Abstract
The emergence of immune checkpoint inhibitors (ICIs) has heralded a transformative era in the therapeutic landscape of non‐small cell lung cancer (NSCLC). While ICIs have demonstrated clinical efficacy in a portion of patients with NSCLC, these treatments concurrently precipitate a spectrum of immune‐related adverse events (irAEs), encompassing mild to severe manifestations, collectively posing a risk of significant organ damage. Consequently, there exists an imperative to augment our comprehension of the pathophysiological underpinnings of irAEs and to formulate more efficacious preventive and ameliorative strategies. In this comprehensive review, we delineate the clinical presentation of organ‐specific irAEs in patients with NSCLC and provide an in‐depth analysis of recent advancements in understanding the mechanisms driving ICI‐induced toxicity. Furthermore, we discuss potential strategies and targets for ameliorating these irAEs. Ultimately, this review aims to furnish valuable insights to guide further research endeavours in the context of irAEs in NSCLC patients.
Keywords: immune checkpoint inhibitors, immune‐related adverse events, mechanism, non‐small cell lung cancer
Immunotherapy not only revolutionises lung cancer treatment but also poses serious toxicity risks.
Elucidating irAE mechanisms in NSCLC: T/B‐cell dysregulation, cytokine imbalances, autoantibody production, genetic predispositions and gut microbiome alterations.
Highlighting precise irAE interventions in NSCLC: corticosteroids, targeted immunosuppressants, monoclonal antibodies targeting lymphocytes, cytokine inhibitors and signalling pathway modulators.

1. BACKGROUND
Non‐small cell lung cancer (NSCLC), accounting for 80%–85% of lung cancer cases, often reaches an advanced diagnosis stage, resulting in a bleak prognosis for approximately 70% of patients. 1 The 5‐year survival likelihood in NSCLC patients with advanced stage barely exceeds 3%. 1 In recent years, immune checkpoint inhibitors (ICIs) such as cytotoxic T‐lymphocyte antigen 4 (CTLA‐4), programmed death‐1 (PD‐1) and programmed death‐ligand 1 (PD‐L1) inhibitors have revolutionised the treatment landscape of NSCLC. 2 , 3 , 4 , 5 , 6 For example, NSCLC patients treated with atezolizumab in the IMpower110 trial exhibited improved median overall survival (OS) relative to chemotherapy (20.2 vs. 13.1 months). 7
However, immunotherapy can induce diverse adverse effects, referred to as immune‐related adverse events (irAEs), that resemble autoimmune pathologies. 8 As demonstrated in numerous clinical trials, irAEs induced by immunotherapy primarily manifest as impairments in the cardiac, respiratory, gastrointestinal, skin and neurological systems. 8 , 9 , 10 In several clinical trials, 45%–60% of NSCLC patients treated with atezolizumab experienced irAEs. 11 , 12 , 13 A systematic analysis encompassing 16 clinical studies reported an overall irAE incidence of 22%, with severe irAEs (grade 3–4) affecting 4% of NSCLC patients under anti‐PD‐1/PD‐L1 treatment. 9 While most cases can be effectively managed, a subset may present with moderate to severe toxicities, thereby severely compromising organ functionality. 14
Given the frequency and severity of irAEs, understanding their underlying mechanisms assumes paramount importance in devising effective prevention and treatment strategies. 15 Yet, the precise mechanisms governing irAEs remains to be fully elucidated, and so preventative action has yet to be consistently applied in clinical practice. 15 Broadly, many irAEs are identified as autoimmune diseases triggered by ICI‐activated CD8+ cytotoxic T cells, and some involve activated B cells and pathogenic antibody production. 16 , 17 Additionally, there is growing interest in exploring the involvement of T follicular helper (Tfh) cells in promoting abnormal B‐cell reactions, potentially disrupting immune tolerance in peripheral tissues. 18 , 19 Consequently, there is a pressing need to delve into the underlying mechanisms of ICI‐induced toxicity and formulate mechanism‐based strategies to mitigate its occurrence. 15 Regrettably, a standardised strategy for patient stratification based on their risk of experiencing toxicity is lacking in the current clinical landscape. 17 This review endeavours to address these challenges by consolidating the existing knowledge on irAE mechanisms in NSCLC and mitigation strategies, thus serving as a valuable resource for clinicians seeking to enhance irAEs management.
2. CLINICAL MANIFESTATION OF ICI‐INDUCED IrAEs IN NSCLC
IrAEs from NSCLC immunotherapy impact various organs, such as the heart, lungs, skin, digestive system, nervous system and muscles (Figure 1). 15 , 20 , 21 A recent systematic review reported that moderate to severe chronic non‐endocrine irAEs could persist for a median (range) of 180 (84–2370) days, with 52% of patients experiencing chronic irAEs that persisted for over 6 months. 20 Additionally, research has emphasised that the incidence of irAEs within the initial year of anti‐PD‐1 treatment is closely associated with prolonged toxicity extending past 1 year in individuals with advanced NSCLC. 21 These findings underscore the variability in terms of the occurrence, severity and timing of these toxicities. 20 , 21 It is noteworthy that certain acute irAEs occur more frequently with dual immunotherapy than with monotherapy. This suggests that the type and severity of irAEs depend on the affected organ and the specific ICI used 22 (Figure 1 and Table 1).
FIGURE 1.

Clinical manifestation and occurrence of immune‐related adverse events (irAEs) in non‐small cell lung cancer (NSCLC).
TABLE 1.
Clinical incidence of organ‐specific immune‐related adverse events (irAEs) in non‐small cell lung cancer (NSCLC) patients receiving immune checkpoint inhibitors (ICIs).
| Incidence of any‐grade irAEs (%) | ||||
|---|---|---|---|---|
| Specific irAEs | PD‐1 inhibitor | PD‐L1 inhibitor | CTLA‐4 inhibitor | Combination |
| Cardiovascular | ||||
| Myocarditis | (a) Pembrolizumab (<1%) 42 , 43 , 44 | (a) Atezolizumab (1%–6.7%) 46 , 47 | (a) IPI + NIVO (≤1%) 62 | |
| Pulmonary | ||||
| Pneumonia |
(a) Cemiplimab (1%–15%) 63 , 64 , 65 |
(a) Tremelimumab (1%–2%) 31 (b) IPI (2%) 71 |
(a) Anti‐TIGIT (tiragolumab) + atezolizumab (2%) 48 (b) Anti‐TIGIT (vibostolimab) + pembrolizumab (6%) 52 |
|
| Interstitial lung disease | (a) NIVO (1%–3%) 38 , 40 | (a) Atezolizumab (1%) 45 | (a) IPI (1%) 71 | (a) IPI + NIVO (1%–2%) 62 , 72 |
| Dermatologic | ||||
| Rash |
(a) NIVO (5.7%–27%) 38 , 39 , 40 (b) Pembrolizumab (7%–22%) 42 , 44 , 73 (c) Cemiplimab (5%) 64 |
(a) Anti‐TIGIT (tiragolumab) + atezolizumab (27%) 48 |
||
| Endocrine | ||||
| Hypo‐thyroidism |
(b) Pembrolizumab (6.7%–28.8%) 42 , 44 , 73 , 76 (c) Cemiplimab (10%) 41 |
Tremelimumab (4%) 75 |
(a) Anti‐TIGIT (tiragolumab) + atezolizumab (10%) 48 |
|
| Hyper‐thyroidism |
(a) Atezolizumab (2.8%–4.1%) 29 |
(a) IPI (2%) 71 | (a) IPI + NIVO (8.7%) 72 | |
| Gastrointestinal | ||||
| Nausea |
(a) Pembrolizumab (2%–17.2%) 42 , 44 (c) Cemiplimab (3%) 64 |
(a) Atezolizumab (7.7%–14.2%) 11 , 28 |
(b) Tremelimumab (10%) 31 |
(a) Durvalumab + tremelimumab (1.2%–18%) 31 , 82 |
| Diarrhoea |
(a) Pembrolizumab (48%) 83 |
(a) IPI (27%–30%) 71 , 81 | (a) IPI + NIVO (6.4%–20%) 49 | |
| Colitis |
(a) NIVO (2%) 38 (b) Pembrolizumab (1%–3.9%) 44 , 73 (c) Cemiplimab (<4%) 64 |
(a) Atezolizumab (2.1%) 7 |
(a) Tremelimumab (8.8%–19%) 31 , 75 (b) IPI (4%) 71 |
(a) Anti‐TIGIT (tiragolumab) + atezolizumab (4%) 48 (b) IPI + NIVO (1%–6%) 62 , 86 (c) Durvalumab + tremelimumab (1.8%) 31 |
| Liver | ||||
| Hepatitis |
(a) Pembrolizumab (2.1%–17.2%) 44 , 87 (b) NIVO (2%–10%) 38 , 39 , 40 (c) Cemiplimab (<2%) 64 |
(a) Atezolizumab (1%–23%) 28 , 30 , 45 (b) Durvalumab (13%) 33 |
(a) Anti‐TIGIT (tiragolumab) + atezolizumab (5%) 48 |
|
| Renal | ||||
| Nephritis/renal injury |
(a) Pembrolizumab (.4%–.6%) 42 , 73 (b) Cemiplimab (1%) 41 |
(a) Durvalumab (.4%) 77 | (a) IPI (<1%) 71 |
(a) IPI + pembrolizumab (1%) 5 (b) IPI + NIVO (1%) 5 |
| Neurologic | ||||
| Encephalitis | (a) NIVO (< 1%) 38 | (a) Avelumab (.3%) 88 | ||
| Musculoskeletal | ||||
| Arthralgia |
(a) Atezolizumab (2%–16.8%) 30 , 45 , 46 (b) Durvalumab (16.7%) 32 |
(a) IPI (7%–14%) 71 , 81 |
(a) Anti‐TIGIT (tiragolumab) + pembrolizumab (12%) 52 (b) Anti‐TIGIT (tiragolumab) + atezolizumab (16%) 48 |
|
| Eyes | ||||
| Uveitis | (a) Pembrolizumab (.6%) 73 | (a) IPI (1.5%) 90 | (a) IPI + pembrolizumab (9%) 4 | |
Abbreviations: CTLA‐4, cytotoxic T‐lymphocyte antigen 4; IPI, ipilimumab; NIVO, nivolumab; PD‐1, programmed death‐1; PD‐L1, programmed death‐ligand 1; TIGIT, T‐cell immunoreceptor with immunoglobulin and immunoreceptor tyrosine‐based inhibitory motif domains.
2.1. irAEs induced by CTLA‐4, PD‐1 and PD‐L1 inhibitors
The primary categories of ICIs encompass CTLA‐4, PD‐1 and PD‐L1 inhibitors. 23 CTLA‐4 inhibitors empower T cells to enhance their activity and effectively eliminate malignant cells. 23 , 24 Nevertheless, loss of function of the CTLA4 gene, whether induced by genetic knockout in vivo or inhibition in humans, is linked to widespread adverse effects related to autoimmunity across various tissues. 25 Additionally, considering significant function of PD‐1 in regulating the immune response of activated T cells, targeting PD‐1/PD‐L1 pathway may impede tumour immune evasion, but may also potentially disrupt the balance between normal tissues and cancer cells, resulting in immune imbalance. 23 , 26
In NSCLC patients treated with anti‐CTLA‐4, common irAEs include skin rashes (13.3%–41%), liver inflammation (5%–42%) and digestive problems such as diarrhoea (27%–30%), nausea (8%–18%) and colitis (8.8%–19%) (Table 1), 7 , 11 , 27 , 28 , 29 , 30 as depicted in Table 1. However, NSCLC patients on CTLA‐4 inhibitors less frequently develop checkpoint inhibitor pneumonitis (CIP) compared to those on PD‐1/PD‐L1 inhibitors, although underreporting is a concern. 7 , 31 , 32 , 33 , 34 , 35 , 36 Moreover, research from clinical trials indicates that the range of affected organ systems and the intensity of irAEs differ across therapeutic agents, especially when comparing CTLA‐4 with PD‐1/PD‐L1 inhibitors and their various combinations. 23 For instance, gastrointestinal toxicity is predominant in NSCLC patients treated with anti‐PD‐1/PD‐L1, with subsequent occurrences of endocrine (6.7%–28.8%), musculoskeletal (5.7%–26%) and pulmonary issues (4.4%–23%) 23 (Table 1). Furthermore, patients on PD‐1 inhibitors have a notably higher rate of experiencing diarrhoea, skin rashes and kidney damage than those on PD‐L1 inhibitors. 37 , 38 , 39 , 40 , 41 Conversely, myocarditis is more common in patients undergoing treatment with anti‐PD‐L1 (1%–6.7%) compared to those on anti‐PD‐1 (<1%) 42 , 43 , 44 , 45 , 46 , 47 (Table 1).
2.2. irAEs induced by combination regimens of ICIs
Compared to mono‐immunotherapy or chemotherapy, NSCLC treatment using a combination of ICIs correlates with an increased frequency of irAEs in patients; however, it offers improved survival outcomes 4 . The incidence of endocrine toxicities (8.7%–16%) is elevated in combined immunotherapy compared to monotherapy, 48 , 49 , 50 as shown in Table 1. Additionally, ipilimumab plus pembrolizumab treatment is associated with a higher occurrence of pneumonia (7%–12.1%) compared to pembrolizumab alone 4 , 51 (Table 1). Conversely, when PD‐1/PD‐L1 inhibitors are used in combination with other types of ICIs for NSCLC treatment, there is an increased irAEs impacting the skin, digestive system and musculoskeletal system, compared to when PD‐1/PD‐L1 and CTLA‐4 inhibitors are used together 4 , 7 , 31 , 48 , 52 (Table 1).
The biological hypotheses explaining how different ICIs cause organ‐specific irAEs are influenced by the affected organ. In the gastrointestinal system, CTLA‐4 is more crucial for regulating gut equilibrium than PD‐1/PD‐L1. 53 CTLA‐4‐targeting monoclonal antibodies (MoAbs) activate the T cells in the gut, leading to colitis characterised by elevated CD4+ effector and regulatory T cells (Tregs), accompanied by significant changes in Treg gene expression. 54 Conversely, while PD‐1/PD‐L1 MoAbs also induce gastrointestinal irAEs, they are generally less severe. 53 Cardiac irAEs stem from disrupted immune balance in the heart, particularly with ICI combinations, with a notable increase in cytotoxic CD8+ T cells and T‐cell receptor (TCR) rearrangement, hint at α‐myosin as a potential driver of myocardial damage. 55 , 56 CIP is more likely with PD‐1 inhibitors, which increase cytokine production and CD4+ T‐cell growth compared to CTLA‐4 or PD‐L1 inhibitors, possibly owing to enhanced interaction of repulsive guidance molecule b and PD‐L2 in lung cells. 57 , 58 As for dermatologic irAEs, lichenoid dermatitis is often associated with PD‐1/PD‐L1 MoAbs, owing mainly to the activation of CD4+ T cells. 59 Conversely, alopecia areata is linked to CTLA‐4 gene variants, while CTLA‐4 IgG supplementation can avert its development in mice. 60 For endocrine‐related irAEs, thyroiditis is often driven by anti‐PD‐1‐activated CD4+ T cells. However, the specific impact of anti‐CTLA‐4 on thyroid function remains unclear. 61 Therefore, organ‐specific irAEs appear to be influenced by distinct molecular mechanisms of various MoAbs, underscoring the complexity and individual variability of irAEs.
3. MECHANISMS OF ICI‐INDUCED irAEs IN NSCLC
Currently, accumulating evidence suggests that checkpoint inhibitors could specifically reactivate T cells and self‐antigen‐mediated cellular immunity in normal tissues, which could result in irAEs. 23 , 67 Other intrinsic factors could include pre‐existing autoimmunity, genetic variants and unbalanced inflammatory cytokines 91 (Figure 2). Nevertheless, the underlying mechanisms behind irAEs in NSCLC are still predominantly undetermined. 23
FIGURE 2.

Potential mechanisms involved in the occurrence of immune‐related adverse events (irAEs) in non‐small cell lung cancer (NSCLC). There are multiple potential mechanisms that could contribute to the occurrence of immune checkpoint inhibitor (ICI)‐mediated toxicity. (1) When T cells are activated by ICIs, cross‐reactivity antigenicity can occur between tumours and normal tissue affected by irAEs. Notably, patients who developed irAEs exhibited an increase in proliferative activated T cells in normal tissue. (2) ICIs can induce upregulation in synthesis of inflammatory cytokines and chemokines, and increase in the tissue‐resident populations of T helper cells (Th1/2/17) and decrease in regulatory T cells (Tregs), thereby participating in early development of irAEs. (3) Germline genetic factors including specific human leukocyte antigen (HLA) alleles and single nucleotide polymorphism (SNP) may contribute to irAEs. (4) ICI‐associated increased B‐cell clonality as well as increased detection of autoantibodies of ICIs commencement are associated with irAEs. Additionally, follicular T helper cells (Tfh) cells typically express programmed death‐1 (PD‐1) and interact with programmed death‐ligand 1 (PD‐L1)‐producing B cells in B‐cell follicle to initiate germinal centre (GC) reaction, whereby B cells undergo maturation. (5) Patients with pre‐existing or new auto‐antibodies may also be an underlying predisposition to the occurrence of irAEs. (6) ICIs can affect the intestinal microbiome profile, leading to production of proinflammatory cytokines.
3.1. T‐cell overactivation in the pathogenesis of irAEs
ICIs have been observed to potentially lead to the growth and stimulation of memory T cells in normal tissue in the development of irAEs. 15 , 23 Recent research suggests that the infiltration of tissues by active T cells might participate in the formation of irAEs in NSCLC. 15
3.1.1. Cross‐reactivity antigenicity of T cells between tumoural and healthy cells
The onset of irAEs is believed to be linked to antigen cross‐reactivity between tumoural antigens and their healthy counterparts. 92 Numerous investigations have empirically validated this observation. For example, active CD8+ T lymphocytes infiltrating healthy cardiac tissue and the consequent T‐cell‐mediated autoimmunity are strongly associated with fulminant myocarditis in melanoma and NSCLC patients receiving anti‐CTLA‐4/PD‐1 therapy, underscoring the pivotal relationship between antigen cross‐presentation and ICI‐induced myocarditis. 36 , 93 Additionally, Berner et al. 94 identified the same TCR sequences in both cancerous and normal tissues in NSCLC patients showing autoimmune skin toxicities post anti‐PD‐1 therapy. Notably, Berner et al. 95 developed a systematic approach aimed at identifying self‐antigens related to ICI responses and irAEs in NSCLC patients. Remarkably, this study found that napsin A‐specific T‐cell clones were disproportionately represented in ICI responders, lung cancer and post‐therapy inflammatory pulmonary lesions. 95 This innovative framework paves the way for the identification of irAE‐specific antigens that are potentially targetable. 95 Historical data further indicate an escalated prevalence of CD8+ T cells in lung samples from NSCLC patients who developed CIP after nivolumab, highlighting the crucial role of antigen cross‐reactivity. 96 Furthermore, Lechner et al. 97 found a clonally expanded segment of cytotoxic C‐X‐C motif chemokine receptor 6 (CXCR6)+ CD8+ T cells in thyroid tissue, a population characterised by interferon‐γ (IFN‐γ) and granzyme B upregulation and spurred on by interleukin (IL)‐21, which was indicative of ICI‐induced thyroiditis in NSCLC and other cancer types. 97 This concept, where T cells recognise tumour‐specific antigens in normal cells leading to irAEs, is reiterated in various studies covering ailments such as vitiligo, colitis and neurological conditions. 94 , 96 , 98 , 99
Furthermore, ICIs have been demonstrated to enhance the variety of T‐cell clones in both bronchoalveolar lavage fluid (BALF) and circulating blood, leading to the development of irAEs. 94 , 100 , 101 Suresh et al. 100 performed an examination of lymphocytes from BALF in patients undergoing ICIs therapy. Their findings revealed a notable increase in CD4+ T cells clones among patients with NSCLC with CIP. 100 Additionally, another study reported an elevated presence of CD8+ T cells with PD‐1, T‐cell immunoreceptor with immunoglobulin and immunoreceptor tyrosine‐based inhibitory motif domains (TIGHIT) and T cell immunoglobulin domain and mucin domain‐3 (Tim‐3) expression in BALF from NSCLC and other cancer patients with PD‐1/PD‐L1 inhibitor‐related interstitial lung disease (ILD), as opposed to other ILD types. 101 Corroborating this, circulating T cells specific to epidermal antigens were identified in autoimmune skin conditions and lung cancer in NSCLC patients post‐anti‐PD‐1 treatment, who showed skin‐related adverse effects. 94 This revelation enhances our comprehension of circulating antigen cross‐presentation and its influence on organ‐specific irAEs in NSCLC. 94 Recent findings also postulate that activated CD4+ T cells can induce inflammatory cell death, thereby managing immune‐evasive tumours. 102 The concurrent presence of activated baseline CD4 memory T‐cell clones and varied TCR, as mapped through single‐cell RNA sequencing (scRNA‐seq), is associated with the development of irAEs in melanoma patients, regardless of the specific organ systems implicated. 103 While such observations have yet to be noted in circulating blood samples from NSCLC patients, future scRNA‐seq investigations into NSCLC could potentially illuminate the indispensable role of T‐cell clones in precipitating irAEs.
3.1.2. Activation of T helper cells and Tregs involved in irAEs
Within the myeloid compartment, dendritic cells and macrophages are crucial for presenting antigens, which in turn helps activate T cells. 18 Following activation by myeloid cells in tumour‐draining lymph nodes (dLNs), CD4+ and CD8+ T cells differentiate into various tumour‐infiltrating lymphocytes (TILs), encompassing T helper (Th1/Th2/Th17) cells, Tregs and Tfh cells (CD4+ CXCR5+ PD‐1high), among others. 18 Post‐differentiation, these TILs exit dLNs, migrate to peripheral tissues and instigate a nascent antibody response. 18 Notably, these TILs, while instrumental in the anti‐tumour immune response, can manifest divergent roles in irAEs. 18
It has been suggested that anti‐PD‐1/PD‐L1 treatments bolster Th1 cell proliferation while concurrently attenuating Th2‐associated cytokine production, which collectively inhibits tumour growth. 15 , 23 Interestingly, it was observed that CD8+ Th1 cells expressing CX3CR1/CXCR3 were significantly expanded in arthritis irAEs among NSCLC and other cancer patients, implicating Th1 cells in irAE pathogenesis. 104 Subsequent research has shown that anti‐PD‐1/PD‐L1 can hinder the transformation of Th1 cells into Tregs, potentially exacerbating immune‐mediated damage. 105 Suresh et al. 100 observed a reduction in CTLA‐4+ and PD‐1+ Tregs in the BALF of CIP patients, suggesting a possible involvement of Tregs in the formation of irAEs. Despite their capacity to inhibit Th1 cell growth, Tregs fail to prevent the transformation of Th17 cells into Th1 cells. 106 Interfering with the PD‐1/PD‐L1 pathway leads to a decrease in Tregs, potentially leading to dysregulation of the Treg/Th17 cell balance, which has been implicated in various autoimmune disorders. 92 Recent scRNA‐seq studies involving patients with NSCLC have observed an elevation in CD4+ Th2 cells and CD4+ Th17 cells linked to pneumonitis and thyroiditis, respectively, highlighting their proinflammatory effects and the potential occurrence of organ‐specific toxic effects. 107 Moreover, the myeloid compartment is vital in the pathogenesis of ICI‐mediated irAEs, acting as essential antigen‐presenting cells for the activation of T cell and significantly influencing PD‐1 expression beyond T cells. 108 , 109 Anti‐PD‐1 therapy can induce alterations within the myeloid compartment, leading to a systemic accumulation of inflammatory cells across various organs and tissues. 110 , 111 This has been particularly demonstrated by the expansion of inflammatory macrophages in mouse models of ICI‐associated myocarditis. 110 , 111 Moreover, the role of macrophages in the onset of ICI‐induced diabetes underscores their substantial influence in the occurrence of irAEs, thus highlighting the extensive effects of myeloid cells in ICI‐related adverse effects. 112
3.1.3. T follicular helper cells involved in irAEs
It is crucial to acknowledge the capacity of activated CD4+ T cells to differentiate into early Tfh cells. These early Tfh cells subsequently engage in intricate interactions with antigen‐specific B cells and migrate into the follicle, where they establish the germinal centre (GC) and initiate the early extrafollicular antibody response. 18 , 19 These Tfh cells display increased expression of costimulatory receptors including inducible T cell co‐stimulator (ICOS), as well as coinhibitory receptors (such as PD‐1) on their cell surface, while also secreting IL‐21 and IL‐4, thus participating in potential pathways that are involved in the regulation of humoral immunity. 18 , 113 Consequently, as has been previously hypothesised, PD‐1 inhibitors could potentially disrupt Tfh cell function, resulting in the generation of aberrant B cells and ultimately leading to the occurrence of irAEs. 18 , 114 More recently, Lechner et al. 97 identified the infiltration of both T peripheral helper (Tph) and Tfh cells into ICI‐related thyroiditis tissue in patients with NSCLC and other types of tumours. They proposed a potential mechanism in vivo in which IL‐21+ Tfh and Tph cells might significantly contribute to ICI‐induced autoimmunity by augmenting the effector characteristics of CD8+ T cells. 97 Specifically, recombinant IL‐21 has been shown to activate CD8+ effectors expressing CXCR6, GZMB and IFN‐γ, thereby enhancing thyrotoxic activity in mice. This suggests that targeting IL‐21 signalling could potentially reduce irAEs. 97 Notably, the observed pattern of irAEs in the mouse model may not fully correspond to that observed in human patients. Thus, future endeavours should amalgamate insights from both patient‐derived irAE data and syngeneic murine tumour models to elucidate the nuanced interplay between IL‐21‐mediated autoimmunity and antitumour responses.
3.2. Unbalanced inflammatory cytokines involved in irAEs
Multiple studies have indicated that the dysregulation of cytokine secretion serves as an additional catalyst for the development of irAEs. 91 Cytokines are critical in regulating immune system functions and supporting the activities of different immune cells. 91 They regulate a variety of signalling pathways essential for T‐cell activation and the transformation of B cells into plasma cells, which are responsible for antibody production. 91 The presence of proinflammatory cytokines has been shown to initiate a systemic inflammatory response, thereby increasing the likelihood of irAEs 115 (Figure 3). Notably, elevated levels of circulating IL‐17 and IL‐6, secreted primarily by Th17 cells that suppress Treg activity, along with IL‐2, known for boosting cytotoxic CD8+ T‐cell activity, are associated with irAEs onset. 91 , 115 , 116 , 117 Lim et al. 115 observed an association between the levels of 11 cytokines including Fractalkine, granulocyte colony stimulating factor (G‐CSF), fibroblast growth factor 2 (FGF‐2), granulocyte‐macrophage colony‐stimulating factor (GM‐CSF), IFN‐α2, IL‐1a, IL‐1b, IL‐1RA, IL‐2, IL‐12p70 and IL‐13 in the blood and the emergence of irAEs in melanoma patients receiving anti‐PD‐1 treatment. This research also created a toxicity index using these 11 cytokines to identify patients at increased risk of experiencing irAEs. 115 Chen et al. 118 conducted scRNA‐seq on lung adenocarcinoma patients undergoing ICI treatments, wherein they observed a notable elevation in the levels of circulating tumour necrosis factor (TNF) protein among patients experiencing irAEs, a trend not observed in those responding to ICI treatments. 118 Another study, primarily involving NSCLC patients, showed a notable link between C‐X‐C motif chemokine ligands (CXCLs) and irAEs incidence. Specifically, CXCL9/10/11 were identified as binding to CXCR3, thereby stimulating T‐cell activation and contributing to the progression of irAEs in NSCLC. 119
FIGURE 3.

Unbalanced inflammatory cytokines in immune‐related adverse events (irAEs). Immunotherapy may lead to cytokine dysregulation, causing systemic inflammatory responses that further promote organ‐specific irAEs. (1) Circulating interleukin (IL)‐17 and IL‐6 secreted by T helper 17 cells (Th17) cells could inhibit the activity of regulatory T cells (Tregs), thereby reducing the production of anti‐inflammatory factors such as IL‐10, IL‐35 and transforming growth factor‐beta (TGF‐β), which are associated with the occurrence of irAEs. (2) Cytokines like IL‐6, IL‐23 and TGF‐β stimulate Th17 to secrete IL‐17 and IL‐6, contributing to irAEs. (3) C‐X‐C motif chemokine ligand 9/10/11 (CXCL9/10/11) binds to C‐X‐C motif chemokine receptor 3 (CXCR3) on cytotoxic T cells, stimulating their activation and promoting CXCR6, granzyme B (GZMB) and interferon‐γ (IFN‐γ) secretion. This leads to the progression of irAEs in non‐small cell lung cancer (NSCLC). (4) Other cytokines such as IL‐2 and IL‐4 stimulate CD4+Th2 cells to secrete proinflammatory factors, including IL‐21.
Regarding organ‐specific irAEs, Lechner et al. 97 discovered IL‐21, secreted by CD4+ Tfh and Tph cells, significantly contributes to the emergence of both ICI‐induced thyroiditis and Hashimoto's thyroiditis in NSCLC patients. Consequently, this finding could elucidate why patients with existing thyroid autoantibodies are more prone to developing thyroid irAEs when undergoing ICI treatments. 120 Furthermore, various research has shown that increased levels of serum inflammatory indicators such as IL‐6, IL‐10, IL‐17, IL‐35 and C‐reactive protein correlate with a heightened risk of severe irAEs in NSCLC. 121 , 122 , 123 Additionally, apart from being present in tissue and serum, certain proinflammatory and chemotactic cytokines linked to CIP are detectable in the BALF of NSCLC patients. 100 , 122 Together, these studies highlight the crucial role of cytokines in the manifestation of irAEs in NSCLC.
3.3. Genetic variants associated with irAEs development
Germline genetic factors exert significant influence on immune homeostasis and our immunological status. 17 , 124 The variability in the onset and severity of irAEs observed among patients receiving similar ICI agents can be partially attributed to germline genetic variation in immune function. 17 , 124 Recent research has demonstrated a correlation between polygenic germline risk for autoimmune conditions and the development of cutaneous and thyroid irAEs in NSCLC. 125 , 126 These results supported that germline genetic factors may contribute to the occurrence of irAEs. However, the extent to which individual genetic variants are associated with irAEs in NSCLC continues to be uncertain. For instance, a study by Groha et al. 127 used a genome‐wide association approach to investigate irAEs, revealing a significant association between a single nucleotide polymorphism known as rs16906115, located within the IL7 gene, and the development of any grade of irAE toxicities. 127 This correlation was noted in 1751 patients with 12 distinct types of cancer, NSCLC included, all of whom received anti‐PD1/PD‐L1 treatment. 127 Notably, rs16906115 represents the first genetic variant to be identified as being associated with irAEs using large sample sizes and validation cohorts. 127 This study primarily focused on patients who were administered PD1/PD‐L1 treatment as single medications. Additionally, another pan‐tumour genome‐wide association study investigating irAEs has identified rs469490 as being associated with any grade of irAEs induced by nivolumab, and rs8023690 as potentially predictive of hypothyroidism in subgroup analysis. 128 However, it is still unknown whether specific genetic variants are also linked to anti‐CTLA‐4‐related irAEs in NSCLC.
Human leukocyte antigen class II (HLA‐II) is vital in presenting tumour antigens to CD8+ and CD4+ T cells, thereby aiding in the successful eradication of cancer cells. 129 Interestingly, recent research has shown that genotyping of HLA antigens is a contributing factor in the emergence of particular irAEs. 130 For example, in a novel prospective study, the links between HLA‐DRB111:01 and pruritus, as well as HLA‐DQB103:01 and colitis, were explored in NSCLC and melanoma patients undergoing ICI therapy. 130 Additionally, other more focused studies have investigated the connections between particular irAEs and HLA variants in a broad tumour spectrum, including NSCLC, such as HLA‐DR15 and hypophysitis, 131 HLA‐DRB1 and arthritis, 132 and HLA‐DR11 and pneumonitis. 133 These results bolster the theory that genetic variations involved in immune regulation might significantly influence the risk of irAEs in NSCLC. Further investigation through association studies is necessary to enhance the utilisation of genetic factors involved in irAEs in NSCLC.
3.4. B‐cell dysregulation in ICI‐induced toxicity
Emerging evidence increasingly underscores the pivotal role of B‐cell dysregulation in mediating both the effectiveness of ICIs and the frequency of irAEs. 134 However, the exact mechanism by which B cells modulate or mediate severe irAEs remains elusive in NSCLC. 134 B‐regulatory cells (Bregs) are important immune modulators, preventing excessive inflammation and maintaining immune homeostasis via cytokine production, including IL‐10, IL‐35 and transforming growth factor beta. 18 Rosser et al. 135 have identified a decrease in the number and function of Bregs in various irAEs, including autoimmune diseases, chronic infections and cancers. 135 More recently, subsequent research from Patel et al. 136 showed a correlation between IL‐10 produced by Bregs and the occurrence of severe irAEs in NSCLC patients treated with ICIs.
Nevertheless, certain Breg subsets, such as the PD‐L1high Bregs, can suppress immune responses independently of IL‐10. 113 These PD‐L1high Bregs have been described as suppressing B cell subsets via PD‐L1 independently of IL‐10 by impeding Tfh cell differentiation, thereby inhibiting the T‐cell response. 113 Tfh cells commonly exhibit PD‐1 and engage with PD‐L1‐expressing B cells in the B‐cell follicle, instigating the GC reaction, leading to B‐cell maturation (Figure 2). Previous evidence has shown that increased presence of Tfh cells and PD‐L1high Bregs is a notable feature in autoimmune disorders, especially rheumatic diseases, resulting in heightened proliferation and cytokine output by CD8+ T cells. 18 , 137 In a recent longitudinal analysis, four phenotypes of PD‐L1high Breg populations were notably prevalent in non‐toxic NSCLC patients treated with anti‐PD‐1/PD‐L1 through functional ex vivo and deep phenotyping mass cytometric assays. 135 These findings suggest that NSCLC patients possessing inherent functional impairments in their Breg array and lacking certain peripheral Breg phenotypes may be more inclined to experience severe autoimmune reactions.
3.5. Autoantibody production and pre‐existing autoantibodies
An expanding range of studies indicates that the emergence of irAEs in NSCLC may be associated with the presence of both pre‐existing and new autoantibodies in immune responses. 92 The potential mechanism by which ICIs facilitate the production of autoantibodies could entail interactions between B and Tfh cells. The disruption of B‐cell homeostasis, particularly Bregs, by ICIs can lead to autoantibody generation, as evidenced in PD‐1 knockout mice and ICI‐treated patients. 92 An increase in circulating plasmablasts and associated irAEs has been observed in patients receiving anti‐CTLA‐4 or combination therapies. 92 , 138 DeFalco et al. 138 provided evidence of somatic hypermutation and clonal expansion in these plasmablasts through B cell receptor sequencing analysis. This indicates a strong connection between B‐cell dysregulation and autoantibody production leading to irAEs. Furthermore, ICIs may disrupt Tfh cells, which are vital for B‐cell maturation and antibody production, implying that Tfh cells contribute to autoantibody production. 18 , 114 Therefore, ICIs may directly contribute to autoantibody production by directly affecting B cells and indirectly by altering Tfh cell function.
Furthermore, it is increasingly evident that autoantibodies can induce irAEs by inducing systemic inflammation, which subsequently results in tissue damage. 139 , 140 , 141 For example, in a retrospective study of 137 NSCLC patients treated with anti‐PD‐1, a notable association was found between pre‐existing antibodies such as rheumatoid factor, antinuclear antibody and anti‐thyroglobulin, and the incidence of irAEs. 141 In addition, Osorio et al. 142 discovered a correlation between ICI‐thyroiditis and anti‐thyroid antibodies in NSCLC. These findings suggest that the presence of heightened levels of pre‐existing or new autoantibodies contributes to irAEs in NSCLC.
3.6. Gut microbiome metabolic pathways associated with irAEs
Emerging evidence suggests that specific gut microbiota members, such as Faecalibacterium, Bacteroides and Clostridium, contribute to maintaining immune balance in the host. This balancing act is achieved through the promotion of Treg growth and cytokine production, factors implicated in the onset of irAEs. 143 Short‐chain fatty acids (SCFAs), including propionate, butyrate, acetate and valeric acid, are essential in engaging with immune cells, particularly colonic macrophages, and suppressing the synthesis of proinflammatory cytokines such as IL‐1, TNF‐α and IL‐6. 144 , 145 , 146 Similar to SCFAs, compounds such as indole‐3‐carboxaldehyde and polyamines, including spermine, are expected to offer defense against the development of irAEs. 143 , 147 Particularly, a study of the stool microbiota in 26 patients showed that most baseline phylotypes in those with colitis belonged to the Firmicutes group, whereas individuals without colitis fell under the Bacteroidetes category. 148 This observation was corroborated by another investigation involving 34 patients, indicating an inverse relationship between the prevalence of the Bacteroidetes phylum and the onset of CTLA4‐related colitis. 139 More recently, Hamada et al. 149 observed a higher abundance of Turicibacter and Acidaminococcus in the group of patients experiencing irAEs. These findings collectively imply a potential mechanistic association between particular bacterial groups in the gut microbiota and irAEs, offering a promising target for novel anti‐tumour immunotherapy.
3.7. Emerging biomarkers for predicting irAEs
Accumulating data have underscored the critical role of molecular pathological epidemiology in understanding the interrelationships between host exposures, tumour molecular characteristics and host immunity in immunotherapy. 150 , 151 External exposures, such as smoking, diet and lifestyle, alongside intrinsic host factors such as the tumour microenvironment, host genomics and systemic factors (e.g., gut microbiota), significantly affect the therapeutic response to ICIs and may possibly lead to irAEs. 145 , 152 , 153 , 154 In‐depth studies on these relationships have contributed to a more comprehensive understanding of how individual modifiable exposure factors affect the occurrence of irAEs. 150 For example, patients with a smoking history are more susceptible to the development of CIP during ICI treatment. 152 Although obesity may be associated with improved responses and survival rates in patients with NSCLC and other solid tumours treated with ICIs, 155 , 156 notably, a retrospective study on NSCLC patients receiving anti‐PD‐1/PD‐L1 therapy showed a positive correlation between increased body mass index (BMI) and irAEs risk. 153 Leiter et al. 157 also found that overweight patients (BMI ≥25 kg/m2) with fewer metabolic complications had a higher risk of irAEs occurrence. These studies highlight the value of lifestyle factors such as diet and smoking in assessing irAE risk. As previously mentioned, the role of the gut microbiome is vital in maintaining homeostasis in the host's gut and other sites, significantly influencing the development of irAEs. 145 , 154 Consequently, the role of specific gut microbiomes is gaining increasing recognition. 139 , 143 , 149 Future research should focus on identifying specific microbiomes that can optimise the outcomes of cancer immunotherapy. Therefore, considering the heterogenous nature of irAEs, future research should comprehensively integrate and analyse data on exposure factors, microbiomes, immune status, etc. 150 , 151 This integrated approach will help optimise immunotherapy strategies, minimise adverse reaction risks and enhance treatment efficacy.
4. MANAGEMENT STRATEGIES TO ABROGATE irAEs OF ICIS IN NSCLC
The effective management of irAEs holds significance for patients with NSCLC owing to their propensity to affect various organ systems and potentially lead to fatal outcomes. 15 , 67 In light of the increasing understanding of the multifaceted mechanisms underlying irAEs, an array of therapeutic approaches has emerged to address the pathogenesis of these events and mitigate their adverse effects. 15 Beyond the conventional use of corticosteroids, current research efforts are concentrated on the development of targeted therapeutic strategies tailored to alleviate irAEs. 158 These strategies encompass the modulation of the T‐cell response and migration, implementation of MoAbs, targeting of secreting cytokines, and inhibition of signalling pathways (Figures 4 and 5).
FIGURE 4.

How to manage immune‐related adverse events. Mechanism‐based therapeutic managements for the management of immune‐related adverse events (irAEs), including modulation of T cells activity and migration, monoclonal antibodies, targeting of cytokines receptors and signalling pathways are demonstrated.
FIGURE 5.

Summary of therapeutic drugs targeting different mechanisms for the management of immune‐related adverse events (irAEs). Therapeutic drugs targeting T cells activity and migration, monoclonal antibodies, targeting of cytokines receptors and signalling pathways are included.
4.1. Modulation of T‐cell activity and migration
Given that T‐cell activation or reactivation is commonly recognised as a key factor in the development of ICI‐related irAEs, 107 targeting T‐cell activity and movement might serve as an efficient approach to control irAEs in NSCLC (Figures 4 and 5).
4.1.1. Corticosteroid treatment
Corticosteroids have wide‐ranging impacts on multiple immune cells, such as boosting Treg cell production and activity, hindering TCR signalling, diminishing T‐cell effector abilities and favouring a proinflammatory cytokine milieu. Additionally, corticosteroids enhance the expression of immune checkpoints such as PD‐1, CTLA‐4, TIM‐3 and lymphocyte‐activation gene 3 (LAG‐3) on T cells in NSCLC and various other cancers. 27 , 159 , 160 , 161 Consistent with established management guidelines, glucocorticoids are recommended as the initial therapeutic approach for moderate to severe irAEs in NSCLC. 162 , 163 , 164 Nevertheless, there are conflicting reports concerning the effects of simultaneous corticosteroid and ICI treatment on tumour progression and patient survival. 161 , 165 , 166 , 167 For instance, Skribek et al. revealed that glucocorticoids effectively mitigated mild irAEs without hindering the effectiveness of ICI therapy in NSCLC patients, 168 while other studies have shown that high‐dose corticosteroids have a detrimental impact on the effectiveness of ICIs. 23 Hence, there is still ambiguity regarding whether the observed survival disadvantage can be solely attributed to the administration of high‐dose corticosteroids or overall aggressive immunosuppression.
4.1.2. Immunosuppressants
Furthermore, the inclusion of synthetic immunosuppressive agents as adjuncts to glucocorticoids is imperative in managing steroid‐resistant irAEs by obstructing T‐cell movement, growth or reactivation in NSCLC. 169 For instance, individuals experiencing steroid‐refractory pneumonitis, hepatitis, nephritis, pancreatitis and uveitis may benefit from treatment with immunosuppressants, such as hydroxychloroquine and mycophenolate, in NSCLC and other cancer types. 169 , 170 Less commonly used immunosuppressive therapies for steroid‐refractory irAEs encompass cyclosporine, tacrolimus and sulphasalazine. 169 The use of these drugs should be contemplated solely for irAEs that are unresponsive to corticosteroids, in conjunction with consultation from relevant specialists in the respective disease field.
4.1.3. Faecal microbiota transplantation and modulation
Recent research has introduced new findings indicating that altering the gut microbiome via faecal microbiota transplantation can efficiently relieve colitis linked to ICIs while reducing side effects from corticosteroids or immunosuppressives. 171 This approach has been found to significantly reduce the density of CD8+ T cells and increase Treg numbers, providing a potential target for new anti‐cancer treatments. 171 A recent forward‐looking clinical trial demonstrated that a microecological preparation (JK5G) corresponded with fewer irAEs compared to the control group in NSCLC patients undergoing ICIs and chemotherapy. 172 Importantly, JK5G postbiotics may also enhance the composition of the gut microbiota and positively modify the tumour milieu by elevating the levels of circulating CD3+CD4+ T cells and CD4/CD8 ratio. 172
4.2. Lymphocyte‐targeted monoclonal antibodies
Currently, the utilisation of lymphocyte‐targeted MoAbs is emerging as a pivotal innovation, particularly for managing severe steroid‐refractory irAEs in NSCLC. 169 , 173 MoAbs, such as rituximab, which targets CD20, and alemtuzumab, which targets CD52, have exhibited efficacy in this respect. 169 , 173 For instance, Santoro et al. 174 indicated the successful efficacy of rituximab in treating a patient with lung carcinoid tumours with steroid‐refractory pancreatitis induced by atezolizumab. Similarly, alemtuzumab, which primarily targets the CD52 antigen on B and T lymphocytes, has demonstrated efficacy in treating ICI‐related myocarditis in NSCLC and melanoma patients. 175 However, management decisions are mostly based on case reports, because prospective or comparative data on the outcome of those MoAbs are lacking. Some prospective studies are underway investigating the efficacy of rituximab/tocilizumab (NCT04375228) and CD24Fc (NCT04552704) in steroid‐refractory irAEs in solid tumours including NSCLC. In recent years, bispecific antibodies have shown strong efficacy in the treatment of malignant tumours, including NSCLC. 176 , 177 Double antibodies targeting two independent epitopes or antigens have undergone evaluation in the transformation and clinical research for NSCLC. 176 , 177 It is expected that these drugs will lead to a breakthrough in lung cancer treatment.
4.3. Targeting cytokines and their receptors
In contrast to corticosteroids that generally suppress various inflammatory pathways, cytokine inhibitors offer a more precise clinical method to diminish inflammation caused by ICIs in NSCLC. 91 Cytokine inhibitors, including anti‐TNF‐α agents (infliximab, etanercept, adalimumab) and anti‐IL‐17A agents (secukinumab) inhibitors, as well as cytokine receptor inhibitors, such as IL‐6 (tocilizumab) or IL‐α4β7 receptor (vedolizumab) inhibitors, have been investigated for their efficacy in managing steroid‐resistant irAEs in NSCLC. 91 , 158 , 178 Notably, TNFα inhibition has shown effectiveness in treating severe and refractory irAEs, such as colitis, inflammatory arthritis and hepatitis, in NSCLC and other cancer types. 179 , 180 , 181 A recent multicentre study found that inhibition of the IL‐6 receptor improved irAEs in 73% of patients with NSCLC and other cancers without affecting tumour immunity. 178 Additionally, tocilizumab was notably effective in treating nivolumab‐induced pneumonitis, with a 79.4% positive response in patients with NSCLC. 158 Moreover, a multicentre study revealed that infliximab or vedolizumab has been shown to reduce severe ICI‐related enterocolitis recurrence. 182 These studies provide substantial support for ongoing clinical trials assessing the safety and effectiveness of combining tocilizumab and vedolizumab with ICIs in NSCLC (NCT04691817, NCT04940299 and NCT04407247). Furthermore, IL‐17 inhibitors have shown profound therapeutic potential in addressing irAEs, such as intestinal issues, arthropathy and psoriasis, in NSCLC, although their use may potentially promote tumour immune escape. 183 , 184 Further clinical trials are underway in NSCLC to assess the effectiveness of tocilizumab in managing migrating irAEs (NCT04691817 NCT04940299). Despite the limited research data on the use of cytokine inhibitors for irAEs, the advancement of specialised cytokine treatments remains promising.
However, the use of anti‐TNF agents is under scrutiny for its potential adverse impact on survival outcomes in immunotherapy. For example, van Not et al., 185 in a 2022 JAMA Oncology study, reported reduced OS among patients with NSCLC who received anti‐TNF agents or other immunosuppressants along with steroids. Verheijden et al. 186 also discovered that the use of anti‐TNF agents might adversely impact OS in patients treated with steroid‐resistant ipilimumab and anti‐PD‐1. Moreover, the increased risk of opportunistic infections associated with anti‐TNF agent administration is becoming increasingly concerning, highlighting the need for careful use of such immunosuppressive treatments in managing irAEs. 187 , 188
4.4. Targeting signalling pathways
Cytokines frequently function as cellular messengers by triggering intracellular signalling pathways, suggesting that irAE treatment might be feasible by targeting these pathways. 189 The Janus kinase–signal transducer and activator of transcription (JAK–STAT) pathway is crucial for cellular activities and mediates the downstream effects of cytokines such as IL‐6, IL‐12, IL‐23 and IL‐17, making it a potential strategy for irAE treatment 115 , 189 (Figure 6). Tofacitinib, an inhibitor of this pathway, has been studied in cases of refractory colitis, myocarditis and arthritis, and showed clinical improvement in NSCLC, melanoma and other cancers. 190 , 191 , 192 , 193 Recently, Benesova et al. 194 found that CD8+ T cells from NSCLC and melanoma patients with musculoskeletal irAEs, when treated in vitro with tofacitinib, continued to secrete cytokines and display immune‐effector cell surface markers, inhibiting lung cancer progression. These findings accentuate the potential of JAK inhibitors to augment the capacity for managing severe refractory rheumatic irAEs in NSCLC. 194 Furthermore, the prospective evaluation of tofacitinib for addressing ICI‐related colitis in patients with NSCLC (NCT04768504) remains a topic of active investigation. 189 However, it is imperative to acknowledge that, as of the present, large‐scale studies in this domain remain limited. Consequently, the imperative to instigate additional clinical trials, aimed at the comprehensive evaluation of these inhibitors in preventing irAEs in NSCLC is unequivocal. 189 Beyond JAK–STAT inhibition, several other pathway inhibitors, including phosphoinositide 3‐kinase inhibitors (PI3K), 195 MAP kinase‐interacting serine/threonine‐protein kinase 1/2 (MNK1/2)inhibitors 196 and Bruton's tyrosine kinase (BTK) inhibitors, 197 , 198 have been implicated in autoimmune diseases and reducing irAEs in solid tumours 189 (Figure 6). Despite ongoing evaluations in preclinical models and clinical trials, these compounds hold the potential to herald a pioneering therapeutic paradigm for irAE management in NSCLC. 189 Clearly, the advancement of therapeutic approaches focused on cell signalling pathways in NSCLC presents an exciting prospect.
FIGURE 6.

Targeting intracellular signalling pathways in immune‐related adverse events (irAEs). (1) Janus kinase–signal transducer and activator of transcription (JAK–STAT) pathway activation promotes the transcription of genes for proteins with proliferation and inflammatory functions, including those related to cytokine and chemokine production during immunotherapy (e.g., interleukin [IL]‐6, IL‐12, IL‐17 and IL‐23). Inhibitors targeting the JAK pathway can suppress these proinflammatory factors, preventing the occurrence of irAEs. (2) Phosphoinositide 3‐kinase/protein kinase B/mammalian target of rapamycin (PI3K/Akt/mTOR) pathway activation stimulates inflammation‐related mRNA translation, leading to proinflammatory cytokine production which has been associated with irAEs. PI3K inhibitors can suppress this pathway. (3) MAP kinase interacting serine/threonine kinase 1/2 (MNK1/2), which are activated by the mitogen‐activated protein kinase kinase 5 (MEK5) and extracellular signal‐regulated kinase (ERK) pathways, increase the translation of mRNAs which promote immune activation and inflammation, exacerbating irAEs. MNK1/2 inhibitors can suppress this pathway and alleviate irAEs. (4) Lck/yes novel tyrosine kinase (Lyn) and spleen tyrosine kinase (Syk) tyrosine kinases phosphorylate Bruton's tyrosine kinase (BTK), activating phospholipase C gamma (PLCγ), leading to diacylglycerol (DAG) and inositol trisphosphate (IP3) production. This stimulates macrophages and B cells to produce cytokines (IL‐6, IL‐10 and C‐X‐C motif chemokine ligand 9 [CXCL9]) related to irAEs. BTK inhibitors can mitigate overactive immune responses and reduce inflammatory infiltration.
5. CONCLUSIONS AND FUTURE PERSPECTIVES
ICIs have significantly transformed the landscape of cancer therapy in advanced NSCLC; however, they are still limited due to the occurrence of severe irAEs. 8 This review has offered a detailed examination of the clinical manifestations of irAEs in NSCLC and elucidated potential therapeutic strategies rooted in mechanistic insights. Nevertheless, severe irAEs are often influenced by various factors and mediated by T cells, B cells, the host microbiome and a complex interplay of ligands, receptors and signalling pathways (Figure 7). Currently, the precise mechanisms underlying irAEs remain incompletely understood. Consequently, future research should prioritise the advancement of more efficacious therapeutic interventions in NSCLC, necessitating meticulous mechanistic preclinical and clinical investigations.
FIGURE 7.

Mechanisms involved in the occurrence of immune‐related adverse events (irAEs). (1) Excessive T‐cell activation leads to cross‐reactivity between tumour and normal tissue antigens, resulting in attacks on normal tissues. (2) Increased inflammatory factor production leads to excessive local and/or systemic inflammatory responses. (3) Individual genetic polymorphisms contribute to irAE susceptibility. (4) Abnormal activation and clonal expansion of B cells can occur. (5) Production of pre‐existing or new antibodies. (6) Immune dysregulation in the gut microbiome.
Recent strides in murine models for irAEs, exemplified by patient‐derived xenografts and humanised mouse models, have exhibited potential in devising targeted strategies aimed at ameliorating the toxicity associated with ICIs. 199 Nonetheless, recognising the constraints of preclinical experimental models in accurately depicting irAEs is crucial. 199 These limitations encompass inter‐species disparities in immune responses, the utilisation of allogenic cell lines instead of naturally occurring tumour growth and the inability to faithfully recapitulate genuine irAEs in murine systems. 163 , 200 , 201 Concurrently, the assimilation of clinical and high‐throughput data into the formulation of precise irAEs prediction models has enriched our understanding of irAE aetiology and pathogenesis. This data‐driven approach has also empowered clinicians in discerning the selection of appropriate therapeutic regimens for clinical implementation. 163 , 202 Noteworthy is the pioneering work by Jing et al., 37 who have proffered an innovative strategy that amalgamates real‐world pharmacovigilance and molecular omics data to construct a predictive model involving lymphocyte cytosolic protein 1 (LCP1) and adenosine diphosphate‐dependent glucokinase (ADPGK) for irAEs across diverse cancer types, including NSCLC. Consequently, the accessibility and availability of human multi‐omics data have become increasingly cost‐effective, aided by open repositories, thereby facilitating widespread utilisation. 203
Furthermore, progress in synthetic biology has opened up possibilities for utilising bioengineering and nanotechnology to develop innovative therapeutic strategies, potentially offering alternative treatments for irAEs. 204 An example of such innovation is the design of bi/tri‐specific antibodies capable of simultaneously binding to two or three distinct entities, presenting a promising avenue for mitigating ICI‐induced toxicity while preserving efficacy. 176 Recent advancements encompass the development of recombinant humanised PD‐L1/CTLA‐4 or PD‐1/PD‐L1 bispecific antibody Fc fusion proteins, which have evinced auspicious outcomes in preclinical investigations. 176 , 177 However, it behooves us to acknowledge that their therapeutic efficacy in clinical trials has yet to manifest commensurate promise in the context of NSCLC. Furthermore, alongside the exploration of novel antibodies, there is ongoing research on antibody delivery systems, specifically those with slow‐release capabilities, such as nanoparticles, virus vectors and bacteriophages. 204 Thus, the clinical transformation of novel technology in the management of irAEs has great application prospects.
In summary, while the path forward presents numerous challenges in understanding and mitigating irAEs in NSCLC, the integration of novel therapeutic modalities promises to reshape this landscape. We anticipate that this scenario will offer significant insights for developing irAEs mitigation approaches, thereby aiding in the continuation of anti‐cancer therapies.
AUTHOR CONTRIBUTIONS
Design: Xuwen Lin, Jie Yao, Xinying Xue and Mei Xie. Writing: Xuwen Lin, Mei Xie and Ying Jing. Figures: Xuwen Lin and Mei Xie. Review and editing: Xinying Xue, Jie Yao, Xidong Ma, Xinyu Bao, Xin Zhang, Yinguang Zhang, Yiming Liu, Wenya Han, Jialin Song and Yiran Liang. Funding: Xinying Xue. All the authors reviewed and approved the final manuscript.
CONFLICT OF INTEREST STATEMENT
All authors have agreed to publish this manuscript.
ETHICS STATEMENT
Not applicable.
ACKNOWLEDGEMENTS
All figures were created with BioRender.com. This work was supported by the National Natural Science Foundation of China (62176166, 62076254 and 82272768) and the Capital Medical University Affiliated Beijing Shijitan Hospital Talent Training Program during the ‘14th Five‐Year Plan’ period (2023DTRXXY).
Lin X, Xie M, Yao J, et al. Immune‐related adverse events in non‐small cell lung cancer: Occurrence, mechanisms and therapeutic strategies. Clin Transl Med. 2024;14:e1613. 10.1002/ctm2.1613
Xuwen Lin, Mei Xie and Jie Yao contributed equally to this work.
Contributor Information
Ying Jing, Email: jingying@ipm-gba.org.cn.
Xinying Xue, Email: xuexinying2988@bjsjth.cn.
DATA AVAILABILITY STATEMENT
Not applicable.
REFERENCES
- 1. Siegel RL, Miller KD, Fuchs HE, Jemal A. Cancer statistics, 2022. CA Cancer J Clin. 2022;72(1):7‐33. doi: 10.3322/caac.21708 [DOI] [PubMed] [Google Scholar]
- 2. Miller KD, Nogueira L, Devasia T, et al. Cancer treatment and survivorship statistics, 2022. CA Cancer J Clin. 2022;72(5):409‐436. doi: 10.3322/caac.21731 [DOI] [PubMed] [Google Scholar]
- 3. Wu Q, Qian W, Sun X, Jiang S. Small‐molecule inhibitors, immune checkpoint inhibitors, and more: FDA‐approved novel therapeutic drugs for solid tumors from 1991 to 2021. J Hematol Oncol. 2022;15(1):143. doi: 10.1186/s13045-022-01362-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4. Gubens MA, Sequist LV, Stevenson JP, et al. Pembrolizumab in combination with ipilimumab as second‐line or later therapy for advanced non‐small‐cell lung cancer: KEYNOTE‐021 cohorts D and H. Lung Cancer. 2019;130:59‐66. doi: 10.1016/j.lungcan.2018.12.015 [DOI] [PubMed] [Google Scholar]
- 5. Gettinger SN, Redman MW, Bazhenova L, et al. Nivolumab plus ipilimumab vs nivolumab for previously treated patients with stage IV squamous cell lung cancer: the lung‐MAP S1400I phase 3 randomized clinical trial. JAMA Oncol. 2021;7(9):1368‐1377. doi: 10.1001/jamaoncol.2021.2209 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6. Reck M, Rodriguez‐Abreu D, Robinson AG, et al. Five‐year outcomes with pembrolizumab versus chemotherapy for metastatic non‐small‐cell lung cancer with PD‐L1 tumor proportion score >/= 50. J Clin Oncol. 2021;39(21):2339‐2349. doi: 10.1200/JCO.21.00174 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7. Herbst RS, Giaccone G, de Marinis F, et al. Atezolizumab for first‐line treatment of PD‐L1‐selected patients with NSCLC. N Engl J Med. 2020;383(14):1328‐1339. doi: 10.1056/NEJMoa1917346 [DOI] [PubMed] [Google Scholar]
- 8. Zhou X, Yao Z, Yang H, Liang N, Zhang X, Zhang F. 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. 2020;18(1):87. doi: 10.1186/s12916-020-01549-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9. Sun X, Roudi R, Dai T, et al. Immune‐related adverse events associated with programmed cell death protein‐1 and programmed cell death ligand 1 inhibitors for non‐small cell lung cancer: a PRISMA systematic review and meta‐analysis. BMC Cancer. 2019;19(1):558. doi: 10.1186/s12885-019-5701-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10. Amoroso V, Gallo F, Alberti A, et al. Immune‐related adverse events as potential surrogates of immune checkpoint inhibitors' efficacy: a systematic review and meta‐analysis of randomized studies. ESMO Open. 2023;8(2):100787. doi: 10.1016/j.esmoop.2023.100787 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. Socinski MA, Jotte RM, Cappuzzo F, et al. Atezolizumab for first‐line treatment of metastatic nonsquamous NSCLC. N Engl J Med. 2018;378(24):2288‐2301. doi: 10.1056/NEJMoa1716948 [DOI] [PubMed] [Google Scholar]
- 12. West H, McCleod M, Hussein M, et al. Atezolizumab in combination with carboplatin plus nab‐paclitaxel chemotherapy compared with chemotherapy alone as first‐line treatment for metastatic non‐squamous non‐small‐cell lung cancer (IMpower130): a multicentre, randomised, open‐label, phase 3 trial. Lancet Oncol. 2019;20(7):924‐937. doi: 10.1016/S1470-2045(19)30167-6 [DOI] [PubMed] [Google Scholar]
- 13. Nishio M, Barlesi F, West H, et al. Atezolizumab plus chemotherapy for first‐line treatment of nonsquamous NSCLC: results from the randomized phase 3 IMpower132 trial. J Thorac Oncol. 2021;16(4):653‐664. doi: 10.1016/j.jtho.2020.11.025 [DOI] [PubMed] [Google Scholar]
- 14. Yan YD, Cui JJ, Fu J, et al. A network comparison on safety profiling of immune checkpoint inhibitors in advanced lung cancer. Front Immunol. 2021;12:760737. doi: 10.3389/fimmu.2021.760737 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15. Conroy M, Naidoo J. Immune‐related adverse events and the balancing act of immunotherapy. Nat Commun. 2022;13(1):392. doi: 10.1038/s41467-022-27960-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16. Yang H, Yao Z, Zhou X, Zhang W, Zhang X, Zhang F. Immune‐related adverse events of checkpoint inhibitors: insights into immunological dysregulation. Clin Immunol. 2020;213:108377. doi: 10.1016/j.clim.2020.108377 [DOI] [PubMed] [Google Scholar]
- 17. Sullivan RJ, Weber JS. Immune‐related toxicities of checkpoint inhibitors: mechanisms and mitigation strategies. Nat Rev Drug Discov. 2022;21(7):495‐508. doi: 10.1038/s41573-021-00259-5 [DOI] [PubMed] [Google Scholar]
- 18. Gutierrez‐Melo N, Baumjohann D. T follicular helper cells in cancer. Trends Cancer. 2023;9(4):309‐325. doi: 10.1016/j.trecan.2022.12.007 [DOI] [PubMed] [Google Scholar]
- 19. Shi J, Hou S, Fang Q, Liu X, Liu X, Qi H. PD‐1 controls follicular T helper cell positioning and function. Immunity. 2018;49(2):264‐274. doi: 10.1016/j.immuni.2018.06.012 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20. Barron CC, Stefanova I, Cha Y, et al. Chronic immune‐related adverse events in patients with cancer receiving immune checkpoint inhibitors: a systematic review. J Immunother Cancer. 2023;11(8). doi: 10.1136/jitc-2022-006500 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21. Hsu ML, Murray JC, Psoter KJ, et al. Clinical features, survival, and burden of toxicities in survivors more than one year after lung cancer immunotherapy. Oncologist. 2022;27(11):971‐981. doi: 10.1093/oncolo/oyac140 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22. Yoest JM. Clinical features, predictive correlates, and pathophysiology of immune‐related adverse events in immune checkpoint inhibitor treatments in cancer: a short review. Immunotargets Ther. 2017;6:73‐82. doi: 10.2147/ITT.S126227 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23. Morad G, Helmink BA, Sharma P, Wargo JA. Hallmarks of response, resistance, and toxicity to immune checkpoint blockade. Cell. 2022;185(3):576. doi: 10.1016/j.cell.2022.01.008 [DOI] [PubMed] [Google Scholar]
- 24. Rudd CE, Taylor A, Schneider H. CD28 and CTLA‐4 coreceptor expression and signal transduction. Immunol Rev. 2009;229(1):12‐26. doi: 10.1111/j.1600-065X.2009.00770.x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25. Lu C, Redd PS, Lee JR, Savage N, Liu K. The expression profiles and regulation of PD‐L1 in tumor‐induced myeloid‐derived suppressor cells. Oncoimmunology. 2016;5(12):e1247135. doi: 10.1080/2162402X.2016.1247135 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26. Garcia‐Diaz A, Shin DS, Moreno BH, et al. Interferon receptor signaling pathways regulating PD‐L1 and PD‐L2 expression. Cell Rep. 2019;29(11):3766. doi: 10.1016/j.celrep.2019.11.113 [DOI] [PubMed] [Google Scholar]
- 27. Pauken KE, Dougan M, Rose NR, Lichtman AH, Sharpe AH. Adverse events following cancer immunotherapy: obstacles and opportunities. Trends Immunol. 2019;40(6):511‐523. doi: 10.1016/j.it.2019.04.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28. Seto T, Nosaki K, Shimokawa M, et al. Phase II study of atezolizumab with bevacizumab for non‐squamous non‐small cell lung cancer with high PD‐L1 expression (@Be Study). J Immunother Cancer. 2022;10(2):e004025. doi: 10.1136/jitc-2021-004025 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29. Reck M, Wehler T, Orlandi F, et al. Safety and patient‐reported outcomes of atezolizumab plus chemotherapy with or without bevacizumab versus bevacizumab plus chemotherapy in non‐small‐cell lung cancer. J Clin Oncol. 2020;38(22):2530‐2542. doi: 10.1200/JCO.19.03158 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30. Pujol JL, Greillier L, Audigier‐Valette C, et al. A randomized non‐comparative phase II study of anti‐programmed cell death‐ligand 1 atezolizumab or chemotherapy as second‐line therapy in patients with small cell lung cancer: results from the IFCT‐1603 trial. J Thorac Oncol. 2019;14(5):903‐913. doi: 10.1016/j.jtho.2019.01.008 [DOI] [PubMed] [Google Scholar]
- 31. Planchard D, Reinmuth N, Orlov S, et al. ARCTIC: durvalumab with or without tremelimumab as third‐line or later treatment of metastatic non‐small‐cell lung cancer. Ann Oncol. 2020;31(5):609‐618. doi: 10.1016/j.annonc.2020.02.006 [DOI] [PubMed] [Google Scholar]
- 32. Herbst RS, Majem M, Barlesi F, et al. COAST: an open‐label, phase II, multidrug platform study of durvalumab alone or in combination with oleclumab or monalizumab in patients with unresectable, stage III non‐small‐cell lung cancer. J Clin Oncol. 2022;40(29):3383‐3393. doi: 10.1200/JCO.22.00227 [DOI] [PubMed] [Google Scholar]
- 33. Rothschild SI, Zippelius A, Eboulet EI, et al. SAKK 16/14: durvalumab in addition to neoadjuvant chemotherapy in patients with stage IIIA(N2) non‐small‐cell lung cancer—a multicenter single‐arm phase II trial. J Clin Oncol. 2021;39(26):2872‐2880. doi: 10.1200/JCO.21.00276 [DOI] [PubMed] [Google Scholar]
- 34. Ikeda S, Kato T, Kenmotsu H, et al. A phase II study of atezolizumab for pretreated advanced/recurrent non‐small cell lung cancer with idiopathic interstitial pneumonias: rationale and design for the TORG1936/AMBITIOUS study. Ther Adv Med Oncol. 2020;12:1758835920922022. doi: 10.1177/1758835920922022 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35. Peters S, Gettinger S, Johnson ML, et al. Phase II trial of atezolizumab as first‐line or subsequent therapy for patients with programmed death‐ligand 1‐selected advanced non‐small‐cell lung cancer (BIRCH). J Clin Oncol. 2017;35(24):2781‐2789. doi: 10.1200/JCO.2016.71.9476 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36. Mahmood SS, Fradley MG, Cohen JV, et al. Myocarditis in patients treated with immune checkpoint inhibitors. J Am Coll Cardiol. 2018;71(16):1755‐1764. doi: 10.1016/j.jacc.2018.02.037 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37. Jing Y, Liu J, Ye Y, et al. Multi‐omics prediction of immune‐related adverse events during checkpoint immunotherapy. Nat Commun. 2020;11(1):4946. doi: 10.1038/s41467-020-18742-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38. Antonia SJ, Borghaei H, Ramalingam SS, et al. Four‐year survival with nivolumab in patients with previously treated advanced non‐small‐cell lung cancer: a pooled analysis. Lancet Oncol. 2019;20(10):1395‐1408. doi: 10.1016/S1470-2045(19)30407-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39. Felip E, Ardizzoni A, Ciuleanu T, et al. CheckMate 171: a phase 2 trial of nivolumab in patients with previously treated advanced squamous non‐small cell lung cancer, including ECOG PS 2 and elderly populations. Eur J Cancer. 2020;127:160‐172. doi: 10.1016/j.ejca.2019.11.019 [DOI] [PubMed] [Google Scholar]
- 40. Lu S, Wang J, Cheng Y, et al. Nivolumab versus docetaxel in a predominantly Chinese patient population with previously treated advanced non‐small cell lung cancer: 2‐year follow‐up from a randomized, open‐label, phase 3 study (CheckMate 078). Lung Cancer. 2021;152:7‐14. doi: 10.1016/j.lungcan.2020.11.013 [DOI] [PubMed] [Google Scholar]
- 41. Stratigos AJ, Sekulic A, Peris K, et al. Cemiplimab in locally advanced basal cell carcinoma after hedgehog inhibitor therapy: an open‐label, multi‐centre, single‐arm, phase 2 trial. Lancet Oncol. 2021;22(6):848‐857. doi: 10.1016/S1470-2045(21)00126-1 [DOI] [PubMed] [Google Scholar]
- 42. Nosaki K, Saka H, Hosomi Y, et al. Safety and efficacy of pembrolizumab monotherapy in elderly patients with PD‐L1‐positive advanced non‐small‐cell lung cancer: pooled analysis from the KEYNOTE‐010, KEYNOTE‐024, and KEYNOTE‐042 studies. Lung Cancer. 2019;135:188‐195. doi: 10.1016/j.lungcan.2019.07.004 [DOI] [PubMed] [Google Scholar]
- 43. de Castro G Jr, Kudaba I, Wu YL, et al. Five‐year outcomes with pembrolizumab versus chemotherapy as first‐line therapy in patients with non‐small‐cell lung cancer and programmed death ligand‐1 tumor proportion score >/= 1% in the KEYNOTE‐042 study. J Clin Oncol. 2023;41(11):1986‐1991. doi: 10.1200/JCO.21.02885 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44. Mok TSK, Wu YL, Kudaba I, et al. Pembrolizumab versus chemotherapy for previously untreated, PD‐L1‐expressing, locally advanced or metastatic non‐small‐cell lung cancer (KEYNOTE‐042): a randomised, open‐label, controlled, phase 3 trial. Lancet. 2019;393(10183):1819‐1830. doi: 10.1016/S0140-6736(18)32409-7 [DOI] [PubMed] [Google Scholar]
- 45. Felip E, Altorki N, Zhou C, et al. Adjuvant atezolizumab after adjuvant chemotherapy in resected stage IB‐IIIA non‐small‐cell lung cancer (IMpower010): a randomised, multicentre, open‐label, phase 3 trial. Lancet. 2021;398(10308):1344‐1357. doi: 10.1016/S0140-6736(21)02098-5 [DOI] [PubMed] [Google Scholar]
- 46. Mazieres J, Rittmeyer A, Gadgeel S, et al. Atezolizumab versus docetaxel in pretreated patients with NSCLC: final results from the randomized phase 2 POPLAR and phase 3 OAK clinical trials. J Thorac Oncol. 2021;16(1):140‐150. doi: 10.1016/j.jtho.2020.09.022 [DOI] [PubMed] [Google Scholar]
- 47. Lee JM, Vallieres E, Ding B, et al. Safety of adjuvant atezolizumab after pneumonectomy/bilobectomy in stage II‐IIIA non‐small cell lung cancer in the randomized phase III IMpower010 trial. J Thorac Cardiovasc Surg. 2023;166(3):655‐666 e7. doi: 10.1016/j.jtcvs.2023.01.012 [DOI] [PubMed] [Google Scholar]
- 48. Cho BC, Abreu DR, Hussein M, et al. Tiragolumab plus atezolizumab versus placebo plus atezolizumab as a first‐line treatment for PD‐L1‐selected non‐small‐cell lung cancer (CITYSCAPE): primary and follow‐up analyses of a randomised, double‐blind, phase 2 study. Lancet Oncol. 2022;23(6):781‐792. doi: 10.1016/S1470-2045(22)00226-1 [DOI] [PubMed] [Google Scholar]
- 49. Baas P, Scherpereel A, Nowak AK, et al. First‐line nivolumab plus ipilimumab in unresectable malignant pleural mesothelioma (CheckMate 743): a multicentre, randomised, open‐label, phase 3 trial. Lancet. 2021;397(10272):375‐386. doi: 10.1016/S0140-6736(20)32714-8 [DOI] [PubMed] [Google Scholar]
- 50. Reck M, Ciuleanu TE, Cobo M, et al. First‐line nivolumab plus ipilimumab with two cycles of chemotherapy versus chemotherapy alone (four cycles) in advanced non‐small‐cell lung cancer: CheckMate 9LA 2‐year update. ESMO Open. 2021;6(5):100273. doi: 10.1016/j.esmoop.2021.100273 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51. Boyer M, Sendur MAN, Rodriguez‐Abreu D, et al. Pembrolizumab plus ipilimumab or placebo for metastatic non‐small‐cell lung cancer with PD‐L1 tumor proportion score >/= 50%: randomized, double‐blind phase III KEYNOTE‐598 study. J Clin Oncol. 2021;39(21):2327‐2338. doi: 10.1200/JCO.20.03579 [DOI] [PubMed] [Google Scholar]
- 52. Niu J, Maurice‐Dror C, Lee DH, et al. First‐in‐human phase 1 study of the anti‐TIGIT antibody vibostolimab as monotherapy or with pembrolizumab for advanced solid tumors, including non‐small‐cell lung cancer(☆). Ann Oncol. 2022;33(2):169‐180. doi: 10.1016/j.annonc.2021.11.002 [DOI] [PubMed] [Google Scholar]
- 53. Joosse ME, Nederlof I, Walker LSK, Samsom JN. Tipping the balance: inhibitory checkpoints in intestinal homeostasis. Mucosal Immunol. 2019;12(1):21‐35. doi: 10.1038/s41385-018-0113-5 [DOI] [PubMed] [Google Scholar]
- 54. Luoma AM, Suo S, Williams HL, et al. Molecular pathways of colon inflammation induced by cancer immunotherapy. Cell. 2020;182(3):655‐671.e22. doi: 10.1016/j.cell.2020.06.001 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55. Alissafi T, Hatzioannou A, Legaki AI, Varveri A, Verginis P. Balancing cancer immunotherapy and immune‐related adverse events: the emerging role of regulatory T cells. J Autoimmun. 2019;104:102310. doi: 10.1016/j.jaut.2019.102310 [DOI] [PubMed] [Google Scholar]
- 56. Axelrod ML, Meijers WC, Screever EM, et al. T cells specific for alpha‐myosin drive immunotherapy‐related myocarditis. Nature. 2022;611(7937):818‐826. doi: 10.1038/s41586-022-05432-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57. Li M, Gan L, Song A, Xue J, Lu Y. Rethinking pulmonary toxicity in advanced non‐small cell lung cancer in the era of combining anti‐PD‐1/PD‐L1 therapy with thoracic radiotherapy. Biochim Biophys Acta Rev Cancer. 2019;1871(2):323‐330. doi: 10.1016/j.bbcan.2019.02.004 [DOI] [PubMed] [Google Scholar]
- 58. Xiao Y, Yu S, Zhu B, et al. RGMb is a novel binding partner for PD‐L2 and its engagement with PD‐L2 promotes respiratory tolerance. J Exp Med. 2014;211(5):943‐959. doi: 10.1084/jem.20130790 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59. Curry JL, Tetzlaff MT, Nagarajan P, et al. Diverse types of dermatologic toxicities from immune checkpoint blockade therapy. J Cutan Pathol. 2017;44(2):158‐176. doi: 10.1111/cup.12858 [DOI] [PubMed] [Google Scholar]
- 60. Ellis SR, Vierra AT, Millsop JW, Lacouture ME, Kiuru M. Dermatologic toxicities to immune checkpoint inhibitor therapy: a review of histopathologic features. J Am Acad Dermatol. 2020;83(4):1130‐1143. doi: 10.1016/j.jaad.2020.04.105 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61. Iwama S, Kobayashi T, Yasuda Y, Arima H. Immune checkpoint inhibitor‐related thyroid dysfunction. Best Pract Res Clin Endocrinol Metab. 2022;36(3):101660. doi: 10.1016/j.beem.2022.101660 [DOI] [PubMed] [Google Scholar]
- 62. Hellmann MD, Ciuleanu TE, Pluzanski A, et al. Nivolumab plus ipilimumab in lung cancer with a high tumor mutational burden. N Engl J Med. 2018;378(22):2093‐2104. doi: 10.1056/NEJMoa1801946 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63. Akinboro O, Larkins E, Pai‐Scherf LH, et al. FDA approval summary: pembrolizumab, atezolizumab, and cemiplimab‐rwlc as single agents for first‐line treatment of advanced/metastatic PD‐L1‐high NSCLC. Clin Cancer Res. 2022;28(11):2221‐2228. doi: 10.1158/1078-0432.CCR-21-3844 [DOI] [PubMed] [Google Scholar]
- 64. Sezer A, Kilickap S, Gumus M, et al. Cemiplimab monotherapy for first‐line treatment of advanced non‐small‐cell lung cancer with PD‐L1 of at least 50%: a multicentre, open‐label, global, phase 3, randomised, controlled trial. Lancet. 2021;397(10274):592‐604. doi: 10.1016/S0140-6736(21)00228-2 [DOI] [PubMed] [Google Scholar]
- 65. Moreno V, Garrido P, Papadopoulos KP, et al. Tolerability and antitumor activity of cemiplimab, a human monoclonal anti‐PD‐1, as monotherapy in patients with pretreated non‐small cell lung cancer (NSCLC): data from the phase 1 NSCLC expansion cohort. Lung Cancer. 2021;155:151‐155. doi: 10.1016/j.lungcan.2021.02.034 [DOI] [PubMed] [Google Scholar]
- 66. Borghaei H, Paz‐Ares L, Horn L, et al. Nivolumab versus docetaxel in advanced nonsquamous non‐small‐cell lung cancer. N Engl J Med. 2015;373(17):1627‐1639. doi: 10.1056/NEJMoa1507643 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67. Okiyama N, Tanaka R. Immune‐related adverse events in various organs caused by immune checkpoint inhibitors. Allergol Int. 2022;71(2):169‐178. doi: 10.1016/j.alit.2022.01.001 [DOI] [PubMed] [Google Scholar]
- 68. Rizvi NA, Mazieres J, Planchard D, et al. Activity and safety of nivolumab, an anti‐PD‐1 immune checkpoint inhibitor, for patients with advanced, refractory squamous non‐small‐cell lung cancer (CheckMate 063): a phase 2, single‐arm trial. Lancet Oncol. 2015;16(3):257‐265. doi: 10.1016/S1470-2045(15)70054-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69. Garon EB, Kim JS, Govindan R. Pemetrexed maintenance with or without pembrolizumab in non‐squamous non‐small cell lung cancer: a cross‐trial comparison of KEYNOTE‐189 versus PARAMOUNT, PRONOUNCE, and JVBL. Lung Cancer. 2021;151:25‐29. doi: 10.1016/j.lungcan.2020.11.018 [DOI] [PubMed] [Google Scholar]
- 70. Mansfield AS, Herbst RS. Outcomes with pembrolizumab monotherapy in patients with programmed death‐ligand 1‐positive NSCLC with brain metastases: pooled analysis of KEYNOTE‐001, 010, 024, and 042. JTO Clin Res Rep. 2021;2(8):100205. doi: 10.1016/j.jtocrr.2021.100205 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71. Govindan R, Szczesna A, Ahn MJ, et al. Phase III trial of ipilimumab combined with paclitaxel and carboplatin in advanced squamous non‐small‐cell lung cancer. J Clin Oncol. 2017;35(30):3449‐3457. doi: 10.1200/JCO.2016.71.7629 [DOI] [PubMed] [Google Scholar]
- 72. Paz‐Ares LG, Ramalingam SS, Ciuleanu TE, et al. First‐line nivolumab plus ipilimumab in advanced NSCLC: 4‐year outcomes from the randomized, open‐label, phase 3 CheckMate 227 part 1 trial. J Thorac Oncol. 2022;17(2):289‐308. doi: 10.1016/j.jtho.2021.09.010 [DOI] [PubMed] [Google Scholar]
- 73. Reck M, Rodriguez‐Abreu D, Robinson AG, et al. Updated analysis of KEYNOTE‐024: pembrolizumab versus platinum‐based chemotherapy for advanced non‐small‐cell lung cancer with PD‐L1 tumor proportion score of 50% or greater. J Clin Oncol. 2019;37(7):537‐546. doi: 10.1200/JCO.18.00149 [DOI] [PubMed] [Google Scholar]
- 74. Rijavec E, Genova C, Barletta G, et al. Ipilimumab in non‐small cell lung cancer and small‐cell lung cancer: new knowledge on a new therapeutic strategy. Expert Opin Biol Ther. 2014;14(7):1007‐1017. doi: 10.1517/14712598.2014.907786 [DOI] [PubMed] [Google Scholar]
- 75. Riudavets M, Naigeon M, Texier M, et al. Gefitinib plus tremelimumab combination in refractory non‐small cell lung cancer patients harbouring EGFR mutations: the GEFTREM phase I trial. Lung Cancer. 2022;166:255‐264. doi: 10.1016/j.lungcan.2021.11.018 [DOI] [PubMed] [Google Scholar]
- 76. Fujimoto D, Yomota M, Sekine A, et al. Nivolumab for advanced non‐small cell lung cancer patients with mild idiopathic interstitial pneumonia: a multicenter, open‐label single‐arm phase II trial. Lung Cancer. 2019;134:274‐278. doi: 10.1016/j.lungcan.2019.06.001 [DOI] [PubMed] [Google Scholar]
- 77. Antonia SJ, Villegas A, Daniel D, et al. Durvalumab after chemoradiotherapy in stage III non‐small‐cell lung cancer. N Engl J Med. 2017;377(20):1919‐1929. doi: 10.1056/NEJMoa1709937 [DOI] [PubMed] [Google Scholar]
- 78. Haratani K, Hayashi H, Chiba Y, et al. Association of immune‐related adverse events with nivolumab efficacy in non‐small‐cell lung cancer. JAMA Oncol. 2018;4(3):374‐378. doi: 10.1001/jamaoncol.2017.2925 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79. Provencio M, Nadal E, Insa A, et al. Neoadjuvant chemotherapy and nivolumab in resectable non‐small‐cell lung cancer (NADIM): an open‐label, multicentre, single‐arm, phase 2 trial. Lancet Oncol. 2020;21(11):1413‐1422. doi: 10.1016/S1470-2045(20)30453-8 [DOI] [PubMed] [Google Scholar]
- 80. Borghaei H, Gettinger S, Vokes EE, et al. Five‐year outcomes from the randomized, phase III trials CheckMate 017 and 057: nivolumab versus docetaxel in previously treated non‐small‐cell lung cancer. J Clin Oncol. 2021;39(7):723‐733. doi: 10.1200/JCO.20.01605 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81. Lynch TJ, Bondarenko I, Luft A, et al. Ipilimumab in combination with paclitaxel and carboplatin as first‐line treatment in stage IIIB/IV non‐small‐cell lung cancer: results from a randomized, double‐blind, multicenter phase II study. J Clin Oncol. 2012;30(17):2046‐2054. doi: 10.1200/JCO.2011.38.4032 [DOI] [PubMed] [Google Scholar]
- 82. Leighl NB, Laurie SA, Goss GD, et al. CCTG BR34: a randomized phase 2 trial of durvalumab and tremelimumab with or without platinum‐based chemotherapy in patients with metastatic NSCLC. J Thorac Oncol. 2022;17(3):434‐445. doi: 10.1016/j.jtho.2021.10.023 [DOI] [PubMed] [Google Scholar]
- 83. Arrieta O, Barron F, Ramirez‐Tirado LA, et al. Efficacy and safety of pembrolizumab plus docetaxel vs docetaxel alone in patients with previously treated advanced non‐small cell lung cancer: the PROLUNG phase 2 randomized clinical trial. JAMA Oncol. 2020;6(6):856‐864. doi: 10.1001/jamaoncol.2020.0409 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84. Carbone DP, Reck M, Paz‐Ares L, et al. First‐line nivolumab in stage IV or recurrent non‐small‐cell lung cancer. N Engl J Med. 2017;376(25):2415‐2426. doi: 10.1056/NEJMoa1613493 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85. Schoenfeld JD, Giobbie‐Hurder A, Ranasinghe S, et al. Durvalumab plus tremelimumab alone or in combination with low‐dose or hypofractionated radiotherapy in metastatic non‐small‐cell lung cancer refractory to previous PD(L)‐1 therapy: an open‐label, multicentre, randomised, phase 2 trial. Lancet Oncol. 2022;23(2):279‐291. doi: 10.1016/S1470-2045(21)00658-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86. O'Byrne KJ, Lee KH, Kim SW, et al. First‐line nivolumab + ipilimumab in advanced NSCLC: CheckMate 227 subpopulation analyses in Asian patients. ESMO Open. 2022;7(1):100394. doi: 10.1016/j.esmoop.2022.100394 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87. Gandhi L, Rodriguez‐Abreu D, Gadgeel S, et al. Pembrolizumab plus chemotherapy in metastatic non‐small‐cell lung cancer. N Engl J Med. 2018;378(22):2078‐2092. doi: 10.1056/NEJMoa1801005 [DOI] [PubMed] [Google Scholar]
- 88. Park K, Ozguroglu M, Vansteenkiste J, et al. Avelumab versus docetaxel in patients with platinum‐treated advanced NSCLC: 2‐year follow‐up from the JAVELIN lung 200 phase 3 trial. J Thorac Oncol. 2021;16(8):1369‐1378. doi: 10.1016/j.jtho.2021.03.009 [DOI] [PubMed] [Google Scholar]
- 89. Paz‐Ares L, Luft A, Vicente D, et al. Pembrolizumab plus chemotherapy for squamous non‐small‐cell lung cancer. N Engl J Med. 2018;379(21):2040‐2051. doi: 10.1056/NEJMoa1810865 [DOI] [PubMed] [Google Scholar]
- 90. Pinto JA, Raez LE, Oliveres H, Rolfo CC. Current knowledge of ipilimumab and its use in treating non‐small cell lung cancer. Expert Opin Biol Ther. 2019;19(6):509‐515. doi: 10.1080/14712598.2019.1610380 [DOI] [PubMed] [Google Scholar]
- 91. Kang JH, Bluestone JA, Young A. Predicting and preventing immune checkpoint inhibitor toxicity: targeting cytokines. Trends Immunol. 2021;42(4):293‐311. doi: 10.1016/j.it.2021.02.006 [DOI] [PubMed] [Google Scholar]
- 92. Knochelmann HM, Dwyer CJ, Bailey SR, et al. When worlds collide: Th17 and Treg cells in cancer and autoimmunity. Cell Mol Immunol. 2018;15(5):458‐469. doi: 10.1038/s41423-018-0004-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93. Johnson DB, Balko JM, Compton ML, et al. Fulminant myocarditis with combination immune checkpoint blockade. N Engl J Med. 2016;375(18):1749‐1755. doi: 10.1056/NEJMoa1609214 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94. Berner F, Bomze D, Diem S, et al. Association of checkpoint inhibitor‐induced toxic effects with shared cancer and tissue antigens in non‐small cell lung cancer. JAMA Oncol. 2019;5(7):1043‐1047. doi: 10.1001/jamaoncol.2019.0402 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95. Berner F, Bomze D, Lichtensteiger C, et al. Autoreactive napsin A‐specific T cells are enriched in lung tumors and inflammatory lung lesions during immune checkpoint blockade. Sci Immunol. 2022;7(75):eabn9644. doi: 10.1126/sciimmunol.abn9644 [DOI] [PubMed] [Google Scholar]
- 96. Naidoo J, Cottrell TR, Lipson EJ, et al. Chronic immune checkpoint inhibitor pneumonitis. J Immunother Cancer. 2020;8(1):e000840. doi: 10.1136/jitc-2020-000840 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97. Lechner MG, Zhou Z, Hoang AT, et al. Clonally expanded, thyrotoxic effector CD8(+) T cells driven by IL‐21 contribute to checkpoint inhibitor thyroiditis. Sci Transl Med. 2023;15(696):eadg0675. doi: 10.1126/scitranslmed.adg0675 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98. Yshii LM, Gebauer CM, Pignolet B, et al. CTLA4 blockade elicits paraneoplastic neurological disease in a mouse model. Brain. 2016;139(11):2923‐2934. doi: 10.1093/brain/aww225 [DOI] [PubMed] [Google Scholar]
- 99. Larsabal M, Marti A, Jacquemin C, et al. Vitiligo‐like lesions occurring in patients receiving anti‐programmed cell death‐1 therapies are clinically and biologically distinct from vitiligo. J Am Acad Dermatol. 2017;76(5):863‐870. doi: 10.1016/j.jaad.2016.10.044 [DOI] [PubMed] [Google Scholar]
- 100. Suresh K, Naidoo J, Zhong Q, et al. The alveolar immune cell landscape is dysregulated in checkpoint inhibitor pneumonitis. J Clin Invest. 2019;129(10):4305‐4315. doi: 10.1172/JCI128654 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101. Suzuki K, Yanagihara T, Matsumoto K, et al. Immune‐checkpoint profiles for T cells in bronchoalveolar lavage fluid of patients with immune‐checkpoint inhibitor‐related interstitial lung disease. Int Immunol. 2020;32(8):547‐557. doi: 10.1093/intimm/dxaa022 [DOI] [PubMed] [Google Scholar]
- 102. Kruse B, Buzzai AC, Shridhar N, et al. CD4(+) T cell‐induced inflammatory cell death controls immune‐evasive tumours. Nature. 2023;618(7967):1033‐1040. doi: 10.1038/s41586-023-06199-x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103. Lozano AX, Chaudhuri AA, Nene A, et al. T cell characteristics associated with toxicity to immune checkpoint blockade in patients with melanoma. Nat Med. 2022;28(2):353‐362. doi: 10.1038/s41591-021-01623-z [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104. Kim ST, Chu Y, Misoi M, et al. Distinct molecular and immune hallmarks of inflammatory arthritis induced by immune checkpoint inhibitors for cancer therapy. Nat Commun. 2022;13(1):1970. doi: 10.1038/s41467-022-29539-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105. Nedoszytko B, Lange M, Sokolowska‐Wojdylo M, et al. The role of regulatory T cells and genes involved in their differentiation in pathogenesis of selected inflammatory and neoplastic skin diseases. Part I: Treg properties and functions. Postepy Dermatol Alergol. 2017;34(4):285‐294. doi: 10.5114/ada.2017.69305 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106. Basdeo SA, Cluxton D, Sulaimani J, et al. Ex‐Th17 (Nonclassical Th1) cells are functionally distinct from classical Th1 and Th17 cells and are not constrained by regulatory T cells. J Immunol. 2017;198(6):2249‐2259. doi: 10.4049/jimmunol.1600737 [DOI] [PubMed] [Google Scholar]
- 107. Bukhari S, Henick BS, Winchester RJ, et al. Single‐cell RNA sequencing reveals distinct T cell populations in immune‐related adverse events of checkpoint inhibitors. Cell Rep Med. 2023;4(1):100868. doi: 10.1016/j.xcrm.2022.100868 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108. Strauss L, Mahmoud MAA, Weaver JD, et al. Targeted deletion of PD‐1 in myeloid cells induces antitumor immunity. Sci Immunol. 2020;5(43). doi: 10.1126/sciimmunol.aay1863 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109. Nam S, Lee A, Lim J, Lim JS. Analysis of the expression and regulation of PD‐1 protein on the surface of myeloid‐derived suppressor cells (MDSCs). Biomol Ther (Seoul). 2019;27(1):63‐70. doi: 10.4062/biomolther.2018.201 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110. Ma P, Liu J, Qin J, et al. Expansion of pathogenic cardiac macrophages in immune checkpoint inhibitor myocarditis. Circulation. 2024;149(1):48‐66. doi: 10.1161/CIRCULATIONAHA.122.062551 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111. Bajpai G, Bredemeyer A, Li W, et al. Tissue resident CCR2– and CCR2+ cardiac macrophages differentially orchestrate monocyte recruitment and fate specification following myocardial injury. Circ Res. 2019;124(2):263‐278. doi: 10.1161/CIRCRESAHA.118.314028 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112. Hu H, Zakharov PN, Peterson OJ, Unanue ER. Cytocidal macrophages in symbiosis with CD4 and CD8 T cells cause acute diabetes following checkpoint blockade of PD‐1 in NOD mice. Proc Natl Acad Sci U S A. 2020;117(49):31319‐31330. doi: 10.1073/pnas.2019743117 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113. Khan AR, Hams E, Floudas A, Sparwasser T, Weaver CT, Fallon PG. PD‐L1hi B cells are critical regulators of humoral immunity. Nat Commun. 2015;6:5997. doi: 10.1038/ncomms6997 [DOI] [PubMed] [Google Scholar]
- 114. Baumjohann D, Brossart P. T follicular helper cells: linking cancer immunotherapy and immune‐related adverse events. J Immunother Cancer. 2021;9(6):e002588. doi: 10.1136/jitc-2021-002588 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115. Lim SY, Lee JH, Gide TN, et al. Circulating cytokines predict immune‐related toxicity in melanoma patients receiving anti‐PD‐1‐based immunotherapy. Clin Cancer Res. 2019;25(5):1557‐1563. doi: 10.1158/1078-0432.CCR-18-2795 [DOI] [PubMed] [Google Scholar]
- 116. von Euw E, Chodon T, Attar N, et al. CTLA4 blockade increases Th17 cells in patients with metastatic melanoma. J Transl Med. 2009;7:35. doi: 10.1186/1479-5876-7-35 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117. Weaver CT, Elson CO, Fouser LA, Kolls JK. The Th17 pathway and inflammatory diseases of the intestines, lungs, and skin. Annu Rev Pathol. 2013;8:477‐512. doi: 10.1146/annurev-pathol-011110-130318 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118. Chen M, Ma P, Zhang Y, et al. Divergent tumor and immune cell reprogramming underlying immunotherapy response and immune‐related adverse events in lung squamous cell carcinoma. J Immunother Cancer. 2023;11(10):e007305. doi: 10.1136/jitc-2023-007305 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119. Khan S, Khan SA, Luo X, et al. Immune dysregulation in cancer patients developing immune‐related adverse events. Br J Cancer. 2019;120(1):63‐68. doi: 10.1038/s41416-018-0155-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120. Muir CA, Clifton‐Bligh RJ, Long GV, et al. Thyroid immune‐related adverse events following immune checkpoint inhibitor treatment. J Clin Endocrinol Metab. 2021;106(9):e3704‐e3713. doi: 10.1210/clinem/dgab263 [DOI] [PubMed] [Google Scholar]
- 121. Lin X, Deng H, Yang Y, et al. Peripheral blood biomarkers for early diagnosis, severity, and prognosis of checkpoint inhibitor‐related pneumonitis in patients with lung cancer. Front Oncol. 2021;11:698832. doi: 10.3389/fonc.2021.698832 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122. Wang YN, Lou DF, Li DY, et al. Elevated levels of IL‐17A and IL‐35 in plasma and bronchoalveolar lavage fluid are associated with checkpoint inhibitor pneumonitis in patients with non‐small cell lung cancer. Oncol Lett. 2020;20(1):611‐622. doi: 10.3892/ol.2020.11618 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123. Liu W, Liu Y, Ma F, et al. Peripheral blood markers associated with immune‐related adverse effects in patients who had advanced non‐small cell lung cancer treated with PD‐1 inhibitors. Cancer Manag Res. 2021;13:765‐771. doi: 10.2147/CMAR.S293200 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124. Pagadala M, Sears TJ, Wu VH, et al. Germline modifiers of the tumor immune microenvironment implicate drivers of cancer risk and immunotherapy response. Nat Commun. 2023;14(1):2744. doi: 10.1038/s41467-023-38271-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125. Khan Z, Hammer C, Carroll J, et al. Genetic variation associated with thyroid autoimmunity shapes the systemic immune response to PD‐1 checkpoint blockade. Nat Commun. 2021;12(1):3355. doi: 10.1038/s41467-021-23661-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126. Luo J, Martucci VL, Quandt Z, et al. Immunotherapy‐mediated thyroid dysfunction: genetic risk and impact on outcomes with PD‐1 blockade in non‐small cell lung cancer. Clin Cancer Res. 2021;27(18):5131‐5140. doi: 10.1158/1078-0432.CCR-21-0921 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127. Groha S, Alaiwi SA, Xu W, et al. Germline variants associated with toxicity to immune checkpoint blockade. Nat Med. 2022;28(12):2584‐2591. doi: 10.1038/s41591-022-02094-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128. Udagawa C, Nakano MH, Yoshida T, et al. Association between genetic variants and the risk of nivolumab‐induced immune‐related adverse events. Pharmacogenomics. 2022;23(16):887‐901. doi: 10.2217/pgs-2022-0113 [DOI] [PubMed] [Google Scholar]
- 129. Dendrou CA, Petersen J, Rossjohn J, Fugger L. HLA variation and disease. Nat Rev Immunol. 2018;18(5):325‐339. doi: 10.1038/nri.2017.143 [DOI] [PubMed] [Google Scholar]
- 130. Hasan Ali O, Berner F, Bomze D, et al. Human leukocyte antigen variation is associated with adverse events of checkpoint inhibitors. Eur J Cancer. 2019;107:8‐14. doi: 10.1016/j.ejca.2018.11.009 [DOI] [PubMed] [Google Scholar]
- 131. Kobayashi T, Iwama S, Sugiyama D, et al. Anti‐pituitary antibodies and susceptible human leukocyte antigen alleles as predictive biomarkers for pituitary dysfunction induced by immune checkpoint inhibitors. J Immunother Cancer. 2021;9(5):e002493. doi: 10.1136/jitc-2021-002493 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132. Cappelli LC, Dorak MT, Bettinotti MP, Bingham CO, Shah AA. Association of HLA‐DRB1 shared epitope alleles and immune checkpoint inhibitor‐induced inflammatory arthritis. Rheumatology (Oxford). 2019;58(3):476‐480. doi: 10.1093/rheumatology/key358 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133. Correale P, Saladino RE, Giannarelli D, et al. HLA expression correlates to the risk of immune checkpoint inhibitor‐induced pneumonitis. Cells. 2020;9(9):1964. doi: 10.3390/cells9091964 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134. Petitprez F, de Reynies A, Keung EZ, et al. B cells are associated with survival and immunotherapy response in sarcoma. Nature. 2020;577(7791):556‐560. doi: 10.1038/s41586-019-1906-8 [DOI] [PubMed] [Google Scholar]
- 135. Rosser EC, Mauri C. Regulatory B cells: origin, phenotype, and function. Immunity. 2015;42(4):607‐612. doi: 10.1016/j.immuni.2015.04.005 [DOI] [PubMed] [Google Scholar]
- 136. Patel AJ, Willsmore ZN, Khan N, et al. Regulatory B cell repertoire defects predispose lung cancer patients to immune‐related toxicity following checkpoint blockade. Nat Commun. 2022;13(1):3148. doi: 10.1038/s41467-022-30863-x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137. Deng J, Wei Y, Fonseca VR, Graca L, Yu D. T follicular helper cells and T follicular regulatory cells in rheumatic diseases. Nat Rev Rheumatol. 2019;15(8):475‐490. doi: 10.1038/s41584-019-0254-2 [DOI] [PubMed] [Google Scholar]
- 138. DeFalco J, Harbell M, Manning‐Bog A, et al. Non‐progressing cancer patients have persistent B cell responses expressing shared antibody paratopes that target public tumor antigens. Clin Immunol. 2018;187:37‐45. doi: 10.1016/j.clim.2017.10.002 [DOI] [PubMed] [Google Scholar]
- 139. Zaenker P, Gray ES, Ziman MR. Autoantibody production in cancer—the humoral immune response toward autologous antigens in cancer patients. Autoimmun Rev. 2016;15(5):477‐483. doi: 10.1016/j.autrev.2016.01.017 [DOI] [PubMed] [Google Scholar]
- 140. Ludwig RJ, Vanhoorelbeke K, Leypoldt F, et al. Mechanisms of autoantibody‐induced pathology. Front Immunol. 2017;8:603. doi: 10.3389/fimmu.2017.00603 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141. Toi Y, Sugawara S, Sugisaka J, et al. Profiling preexisting antibodies in patients treated with anti‐PD‐1 therapy for advanced non‐small cell lung cancer. JAMA Oncol. 2019;5(3):376‐383. doi: 10.1001/jamaoncol.2018.5860 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142. Osorio JC, Ni A, Chaft JE, et al. Antibody‐mediated thyroid dysfunction during T‐cell checkpoint blockade in patients with non‐small‐cell lung cancer. Ann Oncol. 2017;28(3):583‐589. doi: 10.1093/annonc/mdw640 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143. Dora D, Bokhari SMZ, Aloss K, et al. Implication of the gut microbiome and microbial‐derived metabolites in immune‐related adverse events: emergence of novel biomarkers for cancer immunotherapy. Int J Mol Sci. 2023;24(3):2769. doi: 10.3390/ijms24032769 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144. Chen Y, Liu Y, Wang Y, et al. Prevotellaceae produces butyrate to alleviate PD‐1/PD‐L1 inhibitor‐related cardiotoxicity via PPARalpha‐CYP4×1 axis in colonic macrophages. J Exp Clin Cancer Res. 2022;41(1):1. doi: 10.1186/s13046-021-02201-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145. Simpson RC, Shanahan ER, Batten M, et al. Diet‐driven microbial ecology underpins associations between cancer immunotherapy outcomes and the gut microbiome. Nat Med. 2022;28(11):2344‐2352. doi: 10.1038/s41591-022-01965-2 [DOI] [PubMed] [Google Scholar]
- 146. Iraporda C, Romanin DE, Bengoa AA, et al. Local treatment with lactate prevents intestinal inflammation in the TNBS‐induced colitis model. Front Immunol. 2016;7:651. doi: 10.3389/fimmu.2016.00651 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147. Renga G, Nunzi E, Pariano M, et al. Optimizing therapeutic outcomes of immune checkpoint blockade by a microbial tryptophan metabolite. J Immunother Cancer. 2022;10(3):e003725. doi: 10.1136/jitc-2021-003725 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148. Chaput N, Lepage P, Coutzac C, et al. Baseline gut microbiota predicts clinical response and colitis in metastatic melanoma patients treated with ipilimumab. Ann Oncol. 2017;28(6):1368‐1379. doi: 10.1093/annonc/mdx108 [DOI] [PubMed] [Google Scholar]
- 149. Hamada K, Isobe J, Hattori K, et al. Turicibacter and Acidaminococcus predict immune‐related adverse events and efficacy of immune checkpoint inhibitor. Front Immunol. 2023;14:1164724. doi: 10.3389/fimmu.2023.1164724 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150. Inamura K, Hamada T, Bullman S, Ugai T, Yachida S, Ogino S. Cancer as microenvironmental, systemic and environmental diseases: opportunity for transdisciplinary microbiomics science. Gut. 2022. doi: 10.1136/gutjnl-2022-327209 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151. Ogino S, Nowak JA, Hamada T, Milner DA, Nishihara R. Insights into pathogenic interactions among environment, host, and tumor at the crossroads of molecular pathology and epidemiology. Annu Rev Pathol. 2019;14:83‐103. doi: 10.1146/annurev-pathmechdis-012418-012818 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152. Zhou P, Zhao X, Wang G. Risk factors for immune checkpoint inhibitor‐related pneumonitis in cancer patients: a systemic review and meta‐analysis. Respiration. 2022;101(11):1035‐1050. doi: 10.1159/000526141 [DOI] [PubMed] [Google Scholar]
- 153. Huang Y, Soon YY, Aminkeng F, et al. Risk factors for immune‐related adverse events from anti‐PD‐1 or anti‐PD‐L1 treatment in an Asian cohort of nonsmall cell lung cancer patients. Int J Cancer. 2022;150(4):636‐644. doi: 10.1002/ijc.33822 [DOI] [PubMed] [Google Scholar]
- 154. Huang J, Liu D, Wang Y, et al. Ginseng polysaccharides alter the gut microbiota and kynurenine/tryptophan ratio, potentiating the antitumour effect of antiprogrammed cell death 1/programmed cell death ligand 1 (anti‐PD‐1/PD‐L1) immunotherapy. Gut. 2022;71(4):734‐745. doi: 10.1136/gutjnl-2020-321031 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155. Cortellini A, Bersanelli M, Buti S, et al. A multicenter study of body mass index in cancer patients treated with anti‐PD‐1/PD‐L1 immune checkpoint inhibitors: when overweight becomes favorable. J Immunother Cancer. 2019;7(1):57. doi: 10.1186/s40425-019-0527-y [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156. McQuade JL, Daniel CR, Hess KR, et al. Association of body‐mass index and outcomes in patients with metastatic melanoma treated with targeted therapy, immunotherapy, or chemotherapy: a retrospective, multicohort analysis. Lancet Oncol. 2018;19(3):310‐322. doi: 10.1016/S1470-2045(18)30078-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157. Leiter A, Carroll E, De Alwis S, et al. Metabolic disease and adverse events from immune checkpoint inhibitors. Eur J Endocrinol. 2021;184(6):857‐865. doi: 10.1530/eje-20-1362 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158. Stroud CR, Hegde A, Cherry C, et al. Tocilizumab for the management of immune mediated adverse events secondary to PD‐1 blockade. J Oncol Pharm Pract. 2019;25(3):551‐557. doi: 10.1177/1078155217745144 [DOI] [PubMed] [Google Scholar]
- 159. Shimada T, Hoshino Y, Tsunemi T, et al. Neuromyelitis optica spectrum disorder after treatment with pembrolizumab. Mult Scler Relat Disord. 2020;37:101447. doi: 10.1016/j.msard.2019.101447 [DOI] [PubMed] [Google Scholar]
- 160. Xing K, Gu B, Zhang P, Wu X. Dexamethasone enhances programmed cell death 1 (PD‐1) expression during T cell activation: an insight into the optimum application of glucocorticoids in anti‐cancer therapy. BMC Immunol. 2015;16:39. doi: 10.1186/s12865-015-0103-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161. Acharya N, Madi A, Zhang H, et al. Endogenous glucocorticoid signaling regulates CD8(+) T cell differentiation and development of dysfunction in the tumor microenvironment. Immunity. 2020;53(3):658‐671.e6. doi: 10.1016/j.immuni.2020.08.005 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162. Haanen J, Carbonnel F, Robert C, et al. Management of toxicities from immunotherapy: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow‐up. Ann Oncol. 2017;28(suppl 4):iv119‐iv142. doi: 10.1093/annonc/mdx225 [DOI] [PubMed] [Google Scholar]
- 163. Bayless NL, Bluestone JA, Bucktrout S, et al. Development of preclinical and clinical models for immune‐related adverse events following checkpoint immunotherapy: a perspective from SITC and AACR. J Immunother Cancer. 2021;9(9):e002627. doi: 10.1136/jitc-2021-002627 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164. Pang L, Xie M, Ma X, et al. Clinical characteristics and therapeutic effects of checkpoint inhibitor‐related pneumonitis in patients with non‐small cell lung cancer. BMC Cancer. 2023;23(1):203. doi: 10.1186/s12885-023-10649-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165. Tokunaga A, Sugiyama D, Maeda Y, et al. Selective inhibition of low‐affinity memory CD8(+) T cells by corticosteroids. J Exp Med. 2019;216(12):2701‐2713. doi: 10.1084/jem.20190738 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166. Wang Y, Yang M, Tao M, et al. Corticosteroid administration for cancer‐related indications is an unfavorable prognostic factor in solid cancer patients receiving immune checkpoint inhibitor treatment. Int Immunopharmacol. 2021;99:108031. doi: 10.1016/j.intimp.2021.108031 [DOI] [PubMed] [Google Scholar]
- 167. Petrelli F, Signorelli D, Ghidini M, et al. Association of steroids use with survival in patients treated with immune checkpoint inhibitors: a systematic review and meta‐analysis. Cancers (Basel). 2020;12(3):546. doi: 10.3390/cancers12030546 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168. Skribek M, Rounis K, Afshar S, et al. Effect of corticosteroids on the outcome of patients with advanced non‐small cell lung cancer treated with immune‐checkpoint inhibitors. Eur J Cancer. 2021;145:245‐254. doi: 10.1016/j.ejca.2020.12.012 [DOI] [PubMed] [Google Scholar]
- 169. Thompson JA, Schneider BJ, Brahmer J, et al. Management of immunotherapy‐related toxicities, version 1.2019. J Natl Compr Canc Netw. 2019;17(3):255‐289. doi: 10.6004/jnccn.2019.0013 [DOI] [PubMed] [Google Scholar]
- 170. Roberts J, Smylie M, Walker J, et al. Hydroxychloroquine is a safe and effective steroid‐sparing agent for immune checkpoint inhibitor‐induced inflammatory arthritis. Clin Rheumatol. 2019;38(5):1513‐1519. doi: 10.1007/s10067-019-04451-2 [DOI] [PubMed] [Google Scholar]
- 171. Wang Y, Wiesnoski DH, Helmink BA, et al. Fecal microbiota transplantation for refractory immune checkpoint inhibitor‐associated colitis. Nat Med. 2018;24(12):1804‐1808. doi: 10.1038/s41591-018-0238-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172. Chen M, Ma L, Yu H, et al. JK5G postbiotics attenuate immune‐related adverse events in NSCLC patients by regulating gut microbiota: a randomized controlled trial in China. Front Oncol. 2023;13:1155592. doi: 10.3389/fonc.2023.1155592 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173. Liu W, Luo Z, Liu Y, Sun B. Current landscape and tailored management of immune‐related adverse events. Front Pharmacol. 2023;14:1078338. doi: 10.3389/fphar.2023.1078338 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 174. Santoro A, Masini S, Cavina R, et al. Rituximab in steroid‐refractory immune‐related pancreatitis: a case report. Front Oncol. 2023;13:1205720. doi: 10.3389/fonc.2023.1205720 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 175. Araujo DV, Muniz TP, Yang A, et al. Real world outcomes and hepatotoxicity of infliximab in the treatment of steroid‐refractory immune‐related adverse events. Curr Oncol. 2021;28(3):2173‐2179. doi: 10.3390/curroncol28030201 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 176. Jiang C, Zhang L, Xu X, et al. Engineering a smart agent for enhanced immunotherapy effect by simultaneously blocking PD‐L1 and CTLA‐4. Adv Sci (Weinh). 2021;8(20):e2102500. doi: 10.1002/advs.202102500 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177. Simon N, Antignani A, Hewitt SM, Gadina M, Alewine C, FitzGerald D. Tofacitinib enhances delivery of antibody‐based therapeutics to tumor cells through modulation of inflammatory cells. JCI Insight. 2019;4(5):e123281. doi: 10.1172/jci.insight.123281 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 178. Fa'ak F, Buni M, Falohun A, et al. Selective immune suppression using interleukin‐6 receptor inhibitors for management of immune‐related adverse events. J Immunother Cancer. 2023;11(6):e006814. doi: 10.1136/jitc-2023-006814 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 179. Mooradian MJ, Wang DY, Coromilas A, et al. Mucosal inflammation predicts response to systemic steroids in immune checkpoint inhibitor colitis. J Immunother Cancer. 2020;8(1):e000451. doi: 10.1136/jitc-2019-000451 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 180. Arriola E, Wheater M, Karydis I, Thomas G, Ottensmeier C. Infliximab for IPILIMUMAB‐related colitis‐letter. Clin Cancer Res. 2015;21(24):5642‐5643. doi: 10.1158/1078-0432.CCR-15-2471 [DOI] [PubMed] [Google Scholar]
- 181. Dougan M, Wang Y, Rubio‐Tapia A, Lim JK. AGA clinical practice update on diagnosis and management of immune checkpoint inhibitor colitis and hepatitis: expert review. Gastroenterology. 2021;160(4):1384‐1393. doi: 10.1053/j.gastro.2020.08.063 [DOI] [PubMed] [Google Scholar]
- 182. Badran YR, Zou F, Durbin SM, et al. Concurrent immune checkpoint inhibition and selective immunosuppressive therapy in patients with immune‐related enterocolitis. J Immunother Cancer. 2023;11(6):e007195. doi: 10.1136/jitc-2023-007195 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 183. Johnson D, Patel AB, Uemura MI, et al. IL17A blockade successfully treated psoriasiform dermatologic toxicity from immunotherapy. Cancer Immunol Res. 2019;7(6):860‐865. doi: 10.1158/2326-6066.CIR-18-0682 [DOI] [PubMed] [Google Scholar]
- 184. Song M, Liang J, Wang L, et al. IL‐17A functions and the therapeutic use of IL‐17A and IL‐17RA targeted antibodies for cancer treatment. Int Immunopharmacol. 2023;123:110757. doi: 10.1016/j.intimp.2023.110757 [DOI] [PubMed] [Google Scholar]
- 185. van Not OJ, Verheijden RJ, van den Eertwegh AJM, et al. Association of immune‐related adverse event management with survival in patients with advanced melanoma. JAMA Oncol. 2022;8(12):1794‐1801. doi: 10.1001/jamaoncol.2022.5041 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 186. Verheijden RJ, May AM, Blank CU, 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. 2020;26(9):2268‐2274. doi: 10.1158/1078-0432.CCR-19-3322 [DOI] [PubMed] [Google Scholar]
- 187. Suijkerbuijk KPM, Kapiteijn E, Verheijden RJ. Management of immune‐related adverse events affecting outcome in patients treated with checkpoint inhibitors. JAMA Oncol. 2020;6(8):1300‐1301. doi: 10.1001/jamaoncol.2020.1932 [DOI] [PubMed] [Google Scholar]
- 188. Ogusu S, Harutani Y, Tozuka T, et al. Second‐line immunosuppressant administration for steroid‐refractory immune‐related adverse events in patients with lung cancer. Cancer Immunol Immunother. 2023;72(11):3765‐3772. doi: 10.1007/s00262-023-03528-x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 189. Henderson Berg MH, Del Rincon SV, Miller WH. Potential therapies for immune‐related adverse events associated with immune checkpoint inhibition: from monoclonal antibodies to kinase inhibition. J Immunother Cancer. 2022;10(1):e003551. doi: 10.1136/jitc-2021-003551 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 190. Liu Y, Jiang L. Tofacitinib for treatment in immune‐mediated myocarditis: the first reported cases. J Oncol Pharm Pract. 2020:1078155220947141. doi: 10.1177/1078155220947141 [DOI] [PubMed] [Google Scholar]
- 191. Murray K, Floudas A, Murray C, et al. First use of tofacitinib to treat an immune checkpoint inhibitor‐induced arthritis. BMJ Case Rep. 2021;14(2):e238851. doi: 10.1136/bcr-2020-238851 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 192. Bishu S, Melia J, Sharfman W, Lao CD, Fecher LA, Higgins PDR. Efficacy and outcome of tofacitinib in immune checkpoint inhibitor colitis. Gastroenterology. 2021;160(3):932‐934.e3. doi: 10.1053/j.gastro.2020.10.029 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 193. Sweep MWD, Tjan MJH, Gorris MAJ, Bol KF, Westdorp H. Case report: a severe case of immunosuppressant‐refractory immune checkpoint inhibitor‐mediated colitis rescued by tofacitinib. Front Immunol. 2023;14:1212432. doi: 10.3389/fimmu.2023.1212432 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 194. Benesova K, Kraus FV, Carvalho RA, 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(12):1730‐1741. doi: 10.1136/ard-2022-222451 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 195. Marijt KA, Sluijter M, Blijleven L, et al. Metabolic stress in cancer cells induces immune escape through a PI3K‐dependent blockade of IFNgamma receptor signaling. J Immunother Cancer. 2019;7(1):152. doi: 10.1186/s40425-019-0627-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 196. Huang F, Goncalves C, Bartish M, et al. Inhibiting the MNK1/2‐eIF4E axis impairs melanoma phenotype switching and potentiates antitumor immune responses. J Clin Invest. 2021;131(8):e140752. doi: 10.1172/JCI140752 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 197. Haselmayer P, Camps M, Liu‐Bujalski L, et al. Efficacy and pharmacodynamic modeling of the BTK inhibitor evobrutinib in autoimmune disease models. J Immunol. 2019;202(10):2888‐2906. doi: 10.4049/jimmunol.1800583 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 198. Hong D, Rasco D, Veeder M, et al. A phase 1b/2 study of the Bruton tyrosine kinase inhibitor ibrutinib and the PD‐L1 inhibitor durvalumab in patients with pretreated solid tumors. Oncology. 2019;97(2):102‐111. doi: 10.1159/000500571 [DOI] [PubMed] [Google Scholar]
- 199. Shultz LD, Keck J, Burzenski L, et al. Humanized mouse models of immunological diseases and precision medicine. Mamm Genome. 2019;30(5‐6):123‐142. doi: 10.1007/s00335-019-09796-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 200. Medetgul‐Ernar K, Davis MM. Standing on the shoulders of mice. Immunity. 2022;55(8):1343‐1353. doi: 10.1016/j.immuni.2022.07.008 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 201. Wong CK, Lam TH, Liao SY, Lau YM, Tse HF, So BYF. Immunopathogenesis of immune checkpoint inhibitor induced myocarditis: insights from experimental models and treatment implications. Biomedicines. 2023;11(1):107. doi: 10.3390/biomedicines11010107 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 202. Jing Y, Chen X, Li K, et al. Association of antibiotic treatment with immune‐related adverse events in patients with cancer receiving immunotherapy. J Immunother Cancer. 2022;10(1):e003779. doi: 10.1136/jitc-2021-003779 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 203. Jing Y, Yang J, Johnson DB, Moslehi JJ, Han L. Harnessing big data to characterize immune‐related adverse events. Nat Rev Clin Oncol. 2022;19(4):269‐280. doi: 10.1038/s41571-021-00597-8 [DOI] [PubMed] [Google Scholar]
- 204. Atsavapranee ES, Billingsley MM, Mitchell MJ. Delivery technologies for T cell gene editing: applications in cancer immunotherapy. EBioMedicine. 2021;67:103354. doi: 10.1016/j.ebiom.2021.103354 [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Data Availability Statement
Not applicable.
