DCs) within the adventitia. These results suggest that deficient PD-L1 expression on vascular DCs was unable to sufficiently inhibit T cell overactivation in vascular lesions and that ICI administration mimics GCA immunopathology [132]. Reduced expression of PD-L1 in vascular DCs has been confirmed in a mouse model of lipopolysaccharide-induced vasculitis [133]. Moreover, aberrant PD-1/PD-L1 signal expression has also been reported in anti-neutrophil cytoplasmic-antibody-associated vasculitis [134,135]. Although a variety of drugs reportedly cause drug-induced vasculitis, such as hydralazine, minocycline, propylthiouracil, granulocyte colony-stimulating factor, and anti-TNF-α inhibitors [136,137], it is important to recognize that ICIs also can trigger any type of vasculitis.
Vasculitis caused by ICIs usually requires high-dose glucocorticoid treatment [77]. Other therapeutic options include cyclophosphamide [138], tocilizumab [119], rituximab [139,140], plasmapheresis [141], and JAK inhibitors [142,143]; however, the optimal management of ICI-induced vasculitis has not been established.

3.5. Myalgia (Polymyalgia Rheumatica)

Polymyalgia rheumatica (PMR) is the most common inflammatory rheumatic disease affecting individuals >50 years of age [144]. PMR is characterized by pain in the shoulder and pelvic girdles and often associated with GCA [145]. Many patients respond quickly to low-dose glucocorticoids; however, multiple relapses are common with glucocorticoid tapering [146,147].
PMR recently emerged as a side effect of cancer immunotherapy [148,149,150,151,152,153,154]. Compared with anti-CTLA-4 antibodies, anti-PD-1/PD-L1 antibodies have a higher risk of inducing this manifestation [103]. Ultrasound and 18F-fluorodeoxyglucose-positron emission tomography (FDG-PET) findings are comparable to those of idiopathic PMR [154]. Most patients fulfill the classification criteria for PMR [149,155] and respond well to low-dose glucocorticoids [149]. If patients respond inadequately to glucocorticoids, conventional synthetic DMARDs, such as methotrexate, hydroxychloroquine, and sulfasalazine, and bDMARDs, such as tocilizumab, may be effective [69,149,156].

3.6. Sicca Symptoms (Sjögren Syndrome)

Salivary gland dysfunction is a hallmark of Sjögren syndrome [157]. ICI-induced sicca syndrome (ICIS), characterized by severe sudden-onset dry mouth [158], has been increasingly reported [82,159,160,161]. Patients with anti-PD-1/PD-L1 antibodies are at a relatively higher risk of developing this symptom than those with anti-CTLA-4 antibody [103]. Pathologically, the presence of focal lymphocytic sialadenitis is a shared feature of Sjögren syndrome and ICIS, but the nature of immune cell infiltration is T-cell-dominant in ICIS but B-cell-dominant in Sjögren syndrome [162]. Pringle et al. proposed that ICIS is inflammation-driven by IFN-γ and represents a new type II interferonopathy [162]. In many cases, low-dose glucocorticoids improve subjective symptoms but salivary gland function does not recover [161].

3.7. Skin Rash (Psoriasis)

Psoriasis-like skin rash can be observed as a rheumatic irAE after ICI administration [163,164,165,166,167]. A retrospective study showed that among 8683 ICI-treated patients, 262 (3%) developed psoriasis-like cutaneous irAEs [163]. Of these, 5% of patients with ICI-induced psoriasis showed inverse psoriasis, which required differentiation from fungal infection [163]. Flares of pre-existing psoriasis and psoriatic arthritis are also frequent, with some reports of more than 50% [101,168,169].

3.8. Skin Sclerosis (Systemic Sclerosis)

Systemic-sclerosis-like skin sclerosis or skin thickening after ICI use was initially thought to be an extremely rare rheumatic irAE [170], but a recent review suggests that it may be more common than ever thought [171]. Anti-PD-1 antibodies are at higher risk of skin sclerosis than anti-CTLA-4 antibodies, and renal crisis can be observed at the onset [171].

3.9. Lupus-like Disease

Other rheumatic irAEs include lupus-like disease [172,173,174]. Not only skin lesions but also nephritis can be observed [174,175]. The types of rheumatic irAEs may increase as ICI usage increases.

4. A Potential Strategy to Identify Patients at Risk of Rheumatic irAEs

How can we identify patients at risk of developing rheumatic irAEs? It has been reported that ANA and autoantibodies are prevalent in patients with rheumatic irAEs [176]; however, whether ANA and autoantibodies should be examined in all patients before starting ICIs is debatable. According to the EULAR points to consider for the diagnosis and management of rheumatic irAEs, there is no indication to test all patients for the presence of autoantibodies [77]. In order to identify patients at risk of rheumatic irAEs, a comprehensive framework for analyzing HLA, ANA, autoantibodies, cytokines and complement levels, and cytotoxic and regulatory T cell function may be required. Although ANA immunofluorescence pattern helps identify autoimmune liver disease [177,178], it remains unclear whether ANA patterns are predictive of rheumatic irAEs.

5. Optimal Management of Rheumatic irAEs

As we have seen so far, ICIs cause a variety of rheumatic irAEs that may further increase (Table 1). A multidisciplinary discussion between rheumatologists, oncologists, and patients is of utmost importance for the optimal management of rheumatic irAEs [77]. The continuation or discontinuation of ICIs should be guided by their effectiveness, the severity of irAEs, and the intensity of immunosuppressive therapy. In addition, therapeutic agents not only need to control symptoms, but also maintain the anti-tumor effects of ICIs [77]. Despite growing interest, prospective trials have not yet been conducted, and evidence is still lacking regarding optimal management that also permits the anti-tumor effects of ICIs. In animal models, the prophylactic administration of TNF-α inhibitors or IL-6 inhibitors maintained ICI efficacy with minor toxicity [179,180]. However, these therapeutic strategies have not been proven in humans.

6. Conclusions

A multidisciplinary and interdisciplinary approach is critical to facilitate the early diagnosis and treatment of rheumatic irAEs to prevent permanent organ damage. Further studies are required to determine their optimal management.

Author Contributions

Q.M.D., R.W. and M.S. drafted the manuscript. K.F., T.W.N., T.O. and S.Y. read and revised the manuscript. M.H. read and finalized the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This work was in part supported by JSPS KAKENHI Grant Number 22K08569 and a Grant for Promoting Research and Survey in Rheumatic Diseases by Japan Rheumatism Foundation to RW.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

We thank BioRender.com (https://biorender.com) (accessed on 1 July 2022) and irasutoya (https://www.irasutoya.com/) (accessed on 1 July 2022) for the schematic figure and Editage for the language review (https://www.editage.jp/) (accessed on 1 July 2022).

Conflicts of Interest

R.W. received research grant from AbbVie and a speaker’s fee from Asahi Kasei, Chugai, Eli Lilly, GSK, and Sanofi. T.O. received research grant and/or speaker’s fee from Abbvie, Asahi Kasei, Chugai, Eisai, Eli Lilly, Janssen, Novartis Pharma, and Tanabe Mitsubishi. M.H. received research grants and/or speaker fee from Abbvie, Asahi Kasei, Astellas, Ayumi, Brystol Meyers, Chugai, EA Pharma, Eisai, Daiichi Sankyo, Eli Lilly, Nihon Shinyaku, Novartis Pharma, and Tanabe Mitsubishi. Other authors declare no conflict of interest. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript; or in the decision to publish the results.

References

  1. Smolen, J.S.; Aletaha, D.; McInnes, I.B. Rheumatoid arthritis. Lancet 2016, 388, 2023–2038. [Google Scholar] [CrossRef] [PubMed]
  2. Smolen, J.S.; Landewé, R.B.M.; Bergstra, S.A.; Kerschbaumer, A.; Sepriano, A.; Aletaha, D.; Caporali, R.; Edwards, C.J.; Hyrich, K.L.; Pope, J.; et al. EULAR recommendations for the management of rheumatoid arthritis with synthetic and biological disease-modifying antirheumatic drugs: 2022 update. Ann. Rheum. Dis. 2023, 82, 3–18. [Google Scholar] [CrossRef] [PubMed]
  3. Serhal, L.; Lwin, M.N.; Holroyd, C.; Edwards, C.J. Rheumatoid arthritis in the elderly: Characteristics and treatment considerations. Autoimmun. Rev. 2020, 19, 102528. [Google Scholar] [CrossRef] [PubMed]
  4. Ke, Y.; Dai, X.; Xu, D.; Liang, J.; Yu, Y.; Cao, H.; Chen, W.; Lin, J. Features and Outcomes of Elderly Rheumatoid Arthritis: Does the Age of Onset Matter? A Comparative Study From a Single Center in China. Rheumatol. Ther. 2021, 8, 243–254. [Google Scholar] [CrossRef]
  5. Murata, K.; Ito, H.; Hashimoto, M.; Nishitani, K.; Murakami, K.; Tanaka, M.; Yamamoto, W.; Mimori, T.; Matsuda, S. Elderly onset of early rheumatoid arthritis is a risk factor for bone erosions, refractory to treatment: KURAMA cohort. Int. J. Rheum. Dis. 2019, 22, 1084–1093. [Google Scholar] [CrossRef]
  6. Targońska-Stępniak, B.; Grzechnik, K.; Kolarz, K.; Gągoł, D.; Majdan, M. Systemic Inflammatory Parameters in Patients with Elderly-Onset Rheumatoid Arthritis (EORA) and Young-Onset Rheumatoid Arthritis (YORA)—An Observational Study. J. Clin. Med. 2021, 10, 1204. [Google Scholar] [CrossRef]
  7. Simon, T.A.; Thompson, A.; Gandhi, K.K.; Hochberg, M.C.; Suissa, S. Incidence of malignancy in adult patients with rheumatoid arthritis: A meta-analysis. Arthritis Res. Ther. 2015, 17, 212. [Google Scholar] [CrossRef] [Green Version]
  8. Zhang, Y.; Lin, J.; You, Z.; Tu, H.; He, P.; Li, J.; Gao, R.; Liu, Z.; Xi, Z.; Li, Z.; et al. Cancer risks in rheumatoid arthritis patients who received immunosuppressive therapies: Will immunosuppressants work? Front. Immunol. 2022, 13, 1050876. [Google Scholar] [CrossRef]
  9. Huss, V.; Bower, H.; Wadstrom, H.; Frisell, T.; Askling, J.; The ARTIS Group. Short- and longer-term cancer risks with biologic and targeted synthetic disease-modifying antirheumatic drugs as used against rheumatoid arthritis in clinical practice. Rheumatology 2022, 61, 1810–1818. [Google Scholar] [CrossRef]
  10. Hoos, A. Development of immuno-oncology drugs—From CTLA4 to PD1 to the next generations. Nat. Rev. Drug Discov. 2016, 15, 235–247. [Google Scholar] [CrossRef]
  11. Leach, D.R.; Krummel, M.F.; Allison, J.P. Enhancement of antitumor immunity by CTLA-4 blockade. Science 1996, 271, 1734–1736. [Google Scholar] [CrossRef] [Green Version]
  12. Pardoll, D.M. The blockade of immune checkpoints in cancer immunotherapy. Nat. Rev. Cancer 2012, 12, 252–264. [Google Scholar] [CrossRef] [Green Version]
  13. Watanabe, R.; Zhang, H.; Berry, G.; Goronzy, J.J.; Weyand, C.M. Immune checkpoint dysfunction in large and medium vessel vasculitis. Am. J. Physiol. Heart Circ. Physiol. 2017, 312, H1052–H1059. [Google Scholar] [CrossRef] [Green Version]
  14. Hodi, F.S.; O’Day, S.J.; McDermott, D.F.; Weber, R.W.; Sosman, J.A.; Haanen, J.B.; Gonzalez, R.; Robert, C.; Schadendorf, D.; Hassel, J.C.; et al. Improved survival with ipilimumab in patients with metastatic melanoma. N. Engl. J. Med. 2010, 363, 711–723. [Google Scholar] [CrossRef]
  15. Korman, A.J.; Garrett-Thomson, S.C.; Lonberg, N. The foundations of immune checkpoint blockade and the ipilimumab approval decennial. Nat. Rev. Drug Discov. 2022, 21, 509–528. [Google Scholar] [CrossRef]
  16. Kraehenbuehl, L.; Weng, C.-H.; Eghbali, S.; Wolchok, J.D.; Merghoub, T. Enhancing immunotherapy in cancer by targeting emerging immunomodulatory pathways. Nat. Rev. Clin. Oncol. 2022, 19, 37–50. [Google Scholar] [CrossRef]
  17. Postow, M.A.; Sidlow, R.; Hellmann, M.D. Immune-Related Adverse Events Associated with Immune Checkpoint Blockade. N. Engl. J. Med. 2018, 378, 158–168. [Google Scholar] [CrossRef]
  18. Darnell, E.P.; Mooradian, M.J.; Baruch, E.N.; Yilmaz, M.; Reynolds, K.L. Immune-Related Adverse Events (irAEs): Diagnosis, Management, and Clinical Pearls. Curr. Oncol. Rep. 2020, 22, 39. [Google Scholar] [CrossRef]
  19. Poto, R.; Troiani, T.; Criscuolo, G.; Marone, G.; Ciardiello, F.; Tocchetti, C.G.; Varricchi, G. Holistic Approach to Immune Checkpoint Inhibitor-Related Adverse Events. Front. Immunol. 2022, 13, 804597. [Google Scholar] [CrossRef]
  20. Farkona, S.; Diamandis, E.P.; Blasutig, I.M. Cancer immunotherapy: The beginning of the end of cancer? BMC Med. 2016, 14, 73. [Google Scholar] [CrossRef] [Green Version]
  21. Baumeister, S.H.; Freeman, G.J.; Dranoff, G.; Sharpe, A.H. Coinhibitory Pathways in Immunotherapy for Cancer. Annu. Rev. Immunol. 2016, 34, 539–573. [Google Scholar] [CrossRef] [PubMed]
  22. Buchbinder, E.I.; Desai, A. CTLA-4 and PD-1 Pathways: Similarities, Differences, and Implications of Their Inhibition. Am. J. Clin. Oncol. 2016, 39, 98–106. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Wolchok, J.D.; Chiarion-Sileni, V.; Gonzalez, R.; Rutkowski, P.; Grob, J.-J.; Cowey, C.L.; Lao, C.D.; Wagstaff, J.; Schadendorf, D.; Ferrucci, P.F.; et al. Overall Survival with Combined Nivolumab and Ipilimumab in Advanced Melanoma. N. Engl. J. Med. 2017, 377, 1345–1356. [Google Scholar] [CrossRef] [PubMed]
  24. Al-Habsi, M.; Chamoto, K.; Matsumoto, K.; Nomura, N.; Zhang, B.; Sugiura, Y.; Sonomura, K.; Maharani, A.; Nakajima, Y.; Wu, Y.; et al. Spermidine activates mitochondrial trifunctional protein and improves antitumor immunity in mice. Science 2022, 378, eabj3510. [Google Scholar] [CrossRef]
  25. Tanaka, K.; Chamoto, K.; Saeki, S.; Hatae, R.; Ikematsu, Y.; Sakai, K.; Ando, N.; Sonomura, K.; Kojima, S.; Taketsuna, M.; et al. Combination bezafibrate and nivolumab treatment of patients with advanced non–small cell lung cancer. Sci. Transl. Med. 2022, 14, eabq0021. [Google Scholar] [CrossRef]
  26. Cejuela, M.; Vethencourt, A.; Pernas, S. Immune Checkpoint Inhibitors and Novel Immunotherapy Approaches for Breast Cancer. Curr. Oncol. Rep. 2022, 24, 1801–1819. [Google Scholar] [CrossRef]
  27. Hou, W.; Yi, C.; Zhu, H. Predictive biomarkers of colon cancer immunotherapy: Present and future. Front. Immunol. 2022, 13, 1032314. [Google Scholar] [CrossRef]
  28. Langouo Fontsa, M.; Padonou, F.; Willard-Gallo, K. Biomarkers and immunotherapy: Where are we? Curr. Opin. Oncol. 2022, 34, 579–586. [Google Scholar] [CrossRef]
  29. Nobashi, T.; Baratto, L.; Reddy, S.A.; Srinivas, S.; Toriihara, A.; Hatami, N.; Yohannan, T.K.; Mittra, E. Predicting Response to Immunotherapy by Evaluating Tumors, Lymphoid Cell-Rich Organs, and Immune-Related Adverse Events Using FDG-PET/CT. Clin. Nucl. Med. 2019, 44, e272–e279. [Google Scholar] [CrossRef]
  30. Seyhan, A.A.; Carini, C. Insights and Strategies of Melanoma Immunotherapy: Predictive Biomarkers of Response and Resistance and Strategies to Improve Response Rates. Int. J. Mol. Sci. 2022, 24, 41. [Google Scholar] [CrossRef]
  31. Xiao, Z.; Mayer, A.T.; Nobashi, T.W.; Gambhir, S.S. ICOS Is an Indicator of T-cell–Mediated Response to Cancer Immunotherapy. Cancer Res. 2020, 80, 3023–3032. [Google Scholar] [CrossRef] [Green Version]
  32. Matson, V.; Fessler, J.; Bao, R.; Chongsuwat, T.; Zha, Y.; Alegre, M.-L.; Luke, J.J.; Gajewski, T.F. The commensal microbiome is associated with anti-PD-1 efficacy in metastatic melanoma patients. Science 2018, 359, 104–108. [Google Scholar] [CrossRef] [Green Version]
  33. Lee, K.A.; Thomas, A.M.; Bolte, L.A.; Björk, J.R.; de Ruijter, L.K.; Armanini, F.; Asnicar, F.; Blanco-Miguez, A.; Board, R.; Calbet-Llopart, N.; et al. Cross-cohort gut microbiome associations with immune checkpoint inhibitor response in advanced melanoma. Nat. Med. 2022, 28, 535–544. [Google Scholar] [CrossRef]
  34. Gonzalez-Mazón, I.; Sánchez-Bilbao, L.; Martín-Varillas, J.L.; García-Castaño, A.; Delgado-Ruiz, M.; Piña, I.B.; Hernández, J.L.; Castañeda, S.; Llorca, J.; González-Gay, M.A.; et al. Immune-related adverse events in patients with solid-organ tumours treated with immunotherapy: A 3-year study of 102 cases from a single centre. Clin. Exp. Rheumatol. 2021, 39, 612–620. [Google Scholar] [CrossRef]
  35. Tong, J.; Kartolo, A.; Yeung, C.; Hopman, W.; Baetz, T. Long-Term Toxicities of Immune Checkpoint Inhibitor (ICI) in Melanoma Patients. Curr. Oncol. 2022, 29, 7953–7963. [Google Scholar] [CrossRef]
  36. Granito, A.; Muratori, L.; Lalanne, C.; Quarneti, C.; Ferri, S.; Guidi, M.; Lenzi, M.; Muratori, P. Hepatocellular carcinoma in viral and autoimmune liver diseases: Role of CD4+ CD25+ Foxp3+ regulatory T cells in the immune microenvironment. World J. Gastroenterol. 2021, 27, 2994–3009. [Google Scholar] [CrossRef]
  37. Lee, D.J.; Lee, H.J., Jr.; Farmer, J.R.; Reynolds, K.L. Mechanisms Driving Immune-Related Adverse Events in Cancer Patients Treated with Immune Checkpoint Inhibitors. Curr. Cardiol. Rep. 2021, 23, 98. [Google Scholar] [CrossRef]
  38. Mangan, B.L.; McAlister, R.; Balko, J.M.; Johnson, D.B.; Moslehi, J.J.; Gibson, A.; Phillips, E.J. Evolving insights into the mechanisms of toxicity associated with immune checkpoint inhibitor therapy. Br. J. Clin. Pharmacol. 2020, 86, 1778–1789. [Google Scholar] [CrossRef]
  39. Khoja, L.; Day, D.; Chen, T.W.-W.; Siu, L.L.; Hansen, A.R. Tumour- and class-specific patterns of immune-related adverse events of immune checkpoint inhibitors: A systematic review. Ann. Oncol. 2017, 28, 2377–2385. [Google Scholar] [CrossRef]
  40. Kotwal, A. Hypophysitis from immune checkpoint inhibitors: Challenges in diagnosis and management. Curr. Opin. Endocrinol. Diabetes Obes. 2021, 28, 427–434. [Google Scholar] [CrossRef]
  41. Nobashi, T.W.; Nishimoto, Y.; Kawata, Y.; Yutani, H.; Nakamura, M.; Tsuji, Y.; Yoshida, A.; Sugimoto, A.; Yamamoto, T.; Alam, I.S.; et al. Clinical and radiological features of immune checkpoint inhibitor-related pneumonitis in lung cancer and non-lung cancers. Br. J. Radiol. 2020, 93, 20200409. [Google Scholar] [CrossRef] [PubMed]
  42. Huang, A.C.; Zappasodi, R. A decade of checkpoint blockade immunotherapy in melanoma: Understanding the molecular basis for immune sensitivity and resistance. Nat. Immunol. 2022, 23, 660–670. [Google Scholar] [CrossRef] [PubMed]
  43. Carthon, B.C.; Wolchok, J.D.; Yuan, J.; Kamat, A.; Tang, D.S.N.; Sun, J.; Ku, G.; Troncoso, P.; Logothetis, C.J.; Allison, J.P.; et al. Preoperative CTLA-4 blockade: Tolerability and immune monitoring in the setting of a presurgical clinical trial. Clin. Cancer Res. 2010, 16, 2861–2871. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Ng Tang, D.; Shen, Y.; Sun, J.; Wen, S.; Wolchok, J.D.; Yuan, J.; Allison, J.P.; Sharma, P. Increased frequency of ICOS+CD4 T cells as a pharmacodynamic biomarker for anti-CTLA-4 therapy. Cancer Immunol. Res. 2013, 1, 229–234. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Romano, E.; Kusio-Kobialka, M.; Foukas, P.G.; Baumgaertner, P.; Meyer, C.; Ballabeni, P.; Michielin, O.; Weide, B.; Romero, P.; Speiser, D.E. Ipilimumab-dependent cell-mediated cytotoxicity of regulatory T cells ex vivo by nonclassical monocytes in melanoma patients. Proc. Natl. Acad. Sci. USA 2015, 112, 6140–6145. [Google Scholar] [CrossRef] [Green Version]
  46. Sharma, A.; Subudhi, S.K.; Blando, J.; Scutti, J.; Vence, L.; Wargo, J.; Allison, J.P.; Ribas, A.; Sharma, P. Anti-CTLA-4 Immunotherapy Does Not Deplete FOXP3(+) Regulatory T Cells (Tregs) in Human Cancers. Clin. Cancer Res. 2019, 25, 1233–1238. [Google Scholar] [CrossRef] [Green Version]
  47. Huang, A.C.; Orlowski, R.J.; Xu, X.; Mick, R.; George, S.M.; Yan, P.K.; Manne, S.; Kraya, A.A.; Wubbenhorst, B.; Dorfman, L.; et al. A single dose of neoadjuvant PD-1 blockade predicts clinical outcomes in resectable melanoma. Nat. Med. 2019, 25, 454–461. [Google Scholar] [CrossRef]
  48. Kamada, T.; Togashi, Y.; Tay, C.; Ha, D.; Sasaki, A.; Nakamura, Y.; Sato, E.; Fukuoka, S.; Tada, Y.; Tanaka, A.; et al. PD-1+ regulatory T cells amplified by PD-1 blockade promote hyperprogression of cancer. Proc. Natl. Acad. Sci. USA 2019, 116, 9999–10008. [Google Scholar] [CrossRef] [Green Version]
  49. Kumagai, S.; Togashi, Y.; Kamada, T.; Sugiyama, E.; Nishinakamura, H.; Takeuchi, Y.; Vitaly, K.; Itahashi, K.; Maeda, Y.; Matsui, S.; et al. The PD-1 expression balance between effector and regulatory T cells predicts the clinical efficacy of PD-1 blockade therapies. Nat. Immunol. 2020, 21, 1346–1358. [Google Scholar] [CrossRef]
  50. Johnson, D.B.; Balko, J.M.; Compton, M.L.; Chalkias, S.; Gorham, J.; Xu, Y.; Hicks, M.; Puzanov, I.; Alexander, M.R.; Bloomer, T.L.; et al. Fulminant Myocarditis with Combination Immune Checkpoint Blockade. N. Engl. J. Med. 2016, 375, 1749–1755. [Google Scholar] [CrossRef]
  51. Zhu, H.; Galdos, F.X.; Lee, D.; Waliany, S.; Huang, Y.V.; Ryan, J.; Dang, K.; Neal, J.W.; Wakelee, H.A.; Reddy, S.A.; et al. Identification of Pathogenic Immune Cell Subsets Associated With Checkpoint Inhibitor-Induced Myocarditis. Circulation 2022, 146, 316–335. [Google Scholar] [CrossRef]
  52. Groha, S.; Alaiwi, S.A.; Xu, W.; Naranbhai, V.; Nassar, A.H.; Bakouny, Z.; El Zarif, T.; Saliby, R.M.; Wan, G.; Rajeh, A.; et al. Germline variants associated with toxicity to immune checkpoint blockade. Nat. Med. 2022, 28, 2584–2591. [Google Scholar] [CrossRef]
  53. Taylor, C.A.; Watson, R.A.; Tong, O.; Ye, W.; Nassiri, I.; Gilchrist, J.J.; de Los Aires, A.V.; Sharma, P.K.; Koturan, S.; Cooper, R.A.; et al. IL7 genetic variation and toxicity to immune checkpoint blockade in patients with melanoma. Nat. Med. 2022, 28, 2592–2600. [Google Scholar] [CrossRef]
  54. Barata, J.T.; Durum, S.K.; Seddon, B. Flip the coin: IL-7 and IL-7R in health and disease. Nat. Immunol. 2019, 20, 1584–1593. [Google Scholar] [CrossRef]
  55. Winer, H.; Rodrigues, G.O.; Hixon, J.A.; Aiello, F.B.; Hsu, T.C.; Wachter, B.T.; Li, W.; Durum, S.K. IL-7: Comprehensive review. Cytokine 2022, 160, 156049. [Google Scholar] [CrossRef]
  56. Chatzidionysiou, K.; Liapi, M.; Tsakonas, G.; Gunnarsson, I.; Catrina, A. Treatment of rheumatic immune-related adverse events due to cancer immunotherapy with immune checkpoint inhibitors-is it time for a paradigm shift? Clin. Rheumatol. 2021, 40, 1687–1695. [Google Scholar] [CrossRef]
  57. Tivol, E.A.; Borriello, F.; Schweitzer, A.N.; Lynch, W.P.; Bluestone, J.A.; Sharpe, A.H. Loss of CTLA-4 leads to massive lymphoproliferation and fatal multiorgan tissue destruction, revealing a critical negative regulatory role of CTLA-4. Immunity 1995, 3, 541–547. [Google Scholar] [CrossRef] [Green Version]
  58. Waterhouse, P.; Penninger, J.M.; Timms, E.; Wakeham, A.; Shahinian, A.; Lee, K.P.; Thompson, C.B.; Griesser, H.; Mak, T.W. Lymphoproliferative Disorders with Early Lethality in Mice Deficient in Ctla-4. Science 1995, 270, 985–988. [Google Scholar] [CrossRef]
  59. Nishimura, H.; Nose, M.; Hiai, H.; Minato, N.; Honjo, T. Development of lupus-like autoimmune diseases by disruption of the PD-1 gene encoding an ITIM motif-carrying immunoreceptor. Immunity 1999, 11, 141–151. [Google Scholar] [CrossRef] [Green Version]
  60. Nishimura, H.; Okazaki, T.; Tanaka, Y.; Nakatani, K.; Hara, M.; Matsumori, A.; Sasayama, S.; Mizoguchi, A.; Hiai, H.; Minato, N.; et al. Autoimmune dilated cardiomyopathy in PD-1 receptor-deficient mice. Science 2001, 291, 319–322. [Google Scholar] [CrossRef]
  61. Watanabe, R.; Hashimoto, M. Elucidation of disease mechanisms underlying rheumatic immune-related adverse events may lead to novel therapeutic strategies for autoimmune diseases. Ann. Rheum. Dis. 2020, 81, e262. [Google Scholar] [CrossRef] [PubMed]
  62. Shen, P.; Deng, X.; Hu, Z.; Chen, Z.; Huang, Y.; Wang, K.; Qin, K.; Huang, Y.; Ba, X.; Yan, J.; et al. Rheumatic Manifestations and Diseases From Immune Checkpoint Inhibitors in Cancer Immunotherapy. Front. Med. 2021, 8, 762247. [Google Scholar] [CrossRef] [PubMed]
  63. Bardoscia, L.; Pasinetti, N.; Triggiani, L.; Cozzi, S.; Sardaro, A. Biological Bases of Immune-Related Adverse Events and Potential Crosslinks With Immunogenic Effects of Radiation. Front. Pharmacol. 2021, 12, 746853. [Google Scholar] [CrossRef]
  64. June, C.H.; Warshauer, J.T.; Bluestone, J.A. Is autoimmunity the Achilles’ heel of cancer immunotherapy? Nat. Med. 2017, 23, 540–547. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Esfahani, K.; Miller, W.H., Jr. Reversal of Autoimmune Toxicity and Loss of Tumor Response by Interleukin-17 Blockade. N. Engl. J. Med. 2017, 376, 1989–1991. [Google Scholar] [CrossRef]
  66. Holmstroem, R.B.; Nielsen, O.H.; Jacobsen, S.; Riis, L.B.; Theile, S.; Bjerrum, J.T.; Vilmann, P.; Johansen, J.S.; Boisen, M.K.; Eefsen, R.H.L.; et al. COLAR: Open-label clinical study of IL-6 blockade with tocilizumab for the treatment of immune checkpoint inhibitor-induced colitis and arthritis. J. Immunother. Cancer 2022, 10, e005111. [Google Scholar] [CrossRef]
  67. Cappelli, L.C.; Bingham, C.O., 3rd. Expert Perspective: Immune Checkpoint Inhibitors and Rheumatologic Complications. Arthritis Rheumatol. 2021, 73, 553–565. [Google Scholar] [CrossRef]
  68. Narváez, J.; Juarez-López, P.; Lluch, J.; Palmero, R.; del Muro, X.G.; Nolla, J.; Domenech, E.D. Rheumatic immune-related adverse events in patients on anti-PD-1 inhibitors: Fasciitis with myositis syndrome as a new complication of immunotherapy. Autoimmun. Rev. 2018, 17, 1040–1045. [Google Scholar] [CrossRef]
  69. Reid, P.; Cappelli, L.C. Treatment of rheumatic adverse events of cancer immunotherapy. Best Pract. Res. Clin. Rheumatol. 2022, 101805. [Google Scholar] [CrossRef]
  70. Roberts, J.; Ennis, D.; Hudson, M.; Ye, C.; Saltman, A.; Himmel, M.; Rottapel, R.; Pope, J.; Hoa, S.; Tisseverasinghe, A.; et al. Rheumatic immune-related adverse events associated with cancer immunotherapy: A nationwide multi-center cohort. Autoimmun. Rev. 2020, 19, 102595. [Google Scholar] [CrossRef]
  71. Xiao, Y.; Zeng, L.; Shen, Q.; Zhou, Z.; Mao, Z.; Wang, Q.; Zhang, X.; Li, Y.; Yao, W. Diagnosis and Treatment of Rheumatic Adverse Events Related to Immune Checkpoint Inhibitors. J. Immunol. Res. 2020, 2020, 2640273. [Google Scholar] [CrossRef]
  72. Zhong, H.; Zhou, J.; Xu, D.; Zeng, X. Rheumatic immune-related adverse events induced by immune checkpoint inhibitors. Asia Pac. J. Clin. Oncol. 2020, 17, 178–185. [Google Scholar] [CrossRef]
  73. Genovese, M.C.; Becker, J.-C.; Schiff, M.; Luggen, M.; Sherrer, Y.; Kremer, J.; Birbara, C.; Box, J.; Natarajan, K.; Nuamah, I.; et al. Abatacept for rheumatoid arthritis refractory to tumor necrosis factor alpha inhibition. N. Engl. J. Med. 2005, 353, 1114–1123. [Google Scholar] [CrossRef] [Green Version]
  74. Okazaki, S.; Watanabe, R.; Harigae, H.; Fujii, H. Better Retention of Abatacept Is Associated with High Rheumatoid Factor: A Five-Year Follow-Up Study of Patients with Rheumatoid Arthritis. Tohoku J. Exp. Med. 2020, 250, 153–159. [Google Scholar] [CrossRef] [Green Version]
  75. Kremer, J.M.; Westhovens, R.; Leon, M.; Di Giorgio, E.; Alten, R.; Steinfeld, S.; Russell, A.; Dougados, M.; Emery, P.; Nuamah, I.F.; et al. Treatment of rheumatoid arthritis by selective inhibition of T-cell activation with fusion protein CTLA4Ig. N. Engl. J. Med. 2003, 349, 1907–1915. [Google Scholar] [CrossRef]
  76. Lingel, H.; Brunner-Weinzierl, M.C. CTLA-4 (CD152): A versatile receptor for immune-based therapy. Semin. Immunol. 2019, 42, 101298. [Google Scholar] [CrossRef]
  77. Kostine, M.; Finckh, A.; Bingham, C.O.; Visser, K.; Leipe, J.; Schulze-Koops, H.; Choy, E.H.; Benesova, K.; Radstake, T.; Cope, A.P.; et al. EULAR points to consider for the diagnosis and management of rheumatic immune-related adverse events due to cancer immunotherapy with checkpoint inhibitors. Ann. Rheum. Dis. 2021, 80, 36–48. [Google Scholar] [CrossRef] [Green Version]
  78. Jespersen, M.S.; Fano, S.; Stenor, C.; Moller, A.K. A case report of immune checkpoint inhibitor-related steroid-refractory myocarditis and myasthenia gravis-like myositis treated with abatacept and mycophenolate mofetil. Eur. Heart J. Case Rep. 2021, 5, ytab342. [Google Scholar] [CrossRef]
  79. Liu, S.; Chan, J.; Brinc, D.; Gandhi, S.; Izenberg, A.; Delgado, D.; Abdel-Qadir, H.; Wintersperger, B.J.; Thavendiranathan, P. Immune Checkpoint Inhibitor-Associated Myocarditis With Persistent Troponin Elevation Despite Abatacept and Prolonged Immunosuppression. JACC Cardio Oncol. 2020, 2, 800–804. [Google Scholar] [CrossRef]
  80. Salem, J.E.; Allenbach, Y.; Vozy, A.; Brechot, N.; Johnson, D.B.; Moslehi, J.J.; Kerneis, M. Abatacept for Severe Immune Checkpoint Inhibitor-Associated Myocarditis. N. Engl. J. Med. 2019, 380, 2377–2379. [Google Scholar] [CrossRef]
  81. Belkhir, R.; Burel, S.L.; Dunogeant, L.; Marabelle, A.; Hollebecque, A.; Besse, B.; Leary, A.; Voisin, A.L.; Pontoizeau, C.; Coutte, L.; et al. Rheumatoid arthritis and polymyalgia rheumatica occurring after immune checkpoint inhibitor treatment. Ann. Rheum. Dis. 2017, 76, 1747–1750. [Google Scholar] [CrossRef] [PubMed]
  82. Cappelli, L.C.; Gutierrez, A.K.; Baer, A.N.; Albayda, J.; Manno, R.L.; Haque, U.; Lipson, E.J.; Bleich, K.B.; Shah, A.A.; Naidoo, J.; et al. Inflammatory arthritis and sicca syndrome induced by nivolumab and ipilimumab. Ann. Rheum. Dis. 2017, 76, 43–50. [Google Scholar] [CrossRef] [PubMed]
  83. Liu, Y.; Jaquith, J.M.; McCarthy-Fruin, K.; Zhu, X.; Zhou, X.; Li, Y.; Crowson, C.; Davis, J.M., 3rd; Thanarajasingam, U.; Zeng, H. Immune checkpoint inhibitor-induced inflammatory arthritis: A novel clinical entity with striking similarities to seronegative rheumatoid arthritis. Clin. Rheumatol. 2020, 39, 3631–3637. [Google Scholar] [CrossRef] [PubMed]
  84. Nobashi, T.; Mittra, E. PD-1 Blockade–induced Inflammatory Arthritis. Radiology 2018, 289, 616. [Google Scholar] [CrossRef] [Green Version]
  85. Ponce, A.; Frade-Sosa, B.; Sarmiento-Monroy, J.C.; Sapena, N.; Ramírez, J.; Azuaga, A.B.; Morlà, R.; Ruiz-Esquide, V.; Cañete, J.D.; Sanmartí, R.; et al. Imaging Findings in Patients with Immune Checkpoint Inhibitor-Induced Arthritis. Diagnostics 2022, 12, 1961. [Google Scholar] [CrossRef]
  86. Pundole, X.; Abdel-Wahab, N.; Suarez-Almazor, M.E. Arthritis risk with immune checkpoint inhibitor therapy for cancer. Curr. Opin. Rheumatol. 2019, 31, 293–299. [Google Scholar] [CrossRef]
  87. Richter, M.D.; Crowson, C.; Kottschade, L.A.; Finnes, H.D.; Markovic, S.N.; Thanarajasingam, U. Rheumatic Syndromes Associated With Immune Checkpoint Inhibitors: A Single-Center Cohort of Sixty-One Patients. Arthritis Rheumatol. 2019, 71, 468–475. [Google Scholar] [CrossRef] [Green Version]
  88. Cappelli, L.C.; Bingham, C.O.; Forde, P.M.; Anagnostou, V.; Brahmer, J.; Lipson, E.J.; Mammen, J.; Schollenberger, M.; Shah, A.A.; Darrah, E. Anti-RA33 antibodies are present in a subset of patients with immune checkpoint inhibitor-induced inflammatory arthritis. RMD Open 2022, 8, e002511. [Google Scholar] [CrossRef]
  89. Maxwell, R.; Luksik, A.S.; Garzon-Muvdi, T.; Hung, A.L.; Kim, E.S.; Wu, A.; Xia, Y.; Belcaid, Z.; Gorelick, N.; Choi, J.; et al. Contrasting impact of corticosteroids on anti-PD-1 immunotherapy efficacy for tumor histologies located within or outside the central nervous system. Oncoimmunology 2018, 7, e1500108. [Google Scholar] [CrossRef] [Green Version]
  90. Naidoo, J.; Cappelli, L.C.; Forde, P.M.; Marrone, K.A.; Lipson, E.J.; Hammers, H.J.; Sharfman, W.H.; Le, D.T.; Baer, A.N.; Shah, A.A.; et al. Inflammatory Arthritis: A Newly Recognized Adverse Event of Immune Checkpoint Blockade. Oncologist 2017, 22, 627–630. [Google Scholar] [CrossRef] [Green Version]
  91. Yang, H.; Xia, L.; Chen, J.; Zhang, S.; Martin, V.; Li, Q.; Lin, S.; Chen, J.; Calmette, J.; Lu, M.; et al. Stress–glucocorticoid–TSC22D3 axis compromises therapy-induced antitumor immunity. Nat. Med. 2019, 25, 1428–1441. [Google Scholar] [CrossRef]
  92. Abe, K.; Ishikawa, Y.; Fujiwara, M.; Yukawa, H.; Yanagihara, T.; Takei, S.; Arioka, H.; Kita, Y. Immune checkpoint inhibitor-induced refractory polyarthritis rapidly improved by sarilumab and monitoring with joint ultrasonography: A case report. Medicine 2022, 101, e28428. [Google Scholar] [CrossRef]
  93. Subedi, A.; Williams, S.G.; Yao, L.; Maharjan, S.; Strauss, J.; Sharon, E.; Thomas, A.; Apolo, A.B.; Gourh, P.; Hasni, S.A.; et al. Use of Magnetic Resonance Imaging to Identify Immune Checkpoint Inhibitor-Induced Inflammatory Arthritis. JAMA Netw. Open 2020, 3, e200032. [Google Scholar] [CrossRef] [Green Version]
  94. Kim, S.T.; Tayar, J.; Trinh, V.A.; Suarez-Almazor, M.; Garcia, S.; Hwu, P.; Johnson, D.H.; Uemura, M.; Diab, A. Successful treatment of arthritis induced by checkpoint inhibitors with tocilizumab: A case series. Ann. Rheum. Dis. 2017, 76, 2061–2064. [Google Scholar] [CrossRef]
  95. Stroud, C.R.; Hegde, A.; Cherry, C.; Naqash, A.R.; Sharma, N.; Addepalli, S.; Cherukuri, S.; Parent, T.; Hardin, J.; Walker, P. Tocilizumab for the management of immune mediated adverse events secondary to PD-1 blockade. J. Oncol. Pharm. Pract. 2019, 25, 551–557. [Google Scholar] [CrossRef]
  96. Draghi, A.; Borch, T.H.; Radic, H.D.; Chamberlain, C.A.; Gokuldass, A.; Svane, I.M.; Donia, M. Differential effects of corticosteroids and anti-TNF on tumor-specific immune responses: Implications for the management of irAEs. Int. J. Cancer 2019, 145, 1408–1413. [Google Scholar] [CrossRef]
  97. De La Fuente, F.; Belkhir, R.; Henry, J.; Nguyen, C.D.; Pham, T.; Germain, V.; Gavand, P.E.; Labadie, C.; Briere, C.; Lauret, A.; et al. Use of a bDMARD or tsDMARD for the management of inflammatory arthritis under checkpoint inhibitors: An observational study. RMD Open 2022, 8, e002612. [Google Scholar] [CrossRef]
  98. Benesova, K.; Kraus, F.V.; Carvalho, R.; Lorenz, H.; Hörth, C.H.; Günther, J.; Klika, K.D.; Graf, J.; Diekmann, L.; Schank, T.; et al. Distinct immune-effector and metabolic profile of CD8+ T cells in patients with autoimmune polyarthritis induced by therapy with immune checkpoint inhibitors. Ann. Rheum. Dis. 2022, 81, 1730–1741. [Google Scholar] [CrossRef]
  99. Ytterberg, S.R.; Bhatt, D.L.; Mikuls, T.R.; Koch, G.G.; Fleischmann, R.; Rivas, J.L.; Germino, R.; Menon, S.; Sun, Y.; Wang, C.; et al. Cardiovascular and Cancer Risk with Tofacitinib in Rheumatoid Arthritis. N. Engl. J. Med. 2022, 386, 316–326. [Google Scholar] [CrossRef]
  100. Leonardi, G.C.; Gainor, J.F.; Altan, M.; Kravets, S.; Dahlberg, S.E.; Gedmintas, L.; Azimi, R.; Rizvi, H.; Riess, J.W.; Hellmann, M.D.; et al. Safety of Programmed Death-1 Pathway Inhibitors Among Patients With Non-Small-Cell Lung Cancer and Preexisting Autoimmune Disorders. J. Clin. Oncol. 2018, 36, 1905–1912. [Google Scholar] [CrossRef]
  101. Tison, A.; Garaud, S.; Chiche, L.; Cornec, D.; Kostine, M. Immune-checkpoint inhibitor use in patients with cancer and pre-existing autoimmune diseases. Nat. Rev. Rheumatol. 2022, 18, 641–656. [Google Scholar] [CrossRef] [PubMed]
  102. Plaçais, L.; Dalle, S.; Dereure, O.; Trabelsi, S.; Dalac, S.; Legoupil, D.; Montaudié, H.; Arnault, J.-P.; De Quatrebarbes, J.; Saiag, P.; et al. Risk of irAEs in patients with autoimmune diseases treated by immune checkpoint inhibitors for stage III or IV melanoma: Results from a matched case-control study. Ann. Rheum. Dis. 2022, 81, 1445–1452. [Google Scholar] [CrossRef] [PubMed]
  103. Allenbach, Y.; Anquetil, C.; Manouchehri, A.; Benveniste, O.; Lambotte, O.; Lebrun-Vignes, B.; Spano, J.-P.; Ederhy, S.; Klatzmann, D.; Rosenzwajg, M.; et al. Immune checkpoint inhibitor-induced myositis, the earliest and most lethal complication among rheumatic and musculoskeletal toxicities. Autoimmun. Rev. 2020, 19, 102586. [Google Scholar] [CrossRef] [PubMed]
  104. Angelopoulou, F.; Bogdanos, D.; Dimitroulas, T.; Sakkas, L.; Daoussis, D. Immune checkpoint inhibitor-induced musculoskeletal manifestations. Rheumatol. Int. 2021, 41, 33–42. [Google Scholar] [CrossRef]
  105. Nakagomi, Y.; Tajiri, K.; Shimada, S.; Li, S.; Inoue, K.; Murakata, Y.; Murata, M.; Sakai, S.; Sato, K.; Ieda, M. Immune Checkpoint Inhibitor-Related Myositis Overlapping With Myocarditis: An Institutional Case Series and a Systematic Review of Literature. Front. Pharmacol. 2022, 13, 884776. [Google Scholar] [CrossRef]
  106. Hamada, N.; Maeda, A.; Takase-Minegishi, K.; Kirino, Y.; Sugiyama, Y.; Namkoong, H.; Horita, N.; Yoshimi, R.; Nakajima, H.; YCU irAE Working Group. Incidence and Distinct Features of Immune Checkpoint Inhibitor-Related Myositis From Idiopathic Inflammatory Myositis: A Single-Center Experience With Systematic Literature Review and Meta-Analysis. Front. Immunol. 2021, 12, 803410. [Google Scholar]
  107. Seki, M.; Uruha, A.; Ohnuki, Y.; Kamada, S.; Noda, T.; Onda, A.; Ohira, M.; Isami, A.; Hiramatsu, S.; Hibino, M.; et al. Inflammatory myopathy associated with PD-1 inhibitors. J. Autoimmun. 2019, 100, 105–113. [Google Scholar] [CrossRef]
  108. Daoussis, D.; Kraniotis, P.; Liossis, S.N.; Solomou, A. Immune checkpoint inhibitor-induced myo-fasciitis. Rheumatology 2017, 56, 2161. [Google Scholar] [CrossRef] [Green Version]
  109. Isa, M.; Hongo, Y.; Sakamoto, N.; Yamazaki, K.; Takazaki, H.; Asakuma, J.; Ikewaki, K.; Suzuki, K. Immune checkpoint inhibitor-related myositis and myocarditis with multiple myositis-specific/−associated antibodies. J. Neurol. Sci. 2023, 444, 120528. [Google Scholar] [CrossRef]
  110. Yamamoto, A.M.; Gajdos, P.; Eymard, B.; Tranchant, C.; Warter, J.M.; Gomez, L.; Bourquin, C.; Bach, J.F.; Garchon, H.J. Anti-titin antibodies in myasthenia gravis: Tight association with thymoma and heterogeneity of nonthymoma patients. Arch. Neurol. 2001, 58, 885–890. [Google Scholar] [CrossRef] [Green Version]
  111. Soman, B.; Dias, M.C.; Rizvi, S.A.J.; Kardos, A. Myasthenia gravis, myositis and myocarditis: A fatal triad of immune-related adverse effect of immune checkpoint inhibitor treatment. BMJ Case Rep. 2022, 15, e251966. [Google Scholar] [CrossRef]
  112. Brahmer, J.R.; Lacchetti, C.; Schneider, B.J.; Atkins, M.B.; Brassil, K.J.; Caterino, J.M.; Chau, I.; Ernstoff, M.S.; Gardner, J.M.; Ginex, P.; et al. Management of Immune-Related Adverse Events in Patients Treated With Immune Checkpoint Inhibitor Therapy: American Society of Clinical Oncology Clinical Practice Guideline. J. Clin. Oncol. 2018, 36, 1714–1768. [Google Scholar] [CrossRef]
  113. Weyand, C.M.; Goronzy, J.J. Immunology of Giant Cell Arteritis. Circ. Res. 2023, 132, 238–250. [Google Scholar] [CrossRef]
  114. Daxini, A.; Cronin, K.; Sreih, A.G. Vasculitis associated with immune checkpoint inhibitors-a systematic review. Clin. Rheumatol. 2018, 37, 2579–2584. [Google Scholar] [CrossRef]
  115. Watanabe, R.; Berry, G.J.; Liang, D.H.; Goronzy, J.J.; Weyand, C.M. Cellular Signaling Pathways in Medium and Large Vessel Vasculitis. Front. Immunol. 2020, 11, 587089. [Google Scholar] [CrossRef]
  116. Jennette, J.C.; Falk, R.J.; Bacon, P.A.; Basu, N.; Cid, M.C.; Ferrario, F.; Flores-Suarez, L.F.; Gross, W.L.; Guillevin, L.; Hagen, E.C.; et al. 2012 revised International Chapel Hill Consensus Conference Nomenclature of Vasculitides. Arthritis Rheum. 2013, 65, 1–11. [Google Scholar] [CrossRef]
  117. Goldstein, B.L.; Gedmintas, L.; Todd, D.J. Drug-associated polymyalgia rheumatica/giant cell arteritis occurring in two patients after treatment with ipilimumab, an antagonist of ctla-4. Arthritis Rheumatol. 2014, 66, 768–769. [Google Scholar] [CrossRef]
  118. Narala, R.; Reddy, S.A.; Mruthyunjaya, P. Giant cell arteritis manifesting as retinal arterial occlusion and paracentral acute middle maculopathy in a patient on pembrolizumab for metastatic uveal melanoma. Am. J. Ophthalmol. Case Rep. 2020, 20, 100891. [Google Scholar] [CrossRef]
  119. Bloomer, C.H.; Annabathula, R.V.; Aggarwal, V.; Upadhya, B.; Lycan, T.W. A Case Report of Immune Checkpoint Inhibitor-Induced Aortitis Treated with Tocilizumab. Case Rep. Immunol. 2022, 2022, 7971169. [Google Scholar] [CrossRef]
  120. Henderson, D.; Eslamian, G.; Poon, D.; Crabb, S.; Jones, R.; Sankey, P.; Kularatne, B.; Linch, M.; Josephs, D. Immune checkpoint inhibitor induced large vessel vasculitis. BMJ Case Rep. 2020, 13, e233496. [Google Scholar] [CrossRef]
  121. Mort, J.; Maheshwari, S.; Basu, N.; Dillon, P.; Brady, K.; Bear, H.; Millard, T. A Rare Case of Large-Vessel Vasculitis following Checkpoint Inhibitor Therapy and Pegfilgrastim. Case Rep. Oncol. Med. 2022, 2022, 7295305. [Google Scholar] [CrossRef] [PubMed]
  122. Belkaid, S.; Berger, M.; Nouvier, M.; Picard, C.; Dalle, S. A case of Schönlein-Henoch purpura induced by immune checkpoint inhibitor in a patient with metastatic melanoma. Eur. J. Cancer 2020, 139, 169–172. [Google Scholar] [CrossRef] [PubMed]
  123. Nagaoka-Takatori, A.; Ishii, M.; Hayama, K.; Obinata, D.; Yamaguchi, K.; Takahashi, S.; Fujita, H. A Case of IgA Vasculitis During Nivolumab Therapy for Renal Cell Carcinoma. Clin. Cosmet. Investig. Dermatol. 2021, 14, 1885–1888. [Google Scholar] [CrossRef] [PubMed]
  124. Sibille, A.; Alfieri, R.; Malaise, O.; Detrembleur, N.; Pirotte, M.; Louis, R.; Duysinx, B. Granulomatosis With Polyangiitis in a Patient on Programmed Death-1 Inhibitor for Advanced Non-small-cell Lung Cancer. Front. Oncol. 2019, 9, 478. [Google Scholar] [CrossRef]
  125. Hung, W.; Cusnir, I.; Habib, S.; Smylie, M.; Solez, K.; Yacyshyn, E. Immune checkpoint inhibitor-induced granulomatosis with polyangiitis. Rheumatology 2021, 60, e190–e191. [Google Scholar] [CrossRef]
  126. Roger, A.; Groh, M.; Lorillon, G.; Le Pendu, C.; Maillet, J.; Arangalage, D.; Tazi, A.; Lebbe, C.; Baroudjian, B.; Delyon, J. Eosinophilic granulomatosis with polyangiitis (Churg-Strauss) induced by immune checkpoint inhibitors. Ann. Rheum. Dis. 2019, 78, e82. [Google Scholar] [CrossRef]
  127. Ak, T.; Durmus, R.B.; Onel, M. Cutaneous vasculitis associated with molecular tergeted therapies: Systematic review of the literature. Clin. Rheumatol. 2023, 42, 339–357. [Google Scholar] [CrossRef]
  128. Panian, J.; Pan, E.; Shi, V.; Hinds, B.; McKay, R.R. Leukocytoclastic Vasculitis Induced by Immune Checkpoint Inhibition in a Patient With Advanced Renal Cell Carcinoma. Oncology 2022, 36, 316–320. [Google Scholar]
  129. Salem, J.E.; Manouchehri, A.; Moey, M.; Lebrun-Vignes, B.; Bastarache, L.; Pariente, A.; Gobert, A.; Spano, J.P.; Balko, J.M.; Bonaca, M.P.; et al. Cardiovascular toxicities associated with immune checkpoint inhibitors: An observational, retrospective, pharmacovigilance study. Lancet Oncol. 2018, 19, 1579–1589. [Google Scholar] [CrossRef]
  130. Zhang, H.; Watanabe, R.; Berry, G.J.; Vaglio, A.; Liao, Y.J.; Warrington, K.J.; Goronzy, J.J.; Weyand, C.M. Immunoinhibitory checkpoint deficiency in medium and large vessel vasculitis. Proc. Natl. Acad. Sci. USA 2017, 114, E970–E979. [Google Scholar] [CrossRef] [Green Version]
  131. Watanabe, R.; Maeda, T.; Zhang, H.; Berry, G.J.; Zeisbrich, M.; Brockett, R.; Greenstein, A.E.; Tian, L.; Goronzy, J.J.; Weyand, C.M. MMP (Matrix Metalloprotease)-9–Producing Monocytes Enable T Cells to Invade the Vessel Wall and Cause Vasculitis. Circ. Res. 2018, 123, 700–715. [Google Scholar] [CrossRef]
  132. Pugh, D.; Karabayas, M.; Basu, N.; Cid, M.C.; Goel, R.; Goodyear, C.S.; Grayson, P.C.; McAdoo, S.P.; Mason, J.C.; Owen, C.; et al. Large-vessel vasculitis. Nat. Rev. Dis. Prim. 2022, 7, 93. [Google Scholar] [CrossRef]
  133. Sun, L.; Zhang, W.; Zhao, L.; Zhao, Y.; Wang, F.; Lew, A.M.; Xu, Y. Self-Tolerance of Vascular Tissues Is Broken Down by Vascular Dendritic Cells in Response to Systemic Inflammation to Initiate Regional Autoinflammation. Front. Immunol. 2022, 13, 823853. [Google Scholar] [CrossRef]
  134. Wilde, B.; Hua, F.; Dolff, S.; Jun, C.; Cai, X.; Specker, C.; Feldkamp, T.; Kribben, A.; Cohen Tervaert, J.W.; Witzke, O. Aberrant expression of the negative costimulator PD-1 on T cells in granulomatosis with polyangiitis. Rheumatology 2012, 51, 1188–1197. [Google Scholar] [CrossRef] [Green Version]
  135. Zeisbrich, M.; Chevalier, N.; Sehnert, B.; Rizzi, M.; Venhoff, N.; Thiel, J.; Voll, R.E. CMTM6-Deficient Monocytes in ANCA-Associated Vasculitis Fail to Present the Immune Checkpoint PD-L1. Front. Immunol. 2021, 12, 673912. [Google Scholar] [CrossRef]
  136. Watanabe, R.; Berry, G.J.; Liang, D.H.; Goronzy, J.J.; Weyand, C.M. Pathogenesis of Giant Cell Arteritis and Takayasu Arteritis-Similarities and Differences. Curr. Rheumatol. Rep. 2020, 22, 68. [Google Scholar] [CrossRef]
  137. Yaseen, K.; Nevares, A.; Tamaki, H. A Spotlight on Drug-Induced Vasculitis. Curr. Rheumatol. Rep. 2022, 24, 323–336. [Google Scholar] [CrossRef]
  138. Van den Brom, R.R.; Abdulahad, W.H.; Rutgers, A.; Kroesen, B.J.; Roozendaal, C.; de Groot, D.J.; Schroder, C.P.; Hospers, G.A.; Brouwer, E. Rapid granulomatosis with polyangiitis induced by immune checkpoint inhibition. Rheumatology 2016, 55, 1143–1145. [Google Scholar] [CrossRef] [Green Version]
  139. Mamlouk, O.; Lin, J.S.; Abdelrahim, M.; Tchakarov, A.S.; Glass, W.F.; Selamet, U.; Buni, M.; Abdel-Wahab, N.; Abudayyeh, A. Checkpoint inhibitor-related renal vasculitis and use of rituximab. J. Immunother. Cancer 2020, 8, e000750. [Google Scholar] [CrossRef]
  140. Wulfken, L.M.; Becker, J.C.; Hayajneh, R.; Wagner, A.D.; Schaper-Gerhardt, K.; Flatt, N.; Grimmelmann, I.; Gutzmer, R. Case Report: Sustained Remission Due to PD-1-Inhibition in a Metastatic Melanoma Patient With Depleted B Cells. Front. Immunol. 2021, 12, 733961. [Google Scholar] [CrossRef]
  141. Laamech, R.; Terrec, F.; Emprou, C.; Toffart, A.C.; Pierret, T.; Naciri-Bennani, H.; Rostaing, L.; Noble, J. Efficacy of Plasmapheresis in Nivolumab-Associated ANCA Glomerulonephritis: A Case Report and Pathophysiology Discussion. Case Rep. Nephrol. Dial. 2021, 11, 376–383. [Google Scholar] [CrossRef] [PubMed]
  142. Watanabe, R.; Hashimoto, M. Perspectives of JAK Inhibitors for Large Vessel Vasculitis. Front. Immunol. 2022, 13, 881705. [Google Scholar] [CrossRef] [PubMed]
  143. Zhang, H.; Watanabe, R.; Berry, G.J.; Tian, L.; Goronzy, J.J.; Weyand, C.M. Inhibition of JAK-STAT Signaling Suppresses Pathogenic Immune Responses in Medium and Large Vessel Vasculitis. Circulation 2018, 137, 1934–1948. [Google Scholar] [CrossRef] [PubMed]
  144. Gonzalez-Gay, M.A.; Matteson, E.L.; Castaneda, S. Polymyalgia rheumatica. Lancet 2017, 390, 1700–1712. [Google Scholar] [CrossRef]
  145. Watanabe, R.; Goronzy, J.J.; Berry, G.; Liao, Y.J.; Weyand, C.M. Giant Cell Arteritis: From Pathogenesis to Therapeutic Management. Curr. Treat. Options Rheumatol. 2016, 2, 126–137. [Google Scholar] [CrossRef] [Green Version]
  146. Okazaki, S.; Watanabe, R.; Kondo, H.; Kudo, M.; Harigae, H.; Fujii, H. High Relapse Rate in Patients with Polymyalgia Rheumatica despite the Combination of Immunosuppressants and Prednisolone: A Single Center Experience of 89 patients. Tohoku J. Exp. Med. 2020, 251, 125–133. [Google Scholar] [CrossRef]
  147. Salvarani, C.; Cantini, F.; Hunder, G.G. Polymyalgia rheumatica and giant-cell arteritis. Lancet 2008, 372, 234–245. [Google Scholar] [CrossRef]
  148. Betrains, A.E.; Blockmans, D.E. Immune Checkpoint Inhibitor-Associated Polymyalgia Rheumatica/Giant Cell Arteritis Occurring in a Patient After Treatment With Nivolumab. J. Clin. Rheumatol. 2021, 27, S555–S556. [Google Scholar] [CrossRef]
  149. Calabrese, C.; Cappelli, L.C.; Kostine, M.; Kirchner, E.; Braaten, T.; Calabrese, L. Polymyalgia rheumatica-like syndrome from checkpoint inhibitor therapy: Case series and systematic review of the literature. RMD Open 2019, 5, e000906. [Google Scholar] [CrossRef]
  150. Ghosh, N.; Couette, N.; van Binsbergen, W.H.; Weinmann, S.C.; Jivanelli, B.; Shea, B.; Bass, A.R.; Benesova, K.; Bingham, C.O.; Calabrese, C.; et al. Identification of outcome domains in immune checkpoint inhibitor-induced inflammatory arthritis and polymyalgia rheumatica: A scoping review by the OMERACT irAE working group. Semin. Arthritis Rheum. 2023, 58, 152110. [Google Scholar] [CrossRef]
  151. Lundberg, I.E.; Sharma, A.; Turesson, C.; Mohammad, A.J. An update on polymyalgia rheumatica. J. Intern. Med. 2022, 292, 717–732. [Google Scholar] [CrossRef]
  152. De Fremont, G.M.; Belkhir, R.; Henry, J.; Voisin, A.L.; Lambotte, O.; Besson, F.L.; Mariette, X.; Nocturne, G. Features of polymyalgia rheumatica–like syndrome after immune checkpoint inhibitor therapy. Ann. Rheum. Dis. 2022, 81, e52. [Google Scholar] [CrossRef] [Green Version]
  153. Van Der Geest, K.S.M.; Sandovici, M.; Rutgers, A.; Hiltermann, T.J.N.; Oosting, S.F.; Slart, R.H.J.; Brouwer, E. Management of immune checkpoint inhibitor-induced polymyalgia rheumatica. Ann. Rheum. Dis. 2022, 81, e263. [Google Scholar] [CrossRef]
  154. Van der Geest, K.S.M.; Sandovici, M.; Rutgers, A.; Hiltermann, T.J.N.; Oosting, S.F.; Slart, R.; Brouwer, E. Imaging in immune checkpoint inhibitor-induced polymyalgia rheumatica. Ann. Rheum. Dis. 2022, 81, e210. [Google Scholar] [CrossRef] [Green Version]
  155. Dasgupta, B.; Cimmino, M.; Maradit-Kremers, H.; Schmidt, W.; Schirmer, M.; Salvarani, C.; Bachta, A.; Dejaco, C.; Duftner, C.; Jensen, H.S.; et al. 2012 provisional classification criteria for polymyalgia rheumatica: A European League Against Rheumatism/American College of Rheumatology collaborative initiative. Ann. Rheum. Dis. 2012, 71, 484–492. [Google Scholar] [CrossRef] [Green Version]
  156. Moreira, A.; Loquai, C.; Pföhler, C.; Kähler, K.C.; Knauss, S.; Heppt, M.V.; Gutzmer, R.; Dimitriou, F.; Meier, F.; Mitzel-Rink, H.; et al. Myositis and neuromuscular side-effects induced by immune checkpoint inhibitors. Eur J Cancer 2019, 106, 12–23. [Google Scholar] [CrossRef]
  157. Fox, R.I. Sjogren’s syndrome. Lancet 2005, 366, 321–331. [Google Scholar] [CrossRef]
  158. Burbelo, P.D.; Ferre, E.M.N.; Chaturvedi, A.; Chiorini, J.A.; Alevizos, I.; Lionakis, M.S.; Warner, B.M. Profiling Autoantibodies against Salivary Proteins in Sicca Conditions. J. Dent. Res. 2019, 98, 772–778. [Google Scholar] [CrossRef]
  159. Calabrese, C.; Kirchner, E.; Kontzias, A.; Velcheti, V.; Calabrese, L.H. Rheumatic immune-related adverse events of checkpoint therapy for cancer: Case series of a new nosological entity. RMD Open 2017, 3, e000412. [Google Scholar] [CrossRef] [Green Version]
  160. Mavragani, C.P.; Moutsopoulos, H.M. Sicca syndrome following immune checkpoint inhibition. Clin. Immunol. 2020, 217, 108497. [Google Scholar] [CrossRef]
  161. Warner, B.M.; Baer, A.N.; Lipson, E.J.; Allen, C.; Hinrichs, C.; Rajan, A.; Pelayo, E.; Beach, M.; Gulley, J.L.; Madan, R.A.; et al. Sicca Syndrome Associated with Immune Checkpoint Inhibitor Therapy. Oncologist 2019, 24, 1259–1269. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  162. Pringle, S.; Wang, X.; Vissink, A.; Bootsma, H.; Kroese, F.G.M. Checkpoint inhibition-induced sicca: A type II interferonopathy? Clin. Exp. Rheumatol. 2020, 38 (Suppl. S126), 253–260. [Google Scholar] [PubMed]
  163. Jfri, A.; Leung, B.; Said, J.T.; Semenov, Y.; Le Boeuf, N.R. Prevalence of Inverse Psoriasis Subtype with Immune Checkpoint Inhibitors. Immunother. Adv. 2022, 2, ltac016. [Google Scholar] [CrossRef] [PubMed]
  164. Johnson, D.H.; Patel, A.B.; Uemura, M.I.; Trinh, V.A.; Jackson, N.; Zobniw, C.M.; Tetzlaff, M.T.; Hwu, P.; Curry, J.L.; Diab, A. IL17A Blockade Successfully Treated Psoriasiform Dermatologic Toxicity from Immunotherapy. Cancer Immunol. Res. 2019, 7, 860–865. [Google Scholar] [CrossRef] [PubMed]
  165. Ibarguren, A.M.; Enrique, E.A.; Diana, P.L.; Ana, C.; Pedro, H.P. Apremilast for immune checkpoint inhibitor-induced psoriasis: A case series. JAAD Case Rep. 2021, 11, 84–89. [Google Scholar] [CrossRef]
  166. Nigro, O.; Pinotti, G.; Gueli, R.; Grigioni, E.; De Santis, M.; Ceribelli, A.; Selmi, C. Psoriatic arthritis induced by anti-PD1 and treated with apremilast: A case report and review of the literature. Immunotherapy 2020, 12, 549–554. [Google Scholar] [CrossRef]
  167. Suzuki, M.; Matsumoto, S.; Takeda, Y.; Sugiyama, H. Systemic Psoriasiform Dermatitis Appeared after the Administration of Pembrolizumab. Intern. Med. 2020, 59, 871–872. [Google Scholar] [CrossRef] [Green Version]
  168. Halle, B.R.; Warner, A.B.; Zaman, F.Y.; Haydon, A.; Bhave, P.; Dewan, A.K.; Ye, F.; Irlmeier, R.; Mehta, P.; Kurtansky, N.R.; et al. Immune checkpoint inhibitors in patients with pre-existing psoriasis: Safety and efficacy. J. Immunother. Cancer 2021, 9, e003066. [Google Scholar] [CrossRef]
  169. Yu, Y.; Zhou, Y.; Zhang, X.; Tan, K.; Zheng, J.; Li, J.; Cui, H. Immune Checkpoint Inhibitors in the Treatment of Patients With Cancer and Preexisting Psoriasis: A Systematic Review and Meta-Analysis of Observational Studies. Front. Oncol. 2022, 12, 934093. [Google Scholar] [CrossRef]
  170. Barbosa, N.S.; Wetter, D.A.; Wieland, C.N.; Shenoy, N.K.; Markovic, S.N.; Thanarajasingam, U. Scleroderma Induced by Pembrolizumab: A Case Series. Mayo Clin. Proc. 2017, 92, 1158–1163. [Google Scholar] [CrossRef] [Green Version]
  171. Terrier, B.; Humbert, S.; Preta, L.-H.; Delage, L.; Razanamahery, J.; Laurent-Roussel, S.; Mestiri, R.; Beaudeau, L.; Legendre, P.; Goupil, F.; et al. Risk of scleroderma according to the type of immune checkpoint inhibitors. Autoimmun. Rev. 2020, 19, 102596. [Google Scholar] [CrossRef]
  172. Arnaud, L.; Mertz, P.; Gavand, P.-E.; Martin, T.; Chasset, F.; Tebacher-Alt, M.; Lambert, A.; Muller, C.; Sibilia, J.; Lebrun-Vignes, B.; et al. Drug-induced systemic lupus: Revisiting the ever-changing spectrum of the disease using the WHO pharmacovigilance database. Ann. Rheum. Dis. 2019, 78, 504–508. [Google Scholar] [CrossRef]
  173. Michot, J.-M.; Fusellier, M.; Champiat, S.; Velter, C.; Baldini, C.; Voisin, A.-L.; Danlos, F.-X.; El Dakdouki, Y.; Annereau, M.; Mariette, X.; et al. Drug-induced lupus erythematosus following immunotherapy with anti-programmed death-(ligand) 1. Ann. Rheum. Dis. 2019, 78, e67. [Google Scholar] [CrossRef] [Green Version]
  174. Raschi, E.; Antonazzo, I.C.; Poluzzi, E.; De Ponti, F. Drug-induced systemic lupus erythematosus: Should immune checkpoint inhibitors be added to the evolving list? Ann. Rheum. Dis. 2021, 80, e120. [Google Scholar] [CrossRef] [Green Version]
  175. Fadel, F.; El Karoui, K.; Knebelmann, B. Anti-CTLA4 antibody-induced lupus nephritis. N. Engl. J. Med. 2009, 361, 211–212. [Google Scholar] [CrossRef]
  176. Campochiaro, C.; Farina, N.; Tomelleri, A.; Ferrara, R.; Viola, S.; Lazzari, C.; De Luca, G.; Raggi, D.; Bulotta, A.; Matucci-Cerinic, M.; et al. Autoantibody positivity predicts severity of rheumatic immune-related adverse events to immune-checkpoint inhibitors. Eur. J. Intern. Med. 2022, 103, 95–99. [Google Scholar] [CrossRef]
  177. Granito, A.; Muratori, L.; Tovoli, F.; Muratori, P. Diagnostic role of anti-dsDNA antibodies: Do not forget autoimmune hepatitis. Nat. Rev. Rheumatol. 2021, 17, 244. [Google Scholar] [CrossRef]
  178. Granito, A.; Muratori, P.; Muratori, L.; Pappas, G.; Cassani, F.; Worthington, J.; Guidi, M.; Ferri, S.; De Molo, C.; Lenzi, M.; et al. Antinuclear antibodies giving the ‘multiple nuclear dots’ or the ‘rim-like/membranous’ patterns: Diagnostic accuracy for primary biliary cirrhosis. Aliment. Pharmacol. Ther. 2006, 24, 1575–1583. [Google Scholar] [CrossRef]
  179. Dimitriou, F.; Hogan, S.; Menzies, A.M.; Dummer, R.; Long, G.V. Interleukin-6 blockade for prophylaxis and management of immune-related adverse events in cancer immunotherapy. Eur. J. Cancer 2021, 157, 214–224. [Google Scholar] [CrossRef]
  180. Perez-Ruiz, E.; Minute, L.; Otano, I.; Alvarez, M.; Ochoa, M.C.; Belsue, V.; De Andrea, C.; Rodriguez-Ruiz, M.E.; Perez-Gracia, J.L.; Marquez-Rodas, I.; et al. Prophylactic TNF blockade uncouples efficacy and toxicity in dual CTLA-4 and PD-1 immunotherapy. Nature 2019, 569, 428–432. [Google Scholar] [CrossRef]
Figure 1. Immune-related adverse events affect multiple organs. Immune checkpoint inhibitors can cause side effects called immune-related adverse events (irAEs), which involve hypophysitis, meningitis, thyroiditis, uveitis, pneumonitis, myocarditis, pancreatitis (diabetes mellites), hepatitis, erythema, nephropathy, adrenal insufficiency, thrombocytopenia, colitis, thrombosis, and other manifestations. Colitis, hypophysitis, and skin rash occur more frequently with anti-cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) antibodies, whereas pneumonitis, hypothyroidism, and vitiligo are more common with anti-programmed cell death protein-1 (PD-1) and anti-programmed cell death ligand-1 (PD-L1) antibodies.
Figure 1. Immune-related adverse events affect multiple organs. Immune checkpoint inhibitors can cause side effects called immune-related adverse events (irAEs), which involve hypophysitis, meningitis, thyroiditis, uveitis, pneumonitis, myocarditis, pancreatitis (diabetes mellites), hepatitis, erythema, nephropathy, adrenal insufficiency, thrombocytopenia, colitis, thrombosis, and other manifestations. Colitis, hypophysitis, and skin rash occur more frequently with anti-cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) antibodies, whereas pneumonitis, hypothyroidism, and vitiligo are more common with anti-programmed cell death protein-1 (PD-1) and anti-programmed cell death ligand-1 (PD-L1) antibodies.
Ijms 24 05643 g001
Figure 2. Rheumatic immune-related adverse events show diverse pathologies. Immune checkpoint inhibitors sometimes cause rheumatic-disease-like symptoms called rheumatic immune-related adverse events (irAEs), which involve arthritis, myositis, myalgia, sicca symptoms, a rash, skin sclerosis, nephritis, and vasculitis. Autoantibody production may also be increased. Rheumatic irAEs more commonly occur with anti-programmed cell death protein-1 (PD-1) and anti-programmed cell death ligand-1 (PD-L1) antibodies than anti-cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) antibodies.
Figure 2. Rheumatic immune-related adverse events show diverse pathologies. Immune checkpoint inhibitors sometimes cause rheumatic-disease-like symptoms called rheumatic immune-related adverse events (irAEs), which involve arthritis, myositis, myalgia, sicca symptoms, a rash, skin sclerosis, nephritis, and vasculitis. Autoantibody production may also be increased. Rheumatic irAEs more commonly occur with anti-programmed cell death protein-1 (PD-1) and anti-programmed cell death ligand-1 (PD-L1) antibodies than anti-cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) antibodies.
Ijms 24 05643 g002
Figure 3. General management of rheumatic immune-related adverse events (irAEs) due to cancer immunotherapy. Consultation with oncologists is required to decide whether to continue or terminate immune checkpoint inhibitors. If patients are refractory to nonsteroidal anti-inflammatory drugs, systemic glucocorticoids should be considered. Based on the severity, conventional synthetic disease-modifying antirheumatic drugs (DMARDs) and biological DMARDs, such as anti-tumor necrosis factor (TNF) or interleukin (IL)-6 inhibitor, should be considered. Targeted synthetic DMARDs, such as Janus kinase (JAK) inhibitors, are normally avoided because of the risk of exacerbating malignancies.
Figure 3. General management of rheumatic immune-related adverse events (irAEs) due to cancer immunotherapy. Consultation with oncologists is required to decide whether to continue or terminate immune checkpoint inhibitors. If patients are refractory to nonsteroidal anti-inflammatory drugs, systemic glucocorticoids should be considered. Based on the severity, conventional synthetic disease-modifying antirheumatic drugs (DMARDs) and biological DMARDs, such as anti-tumor necrosis factor (TNF) or interleukin (IL)-6 inhibitor, should be considered. Targeted synthetic DMARDs, such as Janus kinase (JAK) inhibitors, are normally avoided because of the risk of exacerbating malignancies.
Ijms 24 05643 g003
Table 1. Brief summary of rheumatic immune-related adverse events (irAEs).
Table 1. Brief summary of rheumatic immune-related adverse events (irAEs).
IncidenceTreatment
ArthritisUp to 5%1. NSAIDs, 2. Glucocorticoids, 3. DMARDs
Myositis<1%1. Glucocorticoids, 2. DMARDs, 3. PE, 4. IVIg
Vasculitis<1%1. Glucocorticoids, 2. CYC, 3. RTX, 4. TCZ
MyalgiaUp to 5%1. Glucocorticoids, 2. DMARDs, 3. TCZ
Sicca symptomRare1. Glucocorticoids
Skin rash (psoriasis)Up to 3%1. Glucocorticoids, 2. DMARDs
Skin sclerosisRare1. Glucocorticoids, 2. CYC, 3. DMARDs
Lupus-like diseaseRare1. Glucocorticoids, 2. CYC
Treatment should be decided based on the severity. CYC: cyclophosphamide, DMARDs: disease-modifying antirheumatic drugs, IVIg: intravenous immunoglobulin, NSAIDs: nonsteroidal anti-inflammatory drugs, PE: plasma exchange, RTX: rituximab, TCZ: tocilizumab.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Dang, Q.M.; Watanabe, R.; Shiomi, M.; Fukumoto, K.; Nobashi, T.W.; Okano, T.; Yamada, S.; Hashimoto, M. Rheumatic Immune-Related Adverse Events due to Immune Checkpoint Inhibitors—A 2023 Update. Int. J. Mol. Sci. 2023, 24, 5643. https://doi.org/10.3390/ijms24065643

AMA Style

Dang QM, Watanabe R, Shiomi M, Fukumoto K, Nobashi TW, Okano T, Yamada S, Hashimoto M. Rheumatic Immune-Related Adverse Events due to Immune Checkpoint Inhibitors—A 2023 Update. International Journal of Molecular Sciences. 2023; 24(6):5643. https://doi.org/10.3390/ijms24065643

Chicago/Turabian Style

Dang, Quang Minh, Ryu Watanabe, Mayu Shiomi, Kazuo Fukumoto, Tomomi W. Nobashi, Tadashi Okano, Shinsuke Yamada, and Motomu Hashimoto. 2023. "Rheumatic Immune-Related Adverse Events due to Immune Checkpoint Inhibitors—A 2023 Update" International Journal of Molecular Sciences 24, no. 6: 5643. https://doi.org/10.3390/ijms24065643

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop