Abstract
Although immune checkpoint inhibitors have greatly improved cancer therapy, they also cause immune-related adverse events, including a wide range of inflammatory side effects resulting from excessive immune activation. Types of immune-related adverse events are diverse and can occur in almost any organ, with different frequencies and severities. Furthermore, immune-related adverse events may occur within the first few weeks after treatment or even several months after treatment discontinuation. Predictive biomarkers include blood cell counts and cell surface markers, serum proteins, autoantibodies, cytokines/chemokines, germline genetic variations and gene expression profiles, human leukocyte antigen genotype, microRNAs and the gut microbiome. Given the inconsistencies in research results and limited practical utility, there is to date no established biomarker that can be used in routine clinical practice, and additional investigations are essential to demonstrate efficacy and subsequently facilitate integration into routine clinical use.
Keywords: immune checkpoint inhibitor, immune-related adverse event, biomarker
There is to date no established biomarker for immune-related adverse events resulted from immune checkpoint inhibitors in routine clinical practice, and further investigations are indispensable to validate efficacy and accuracy.
Introduction
In recent years, there has been considerable progress in cancer treatment through use of immune checkpoint inhibitors (ICIs). The Pharmaceuticals and Medical Devices Agency in Japan has approved several such agents for various cancers. These include anti-cytotoxic T-lymphocyte antigen 4 (anti-CTLA-4) antibodies (ipilimumab and tremelimumab), anti-programmed death 1 (anti-PD-1) antibodies (nivolumab, pembrolizumab and cemiplimab) and anti-programmed death 1 ligand (anti-PD-L1) antibodies (durvalumab, atezolizumab and avelumab). Cytotoxic T cells (CTLs) express inhibitory receptors, including CTLA-4 and PD-1, that interact with cluster differential 80 (CD80)/cluster differential 86 (CD86) and PD-L1 expressed on antigen-presenting or tumor cells. These interactions lead to suppression of the T-cell immune response and help tumor cells to evade T-cell-mediated cell death (1,2). ICIs targeting the CTLA-4 or PD-1/PD-L1 axis prevent binding between these ligands and receptors from impeding signal transduction and therefore enhance the antitumor immune response.
Although use of ICIs has lead to favorable clinical outcomes, it also results in a broad range of inflammatory side effects due to excessive immune activation, known as immune-related adverse events (irAEs) (3). Although the precise mechanisms of irAEs are still unknown, several potential mechanisms include heightened T-cell activity targeting antigens presented in both tumor and healthy tissues, along with escalated inflammatory cytokine levels. Types of irAEs are diverse and can occur in almost any organ and tissue, with different frequencies and severities (4). The most common irAEs often involve the skin, gastrointestinal tract, endocrine glands and liver; they are often difficult to treat and might worsen quality of life (QoL) (5). Pneumonitis and myocarditis are rare, occurring in 0.3 to 1.3% of cases, but can be fatal (6). Furthermore, irAEs may occur within the first few weeks after treatment or even several months after treatment discontinuation (3,7). Given the variation of organs and tissues of origin, severity, and uncertainty of onset, biomarkers for predicting irAEs and managing them as early as possible are highly desired, helping to maintain effective treatment of patients and their QoL.
There are currently no definitive biomarkers for routine clinical practice, and efficacy and accuracy will need to be validated. Here, we review the literature on various types of potential predictive biomarkers for irAEs (Figure 1).
Figure 1.
Biomarkers for organ-specific irAEs. Abbreviations: irAEs, immune-related adverse events; HLA, human leukocyte antigen; IL, interleukin; G-CSF, granulocyte colony-stimulating; GM-CSF, granulocyte-macrophage colony-stimulating; MCP-1, monocyte chemoattractant protein-1.
Blood cell counts and cell surface markers
Because routine laboratory tests of blood counts and blood cell ratios are easily accessible, inexpensive and simple, clinicians are inclined to use them as biomarkers for irAEs. However, most studies to date are retrospective, had inconsistent cutoff values, and may contain many biases, and the results were difficult to interpret (Table 1).
Table 1.
Blood cell count and ratio biomarkers for predicting irAEs
Parameter | Study design (no. patients) | Tumor type | irAE type | Association |
---|---|---|---|---|
Cell counts | ||||
WBC count | Retrospective (n = 101) | Melanoma | Lung and gastrointestinal irAEs | High WBC counts after treatment were associated with high risk of grade 3–4 lung or gastrointestinal irAEs (OR, 1.20; P = 0.014) (8) |
Neutrophil count | Retrospective (n = 150) | NSCLC | All | Low absolute neutrophil counts at baseline and when irAEs occurred were associated with high risk of grade 3–4 irAEs (univariate analysis: P = 0.009; P = 0.000281, respectively) (9) |
Neutrophil-to-lymphocyte ratio | Retrospective (n = 150) | NSCLC | All | Low neutrophil-to-lymphocyte ratio at baseline and when irAEs occurred were associated with high risk of grade 3–4 irAEs (univariate analysis: P = 0.023; P = 0.011, respectively) (9) |
Retrospective (n = 470) | Various cancers | All | Baseline neutrophil-to-lymphocyte ratio ≤ 5.3 was associated with high risk of any-grade irAEs (OR, 2.07; P = 0.01) (10) | |
Prospective (n = 1187) | Solid and blood cancers | All | High baseline neutrophil-to-lymphocyte ratio was associated with high risk of grade 4 or worse irAEs (univariate analysis: P = 0.0137) (11) | |
Eosinophil count | Retrospective (n = 105) | Various cancers | All | High baseline absolute eosinophil counts >0.175 × 109/L were associated with high risk of any-grade irAEs (OR, 0.29; P = 0.02) (12) |
Retrospective (n = 533) | Solid tumors | Liver injury | Baseline absolute eosinophil counts ≥130 μ/L were associated with high risk of grade 2 or worse immune-related liver injury (HR, 3.01; P = 0.012) (13) | |
Eosinophil | Retrospective (n = 149) | NSCLC | All | Baseline percentage of eosinophil ≥1.15 was associated with high risk of any-grade irAEs (OR, 8.30; P = 0.003) (14) |
Monocyte count | Retrospective (n = 470) | Various cancers | All | Baseline absolute monocyte counts >0.29 × 103/μL were associated with high risk of any-grade irAEs (OR, 2.34; P = 0.03) (10) |
Monocyte-to-lymphocyte ratio | Retrospective (n = 470) | Various cancers | All | Baseline monocyte-to-lymphocyte ratio ≤ 0.73 was associated with high risk of any-grade irAEs (OR, 2.96; P = 0.001) (10) |
Lymphocyte count | Retrospective (n = 470) | Various cancers | All | Baseline absolute lymphocyte counts >2.6 × 103/μL were associated with high risk of any-grade irAEs (OR, 4.30; P = 0.002) (10) |
Retrospective (n = 171) | NSCLC | All | Absolute lymphocyte counts >820/mm3 at 2 weeks after treatment initiation were associated with high risk of any-grade irAEs (OR, 3.58; P = 0.007) (15) | |
Retrospective (n = 226) | Solid tumors | All | Low absolute lymphocyte counts when irAEs occurred were associated with high risk of grade 3–4 irAEs (univariate analysis: P = 0.005) (16) | |
Retrospective (n = 101) | Melanoma | Lung and gastrointestinal irAEs | High relative lymphocyte counts after treatment were associated with high risk of grade 3–4 lung or gastrointestinal irAEs (OR, 1.65; P = 0.012) (8) | |
Retrospective (n = 105) | Various cancers | All | Low baseline relative lymphocyte counts were associated with high risk of any-grade irAEs (OR, 3.6; P = 0.027) (12) | |
Platelet count | Retrospective (n = 470) | Various cancers | All | Baseline platelet counts >145 × 109/L were associated with high risk of any-grade irAEs (OR, 2.23; P = 0.03) (10) |
Retrospective (n = 150) | NSCLC | All | Low platelet counts at baseline were associated with high risk of grade 3–4 irAEs (univariate analysis: P = 0.023) (9) | |
Retrospective (n = 105) | Various cancers | All | Low baseline platelet counts were associated with high risk of any-grade irAEs (OR, 2.77; P = 0.025) (12) | |
Platelet-to-lymphocyte ratio | Retrospective (n = 150) | NSCLC | All | Low platelet-to-lymphocyte ratio at baseline and when irAEs occurred were associated with high risk of grade 3–4 irAEs (univariate analysis: P = 0.0016) and grade 1–2 irAEs (univariate analysis: P = 0.011), respectively (9) |
Retrospective (n = 470) | Various cancers | All | Baseline platelet-to-lymphocyte ratio ≤ 534 was associated with high risk of any-grade irAEs (OR, 5.05; P = 0.04) (10) | |
Retrospective (n = 184) | NSCLC | All | Baseline platelet-to-lymphocyte ratio < 180 was associated with high risk of any-grade irAEs (OR, 2.3; P = 0.027) (17) | |
Cell surface markers | ||||
CD8+ T-cell | Retrospective (n = 40) | Various cancers | All | Low baseline CD8+ T-cell counts were associated with high risk of any-grade irAEs (OR, 0.934, P = 0.012) (12) |
Regulatory T-cell | Prospective (n = 26) | Melanoma | Colitis | Low baseline median regulatory T-cell proportion was associated with high risk of any-grade colitis (univariate analysis: P = 0.018) (19) |
CD4 memory T-cell and T-cell receptor | Retrospective (n = 71) | Melanoma | All | Baseline activated CD4 memory T-cell abundance and T-cell receptor diversity correlated with severe irAE development; increased T-cell receptor clonality during treatment correlated with severe irAE development and early irAE onset time (20) |
CD19+ B-cell | Retrospective (n = 40) | Various cancers | All | High baseline proportion of CD19+ B cells was associated with high risk of any-grade irAEs (OR, 15.87, P = 0.02) (12) |
CD21lo B-cell | Prospective (n = 23) | Renal cell carcinoma | All | Low baseline proportion of CD21lo B cells was associated with high risk of any-grade irAEs (univariate analysis: P < 0.01) (21) |
Note: Data were calculated by multivariate analysis, unless otherwise specified; P < 0.05 was statistically significant.
Abbreviations: irAE, immune-related adverse event; WBC, white blood cell; OR, odds ratio; NSCLC, non-small cell lung cancer; HR; hazard ratio.
Many studies have shown that blood cell counts and ratios might be predictive factors of irAE occurrence (8–17). Most indicate that elevated blood cell counts, especially lymphocyte counts and a corresponding decrease in the individual blood cell-to-lymphocyte ratio, at baseline are associated with increased risk of irAEs. However, as is often the case, there are conflicting and inconsistent reports (9,11,12,16). Of particular interest, in a retrospective study of patients with solid tumors, those who experienced grade 3 or higher irAEs had significantly decreased lymphocyte counts at irAE onset (8–12,15,16), suggesting that changes in blood counts during treatment, as well as at baseline, may be important.
It has been reported that ipilimumab leads to increased T-cell repertoire diversification before irAE onset, with greater diversity of CD4+ and CD8+ T-cells in patients with irAEs compared with those without irAEs (18), suggesting that subtypes of T-cells might act as biomarkers to predict the occurrence of irAEs. A retrospective analysis suggested that a low level of CD8+ T-cells at baseline predicts the occurrence of irAEs (odds ratio (OR), 0.934, 95% confidence interval (CI) 0.885–0.985, P = 0.012) (12). Similarly, patients with melanoma experiencing ipilimumab-related colitis tend to have a high level of CD4+ T-cells (P = 0.053) and a low proportion of regulatory T-cells (P = 0.018) at baseline (19). In contrast, no significant difference in absolute regulatory T-cell counts at baseline between groups with or without colitis has been reported. A study on T-cell characteristics associated with irAEs in patients with melanoma reported that activated CD4 memory T-cell abundance and T-cell receptor diversity at baseline correlated with severe irAE development. In addition, increased T-cell receptor clonality during treatment was associated with severe irAE development and onset time (20). In addition, baseline levels of several subtypes of B cells, such as CD19+ B-cells and B-cells with low CD21 expression (CD21lo B-cells), are significantly associated with high risk of irAEs (12,21).
Serum proteins
Similar to blood counts, serum proteins are measured by routine laboratory tests and may be promising biomarkers for both general and organ-specific irAEs. However, these protein levels are susceptible to many factors, including cancer-related inflammation and cancer development, which inevitably limits their specificity as biomarkers.
C-reactive protein (CRP) has been well studied, but results are inconsistent. A retrospective cohort study of 67 patients with primary liver cancer found a transient increase in CRP upon irAE onset; at baseline, however, there was no difference between patients with any grade irAEs and those without (22). Another retrospective study of 37 patients with melanoma treated with ICIs and tyrosine kinase inhibitors showed that CRP dramatically increased in the majority of patients (93%) when irAEs occurred (23). Nevertheless, it has also been reported that a low level of pretreatment CRP is independently associated with irAE occurrence (24). Similarly, results for other serum proteins, such as lactate dehydrogenase and albumin, are also inconsistent (12,25,26).
Some proteins may function as biomarkers of organ-specific irAEs. For example, higher levels of creatine phosphokinase, aspartate aminotransferase (AST) and alanine aminotransferase (ALT) after ICI therapy initiation correlate well with greater risk of ICI-induced myocarditis (27), and cardiac troponin and natriuretic peptides are frequently elevated when ICI-induced myocarditis is diagnosed (28). In a review of ICI-mediated diarrhea and colitis, fecal lactoferrin and calprotectin were reported to serve as useful surrogate markers in patients with ICI-mediated diarrhea and colitis, as in those with inflammatory bowel disease (29). In addition, decreased free thyroxine after ICI therapy initiation has been suggested to be an early biomarker of ICI-induced hypophysitis (30). Elevated levels of thyroglobulin after ICI therapy initiation and thyroid-stimulating hormone at baseline have been associated with ICI-induced thyroid dysfunction (31–33).
Autoantibodies
Some organ-specific autoantibodies are often associated with irAEs in relevant organs; thus, the presence of autoantibodies at baseline is associated with the incidence or severity of irAEs associated with that organ. However, rather than predicting the occurrence of irAEs, autoantibodies may simply contribute to preemptive diagnosis of an immune-related disease present prior to ICI therapy. Further investigation is needed to clarify the association between autoantibody detection and irAE development.
Many studies have demonstrated the association between preexisting autoantibodies and the occurrence of irAEs in patients with multiple cancers (32,34–38). For instance, patients positive for anti-nuclear (ANA) or anti-thyroid antibodies at baseline might be at higher risk of developing organ-nonspecific irAEs than those without (34,38). Anti-pituitary antibodies and anti-thyroid antibodies are positively associated with adrenocorticotropic hormone (ACTH) deficiency and thyroid dysfunction, respectively (39–41). Pituitary endocrine cells express CTLA-4, which can be blocked by CTLA-4 inhibitors, leading to activation of complement components and pituitary gland damage (42). ICI-induced thyroiditis is a T lymphocyte-mediated process with elevated intrathyroidal CD8+ and CD4−CD8− T lymphocytes (43). A possible interpretation is that T cells strengthen PD-1 antibody efficacy and, in turn, induce B cells to produce autoantibodies, leading to irAE occurrence and indicating that the presence of preexisting autoantibodies predicts irAEs (44). In addition to levels at baseline, obvious titer increases in thyroglobulin and thyroid peroxidase autoantibodies after starting ICI therapy and rheumatoid factor positivity at any time correlate with thyroid dysfunction and rheumatic irAEs (31,45).
Conversely, some studies have found that preexisting antibodies do not increase risk of irAE occurrence (46,47). Indeed, one retrospective study demonstrated no association between those baseline antibody titers and irAE severity (46). It has also been reported that although preexisting ANA at baseline is not associated with irAE rates, its appearance correlates positively with irAE severity (47).
In a retrospective analysis involving pretreatment serum samples from 333 patients with metastatic melanoma undergoing ICI therapy, it was observed that heightened levels of anti-MAGEB4 antibodies correlate with increased incidences of irAEs but that elevated anti-FGFR1 antibody levels are linked to decreased irAE occurrences (48).
There are both advantages and disadvantages when considering the use of ICI therapy in patients known to be autoantibody positive or diagnosed with an autoimmune disease. Historically, such patients were routinely excluded from clinical trials, and safety data were limited (49). However, recent reports have indicated that the safety of ICI treatment in these patients is comparable with that of others (50,51). In addition, studies have suggested that a combination of immunosuppressants can be safely used alongside ICI treatment without compromising its effectiveness (50). Therefore, we believe that ICI therapy can be considered a viable option for patients known to be autoantibody positive or diagnosed with an autoimmune disease.
Cytokines/chemokines
Cytokines and chemokines play a crucial role in multiple inflammatory reactions and the tumor microenvironment (52,53). Therefore, they might serve as promising biomarkers for irAEs. However, findings regarding the correlation between cytokines/chemokines and occurrence of irAEs are currently vary and are inconsistent. Many studies have investigated and demonstrated the value of specific cytokines that can amplify both pro- and anti-inflammatory immune responses as predictive biomarkers for identifying an individual’s risk of developing irAEs (53). Among pro-inflammatory cytokines, the interleukin-1 (IL-1) family is associated with innate immune responses, with IL-1α and IL-1β exhibiting the highest inflammatory activity (54). A retrospective study showed that an elevated baseline level of IL-1α was significantly associated with the incidence of myositis in advanced gastrointestinal cancer patients (55). In another prospective study focusing on biomarkers of irAEs in non-small cell squamous carcinoma (NSCLC) patients, the occurrence of irAEs correlated with a higher baseline level of IL-1β (56). Similarly, concerning organ-specific irAEs, it has been suggested that the baseline level of IL-1β is significantly higher in those with than without thyroid dysfunction (31).
As a proinflammatory cytokine, systemic release of IL-6 has been implicated in the mechanism driving irAEs (57). In line with this, it has been proposed that a higher baseline level of IL-6 can be considered a risk factor for organ-specific irAEs, including thyroiditis and colitis (55). However, a different study involving melanoma patients reported the opposite finding, namely, that a lower baseline level of IL-6 correlated significantly with higher risk of irAEs (58). When combined with CRP, an increased level of IL-6 is considered a screening parameter for early detection of irAEs (59).
Through analysis of cytokine expression in melanoma patients experiencing irAEs, it was discovered that patients who develop pneumonitis and grade 3 gastrointestinal irAEs have an increased baseline level of IL-17 (60,61). There are several other proinflammatory cytokines that may serve as biomarkers for irAEs. For instance, higher baseline levels of IL-12p70 and leukemia inhibitory factor (LIF) are associated with myositis, granulocyte-macrophage colony-stimulating factor is linked to rash, and interferon-gamma (IFN-γ) is relevant to various irAEs (14,55,56). Conversely, it was revealed that a low baseline level of granulocyte colony-stimulating factor and an early decrease in IL-8 are significantly linked to the occurrence of irAEs such as thyroid dysfunction and colitis (31,60).
As typical anti-inflammatory cytokines, IL-4 and IL-10 have value as risk factors for irAE occurrence. It was shown that patients with ICI-related rash had higher baseline levels of IL-4 than those without this irAE (55). Moreover, elevated levels of IL-10 both at baseline and after the first cycle of immunotherapy correlated positively with onset of irAEs, especially in the case of pneumonitis (56,62).
As a category of cytokines capable of causing directional cell movement, chemokines are potential biomarkers for predicting irAE occurrence. For example, compared with patients who did not experience any neurological irAEs, those with high-grade irAEs showed an increased baseline level of monocyte chemoattractant protein-1 (MCP-1) (63). Furthermore, a significant association was observed between an early decrease in MCP-1 level during treatment and occurrence of thyroid irAEs (31). A longitudinal analysis of the relationship between cytokines/chemokines and irAEs revealed low baseline levels of C-X-C motif chemokine ligand 9 (CXCL9), CXCL10, CXCL11 and CXCL19 in patients who experienced irAEs. In addition, these patients had large increases in CXCL9 and CXCL10 levels after treatment (64). CXCL9, CXCL10 and CXCL11 are chemotactic factors for activated T cells and involved in T-cell recruitment, which indicates that elevated levels of these chemokines might lead to broad T-cell activation, thereby inducing irAE occurrence. CXCL13, functioning as a B-cell chemoattractant and playing a role in multiple autoimmune and inflammatory diseases, shows a significant association with irAE occurrence, whereby high baseline and treatment-related levels of CXCL13 are significantly linked to irAE occurrence (65).
Furthermore, there are additional potential cytokine/chemokine-related biomarkers capable of predicting the risk of irAEs. These include high baseline levels of angiogenin, angiopoietin-1, CD40L and B-cell-activating factor as well as increased levels of regulated upon activation, normal T cell expressed and secreted (RANTES) and soluble cluster of differentiation 163 during treatment (56,60,66–68).
Genetic variations and gene expression profile
Because gene variants and differential gene expression can contribute to susceptibility to irAEs, there is great promise in utilizing multigene panels to assess an individual’s risk of developing irAEs.
Several studies have highlighted the association between single-nucleotide polymorphisms (SNPs) in the PD-1 protein-encoding gene programmed cell death 1 (PDCD1) and the frequency and severity of irAEs (Table 2). For example, among patients with NSCLC, those who carry the homozygous PDCD1 804C > T (rs2227981) gene variant were found to be less likely to experience irAEs of any grade (69). Conversely, patients with metastatic renal cell carcinoma carrying the PD-1.6 G allele (rs10204525) are more prone to developing more severe and diverse irAEs than those with the AA genotype (70). In a genome-wide study, 12 SNPs were identified as risk factors for irAE occurrence; 18 other SNPs were protective factors (71). It has been suggested that various genetic alterations, including small variations and copy number variations in genes, such as SMAD family member 3 (SMAD3), PR/SET domain 1 (PRDM1), interleukin 1 receptor antagonist (IL1RN), CD274, solute carrier organic anion transporter family member 1B1 (SLCO1B1), thyroid stimulating hormone receptor (TSHR) and FANCD2 and FANCI associated nuclease 1 (FAN1), are associated with irAEs, especially some organ-specific irAEs, including hepatitis and encephalitis (72).
Table 2.
Gene-related biomarkers for predicting irAEs
Gene | Study design (no. patients) |
Tumor type | irAE type | Association |
---|---|---|---|---|
Genetic variation | ||||
PDCD1 | Prospective (n = 322) | NSCLC | All | The homozygous genotype PDCD1 804C > T (rs2227981) was associated with low risk of any-grade irAEs (OR, 0.4; P = 0.039) (69) |
PDCD1 | Retrospective (n = 106) | Renal cell carcinoma | All | The PD-1.6 G allele of PDCD1 (rs10204525) was associated with high risk of grade 2 or worse (OR, 3.39; P = 0.003) and more various (OR, 2.778; P = 0.031) irAEs (70) |
Spleen associated tyrosine kinase (SYK) | Prospective (n = 95) | Melanoma | All | The SYK T/T genotype (rs7036417) was associated with high risk of grade 3 or 4 irAEs (OR, 7.46; P = 0.000143) (94) |
IL-17 | Prospective (n = 214) | Melanoma | All | The A allele of IL-17 (rs16906115) and was associated with high risk of grade 3 or worse irAEs (OR, 2.24; P = 0.046) (95) |
Uracil DNA glycosylase (UNG), interferon omega 1 (IFNW1), PD-L1, interferon lambda 4 (IFNL4) and CTLA4 | Retrospective (n = 94) | Various cancers | All | UNG (rs246079), IFNW1 (rs10964859), PD-L1 (rs4143815), IFNL4 (rs12979860) and CTLA4 (rs3087243) were significantly associated with irAEs (univariate analysis) (96) |
Mitogen-activated protein kinase 1 (MAPK1), protein tyrosine phosphatase receptor type C (PTPRC), adenosine deaminase domain containing 1 (ADAD1) and IL6 | Prospective (n = 340) | Solid tumors | All | The C allele of MAPK1 (rs3810610) was associated with high risk of any-grade irAE (OR, 1.495; P = 0.012); the A allele of PTPRC (rs6428474) was associated with low risk of any-grade irAE (OR, 0.717; P = 0.041); the A allele of ADAD1 (rs17388568) was associated with high risk of severe irAEs (OR, 2.599; P = 0.003); the G allele of IL6 (rs1800796) was associated with low risk of severe irAEs (OR, 0.425; P = 0.018) (97) |
Multiple genes | Retrospective (n = 89) | Melanoma | All | 12 SNPs were risk factors for irAE occurrence; 18 SNPs were protective (71) |
SMAD3, PRDM1, IL1RN, CD274, SLCO1B1, TSHR and FAN1 | Prospective (n = 95) | Melanoma | All | VARs of SMAD3 were associated with pancreatitis; CNVs of PRDM1 and IL1RN were associated with irAEs; CNVs of CD274 and SLCO1B1 were associated with hepatitis; CNVs of PRDM1 and CD274 were associated with encephalitis; CNVs of PRDM1, CD274, TSHR and FAN1 were associated with myositis (univariate analysis) (72) |
Gene expression | ||||
LCP1 and ADPGK | Retrospective (n = 18 706) | Various cancers | All | High expression of LCP1 (P = 0.008) and ADPGK (P = 0.01) were associated with any-grade irAEs (tumor tissues; univariate analysis) (73) |
CD3 epsilon subunit of T-cell receptor complex (CD3E), interleukin 2 receptor subunit gamma (IL2RG), CD4, CD37 and IL-32 | Prospective (n = 162) | Melanoma | Gastrointestinal irAEs |
High baseline expression of CD3E, IL2RG, CD4, CD37 and IL-32 was associated with any-grade irAEs (blood samples; repeated measures analysis of variance) (74) |
C-X-C motif chemokine receptor 1 (CXCR1) | Retrospective (n = 355) | Melanoma | All | High expression of CXCR1 pre- (P = 0.0034) and posttreatment (P < 0.001) was associated with low risk of irAEs (blood samples; univariate analysis) (75) |
16 genes | Prospective (n = 150) | Melanoma | Diarrhea/Colitis | Variations in expression levels of 16 specific genes were associated with diarrhea/colitis severity (blood samples; univariate analysis) (76) |
HLA | ||||
HLA-Cw12, HLA-DR15, HLA-DQ7 and HLA-DPw9 | Retrospective (n = 62) | Various cancers | Endocrine irAEs | HLA-Cw12, HLA-DR15, HLA-DQ7 and HLA-DPw9 were associated with isolated adrenocorticotropic hormone deficiency; HLA-Cw12 and HLA-DR15 were associated with hypophysitis (univariate analysis: all P < 0.05) (39) |
HLA-DRB1*11:01 and HLA-DQB1*03:01 | Prospective (n = 102) | NSCLC and melanoma |
All | HLA-DRB1*11:01 was associated with pruritus (OR, 4.53; P = 0.0021), and HLA-DQB1*03:01 was associated with colitis (OR, 3.94; P = 0.017) (78) |
HLA-B*27:05 | Prospective (n = 5) | Breast and bladder cancers | Encephalitis | HLA-B*27:05 was associated with encephalitis (OR, 59.1; P < 0.001) (79) |
HLA-DR1, HLA-DR4, HLA-DR8 and HLA-DR15 | Retrospective (n = 132) | Melanoma | All | HLA-DR1 was associated with pneumonitis, HLA-DR4 was associated with type 1 diabetes and hepatitis, HLA-DR8 was associated with hypothyroidism, and HLA-DR15 was associated with hypophysitis (univariate analysis: all P < 0.01 except for hepatitis: P < 0.05) (80) |
HLA-DRB1*04:01, HLA-DQB1*03:01 and HLA-DRB1*15:01 | Prospective (n = 179) | NSCLC | All | HLA-DRB1*04:01 was associated with high risk of irAEs (RR, 1.55; P = 0.011; HLA-DQB1*03:01 was associated with low risk of colitis (RR, 0.18; P = 0.029), and HLA-DRB1*15:01 was associated with low risk of arthralgia (P = 0.048) (81) |
HLA-DRB3*01:01, HLA-DPB1*04:02 and HLA-A*26:01 | Retrospective (n = 530) | Various cancers | All | HLA-DRB3*01:01 was associated with thrombocytopenia (OR, 3.48; P = 0.011); HLA-DPB1*04:02 was associated with hypokalemia/hyponatremia (OR, 3.44; P = 0.009), leukopenia (OR, 2.1; P = 0.037), and anemia (OR, 2.33; P = 0.026); HLA-A*26:01 was associated with bilirubin elevation (OR, 2.67; P = 0.037) (82) |
Note: Data were calculated by multivariate analysis, unless otherwise specified.
Abbreviations:VARs, small variations; CNVs, copy number variations; HLA, human leucocyte antigen; RR, relative risk.
Specific gene expression signatures also serve as valuable biomarkers for irAEs (73–76). In a notable example, a large-scale retrospective multiomics analysis involving various cancers revealed that patients who experience irAEs show high expression levels of certain genes. One such gene is lymphocyte cytosolic protein 1 (LCP1), which encodes lymphocyte cytosolic protein 1 and is involved in T-cell activation. Another gene is adenosine diphosphate dependent glucokinase (ADPGK), encoding adenosine diphosphate–dependent glucokinase and playing a role in mediating metabolic shifts during T-cell activation (73). These gene expression patterns provide valuable insights into the underlying mechanisms of irAEs and may serve as predictive markers for their occurrence.
Human leucocyte antigen genotypes
Human leukocyte antigen (HLA) molecules are expressed on the surface of immune cells and play a crucial role in presenting peptide ligands to T-cell receptors. With a growing body of research indicating associations between certain HLA genotypes and organ-specific irAEs, it is reasonable to view HLA genotype as a potential biomarker for predicting organ-specific irAEs.
HLA genes are known to be highly polymorphic, and certain specific variations in these genes can act as biomarkers for autoimmune diseases (77). Similar to autoimmune diseases, it has been suggested that HLA gene variations may influence the occurrence of irAEs. This viewpoint is supported by multiple studies that have identified significant associations between specific HLA variations and irAEs, especially in the context of some organ-specific irAEs (Table 2) (39,78–82). For example, it has been reported that patients with HLA-DR15 are at a high risk of experiencing isolated ACTH deficiency and hypophysitis (39,80). Indeed, the relationship between HLA-DQB1*3:01 and colitis appears to be contradictory. One study suggested that patients with HLA-DQB1*3:01 are more susceptible to developing colitis (78), whereas another study indicated that the same genotype might offer protection against colitis (81). This highlights the complexity of the relationship between HLA gene variations and irAEs, underlining the need for further research to refine these associations for clinical application.
microRNAs
Although microRNAs (miRNAs) have gained interest in recent years, only a limited number of studies have established a significant association between specific miRNAs and irAEs. Indeed, this area of research is still in its early stages, and further investigations into the potential of miRNAs as biomarkers will be necessary in the future.
miR-146a, a member of the small, double-stranded, noncoding RNA family, plays a critical role as a negative regulator of inflammation and autoimmunity (83). In preclinical studies involving mice lacking miR-146a, the mice developed more severe irAEs than wild-type mice (84). This finding was verified by a clinical study in which patients with clear cell renal cell carcinoma who experienced grade 3 or 4 irAEs presented significantly reduced expression of exosomal miR-146a (85). Moreover, these two studies explored the impact of the MIR146A variant (rs2910164) on the severity of irAEs, consistently reporting that patients with the CC genotype were more likely to develop grade 3 or 4 irAEs. The role of exosomal miRNA-34a-5p in inducing cardiac senescence-related injury has also been revealed through experiments involving mouse models treated with PD-1 inhibitors (86). This finding supports the potential of miRNA-34a-5p as a biomarker for cardiac irAEs.
The gut microbiome
The gut microbiome is a critical component of human physiology that influences both health and disorders such as inflammation, suggesting that it may impact irAE occurrence (87). Given that the gut microbiota might serve as a biomarker for irAEs, modifying it through interventions such as antibiotics, probiotics and fecal microbiota transplantation might offer a way to mitigate irAEs.
Nonetheless, the function of the gut microbiome as a biomarker for irAEs remains unclear because of inconsistent study results for numerous gut bacteria, which might not be easily reproducible and are difficult to interpret. For example, one study reported that patients with irAEs had reduced abundance of Agathobacter, though another study suggested that Agathobacter might be linked to more severe irAEs (88,89). As another limitation, collecting fecal samples from patients for analysis can be a tedious process.
Body mass index
In recent years, several studies have provided evidence indicating that a high body mass index (BMI, kg/m2) is a predictive biomarker for irAEs (90). In comparison to patients with normal weight, those with BMI of ≥25 kg/m2 have a higher occurrence of irAEs of any grade. In addition, a retrospective study investigated the impact of weight and factors related to metabolic syndrome on irAEs, suggesting that individuals with overweight and few metabolic comorbidities are more likely to experience grade 2 or worse irAEs (91), though the mechanism remains unclear. Different metabolic statuses might exert opposite influences on PD-1 expression in T cells, resulting in complicated alterations in risk of irAEs.
Future perspectives
With the rapid development and widespread use of ICIs, irAEs and their prediction have become a major concern. Despite considerable research efforts to explore potential biomarkers for irAEs, there is no established biomarker that can be used in routine clinical practice. This situation can be attributed to several factors. On one hand, routine laboratory tests, such as blood cell counts, ratios and serum proteins, are simple, feasible and cost-effective. However, studies investigating these markers as irAE predictors are largely retrospective in nature. On the other hand, evaluating complicated biological parameters such as cytokines/chemokines, genetic variations and gene expression is expensive and often not clinically available. Moreover, findings supporting their use as predictive biomarkers are sometimes inconsistent and lack specificity due to cancer-related inflammation and cancer development (92). This may contribute to the uncertainty of these biomarkers in predicting irAEs. Given these limitations, drawing definitive conclusions about the predictive value of these biomarkers for irAEs remains challenging. More large-scale prospective studies are required to confirm their ability to predict irAEs. Additional investigations are essential to demonstrate efficacy and subsequently facilitate integration into routine clinical use.
It is known that the development of irAEs is associated with better outcomes after ICI therapy across various types of cancers (93). It is quite plausible that biomarkers for irAEs might also predict the efficacy of ICI. For instance, baseline serum cytokine levels might predict the development of irAEs as well as the efficacy of ICI in NSCLC patients (56). Therefore, future research needs to go beyond merely predicting irAEs and, instead, explore ideal biomarkers that can simultaneously predict a lower risk of irAEs and higher treatment efficacy.
Conclusion
Biomarkers that can predict the occurrence and severity of irAEs play a pivotal role in enhancing our ability to predict and manage these adverse events. Early identification and intervention are crucial for minimizing the impact of irAEs on patients undergoing ICI therapies. Many potential biomarkers have been proposed, but their sensitivity and specificity need to undergo validation through further studies before they can be confidently applied in various clinical settings.
Funding
This work was financially supported by JST SPRING (Grant Number JPMJSP2125). Yao Liang would like to take this opportunity to thank the ‘Interdisciplinary Frontier Next-Generation Researcher Program of the Tokai Higher Education and Research System.’
Conflict of interest statement
Dr Yuichi Ando reports grants and personal fees from Chugai Pharmaceutical Co, Ltd, grants and personal fees from Kyowa Kirin Co, Ltd, grants and personal fees from Nippon Kayaku Co, Ltd, grants and personal fees from Yakult Honsha Co, Ltd, personal fees from Eli Lilly Japan K.K., personal fees from Ono Pharmaceutical Co, Ltd, grants and personal fees from Taiho Pharmaceutical Co, Ltd, grants and personal fees from Novartis Pharma K.K., personal fees from Bayer Holding Ltd, personal fees from Sawai Pharmaceutical Co., Ltd, grants and personal fees from Daiichi Sankyo Company, Ltd, grants from Eisai Co, Ltd, personal fees from MSD K.K., personal fees from Astellas Pharma Inc, personal fees from Otsuka Holdings Co, Ltd, personal fees from Sanwa Kagaku Kenkyusho Co, Ltd, personal fees from Hisamitsu Pharmaceutical Co, Inc, personal fees from SymBio Pharceuticals, personal fees from Aptitude Health, grants from BeiGene, Ltd, personal fees from Alfresa Pharma Corporation, personal fees from Sumitomo Pharma Co. Sumitomo Pharma Co., Ltd, outside the submitted work.
Contributor Information
Yao Liang, Department of Clinical Oncology and Chemotherapy, Nagoya University Hospital, Nagoya, Aichi, Japan.
Osamu Maeda, Department of Clinical Oncology and Chemotherapy, Nagoya University Hospital, Nagoya, Aichi, Japan.
Yuichi Ando, Department of Clinical Oncology and Chemotherapy, Nagoya University Hospital, Nagoya, Aichi, Japan.
References
- 1. Bagchi S, Yuan R, Engleman EG. Immune checkpoint inhibitors for the treatment of cancer: clinical impact and mechanisms of response and resistance. Annu Rev Pathol 2021;16:223–49. 10.1146/annurev-pathol-042020-042741. [DOI] [PubMed] [Google Scholar]
- 2. Dine J, Gordon R, Shames Y, Kasler MK, Barton-Burke M. Immune checkpoint inhibitors: an innovation in immunotherapy for the treatment and management of patients with cancer. Asia Pac J Oncol Nurs 2017;4:127–35. 10.4103/apjon.apjon_4_17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3. Postow MA, Sidlow R, Hellmann MD. Immune-related adverse events associated with immune checkpoint blockade. N Engl J Med 2018;378:158–68. 10.1056/NEJMra1703481. [DOI] [PubMed] [Google Scholar]
- 4. Martins F, Sofiya L, Sykiotis GP, et al. Adverse effects of immune-checkpoint inhibitors: epidemiology, management and surveillance. Nat Rev Clin Oncol 2019;16:563–80. 10.1038/s41571-019-0218-0. [DOI] [PubMed] [Google Scholar]
- 5. Weber JS, Hodi FS, Wolchok JD, et al. Safety profile of nivolumab monotherapy: a pooled analysis of patients with advanced melanoma. J Clin Oncol 2017;35:785–92. 10.1200/JCO.2015.66.1389. [DOI] [PubMed] [Google Scholar]
- 6. Wang DY, Salem J-E, Cohen JV, et al. Fatal toxic effects associated with immune checkpoint inhibitors: a systematic review and meta-analysis. JAMA Oncol 2018;4:1721–8. 10.1001/jamaoncol.2018.3923. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7. Johnson DB, Nebhan CA, Moslehi JJ, Balko JM. Immune-checkpoint inhibitors: long-term implications of toxicity. Nat Rev Clin Oncol 2022;19:254–67. 10.1038/s41571-022-00600-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8. Fujisawa Y, Yoshino K, Otsuka A, et al. Fluctuations in routine blood count might signal severe immune-related adverse events in melanoma patients treated with nivolumab. J Dermatol Sci 2017;88:225–31. 10.1016/j.jdermsci.2017.07.007. [DOI] [PubMed] [Google Scholar]
- 9. 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;13:765–71. 10.2147/CMAR.S293200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10. Michailidou D, Khaki AR, Morelli MP, Diamantopoulos L, Singh N, Grivas P. Association of blood biomarkers and autoimmunity with immune related adverse events in patients with cancer treated with immune checkpoint inhibitors. Sci Rep 2021;11:9029. 10.1038/s41598-021-88307-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. Ruste V, Goldschmidt V, Laparra A, et al. The determinants of very severe immune-related adverse events associated with immune checkpoint inhibitors: a prospective study of the French REISAMIC registry. Eur J Cancer 2021;158:217–24. 10.1016/j.ejca.2021.08.048. [DOI] [PubMed] [Google Scholar]
- 12. Bai R, Chen N, Chen X, et al. Analysis of characteristics and predictive factors of immune checkpoint inhibitor-related adverse events. Cancer Biol Med 2021;18:1118–33. 10.20892/j.issn.2095-3941.2021.0052. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13. Yoshikawa Y, Imamura M, Yamauchi M, et al. Prevalence of immune-related adverse events and anti-tumor efficacy following immune checkpoint inhibitor therapy in Japanese patients with various solid tumors. BMC Cancer 2022;22:1232. 10.1186/s12885-022-10327-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14. Hu W-T, Zhang Q, Zhang Z, et al. Eosinophil and IFN-γ associated with immune-related adverse events as prognostic markers in patients with non-small cell lung cancer treated with immunotherapy. Front Immunol 2023;14:1112409. 10.3389/fimmu.2023.1112409. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15. Egami S, Kawazoe H, Hashimoto H, et al. Absolute lymphocyte count predicts immune-related adverse events in patients with non-small-cell lung cancer treated with nivolumab monotherapy: a multicenter retrospective study. Front Oncol 2021;11:618570. 10.3389/fonc.2021.618570. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16. Haraguchi M, Nakao Y, Narita S, et al. Peripheral lymphocyte fluctuation as an indicator of severe immune-related adverse events in patients treated with immune checkpoint inhibitors. Cancer Med 2023;12:10636–46. 10.1002/cam4.5816. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17. Pavan A, Calvetti L, Dal Maso A, et al. Peripheral blood markers identify risk of immune-related toxicity in advanced non-small cell lung cancer treated with immune-checkpoint inhibitors. Oncologist 2019;24:1128–36. 10.1634/theoncologist.2018-0563. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18. Oh DY, Cham J, Zhang L, et al. Immune toxicities elicted by CTLA-4 blockade in cancer patients are associated with early diversification of the T-cell repertoire. Cancer Res 2017;77:1322–30. 10.1158/0008-5472.CAN-16-2324. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19. 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;28:1368–79. 10.1093/annonc/mdx108. [DOI] [PubMed] [Google Scholar]
- 20. 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;28:353–62. 10.1038/s41591-021-01623-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21. Nishimura K, Konishi T, Ochi T, et al. CD21lo B cells could be a potential predictor of immune-related adverse events in renal cell carcinoma. J Pers Med 2022;12:888. 10.3390/jpm12060888. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22. Yu Y, Wang S, Su N, et al. Increased circulating levels of CRP and IL-6 and decreased frequencies of T and B lymphocyte subsets are associated with immune-related adverse events during combination therapy with PD-1 inhibitors for liver cancer. Front Oncol 2022;12:906824. 10.3389/fonc.2022.906824. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23. Abolhassani A-R, Schuler G, Kirchberger MC, Heinzerling L. C-reactive protein as an early marker of immune-related adverse events. J Cancer Res Clin Oncol 2019;145:2625–31. 10.1007/s00432-019-03002-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24. Mehra T, Dongre K, Boesing M, et al. Pre-treatment comorbidities, C-reactive protein and eosinophil count, and immune-related adverse events as predictors of survival with checkpoint inhibition for multiple tumour entities. Cancer Med 2023;12:12253–62. 10.1002/cam4.5919. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25. Matsuoka H, Hayashi T, Takigami K, et al. Correlation between immune-related adverse events and prognosis in patients with various cancers treated with anti PD-1 antibody. BMC Cancer 2020;20:656. 10.1186/s12885-020-07142-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26. Shimozaki K, Sukawa Y, Sato Y, et al. Analysis of risk factors for immune-related adverse events in various solid tumors using real-world data. Future Oncol 2021;17:2593–603. 10.2217/fon-2020-0861. [DOI] [PubMed] [Google Scholar]
- 27. Vasbinder A, Chen Y, Procureur A, et al. Biomarker trends, incidence, and outcomes of immune checkpoint inhibitor-induced myocarditis. JACC CardioOncol 2022;4:689–700. 10.1016/j.jaccao.2022.11.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28. Pirozzi F, Poto R, Aran L, et al. Cardiovascular toxicity of immune checkpoint inhibitors: clinical risk factors. Curr Oncol Rep 2021;23:13. 10.1007/s11912-020-01002-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29. Gong Z, Wang Y. Immune checkpoint inhibitor–mediated diarrhea and colitis: a clinical review. JCO Oncol Pract 2020;16:453–61. 10.1200/OP.20.00002. [DOI] [PubMed] [Google Scholar]
- 30. Seejore K, Giannoudi M, Osborn D, et al. Characterisation of the onset and severity of adrenal and thyroid dysfunction associated with CTLA4-related hypophysitis. Eur J Endocrinol 2021;186:83–93. 10.1530/EJE-21-0760. [DOI] [PubMed] [Google Scholar]
- 31. Kurimoto C, Inaba H, Ariyasu H, et al. Predictive and sensitive biomarkers for thyroid dysfunctions during treatment with immune-checkpoint inhibitors. Cancer Sci 2020;111:1468–77. 10.1111/cas.14363. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32. Kobayashi T, Iwama S, Yamagami A, et al. Elevated TSH level, TgAb, and prior use of ramucirumab or TKIs as risk factors for thyroid dysfunction in PD-L1 blockade. J Clin Endocrinol Metab 2022;107:e4115–23. 10.1210/clinem/dgac467. [DOI] [PubMed] [Google Scholar]
- 33. 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:e3704–13. 10.1210/clinem/dgab263. [DOI] [PubMed] [Google Scholar]
- 34. Castel-Ajgal Z, Goulvestre C, Zaibet S, et al. Preexisting autoantibodies and immune related adverse events in metastatic urothelial carcinoma patients treated by pembrolizumab. Clin Genitourin Cancer 2022;20:e362–8. 10.1016/j.clgc.2022.04.002. [DOI] [PubMed] [Google Scholar]
- 35. Iwama S, Kobayashi T, Yasuda Y, et al. Increased risk of thyroid dysfunction by PD-1 and CTLA-4 blockade in patients without thyroid autoantibodies at baseline. J Clin Endocrinol Metab 2022;107:e1620–30. 10.1210/clinem/dgab829. [DOI] [PubMed] [Google Scholar]
- 36. Johannet P, Liu W, Fenyo D, et al. Baseline serum autoantibody signatures predict recurrence and toxicity in melanoma patients receiving adjuvant immune checkpoint blockade. Clin Cancer Res 2022;28:4121–30. 10.1158/1078-0432.CCR-22-0404. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37. Daban A, Gonnin C, Phan L, et al. Preexisting autoantibodies as predictor of immune related adverse events (irAEs) for advanced solid tumors treated with immune checkpoint inhibitors (ICIs). Oncoimmunology 2023;12:2204754. 10.1080/2162402X.2023.2204754. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38. Hsu S-J, Chao Y-C, Lin X-H, et al. Antinuclear antibody (ANA) status predicts immune-related adverse events in liver cancer patients undergoing anti-PD-1 therapy. Clin Exp Immunol 2023;212:239–48. 10.1093/cei/uxad036. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39. 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:e002493. 10.1136/jitc-2021-002493. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40. Muir CA, Wood CCG, Clifton-Bligh RJ, et al. Association of antithyroid antibodies in checkpoint inhibitor-associated thyroid immune-related adverse events. J Clin Endocrinol Metab 2022;107:e1843–9. 10.1210/clinem/dgac059. [DOI] [PubMed] [Google Scholar]
- 41. Izawa N, Shiokawa H, Onuki R, et al. The clinical utility of comprehensive measurement of autoimmune disease-related antibodies in patients with advanced solid tumors receiving immune checkpoint inhibitors: a retrospective study. ESMO Open 2022;7:100415. 10.1016/j.esmoop.2022.100415. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42. Iwama S, De Remigis A, Callahan MK, Slovin SF, Wolchok JD, Caturegli P. Pituitary expression of CTLA-4 mediates hypophysitis secondary to administration of CTLA-4 blocking antibody. Sci Transl Med 2014;6:230ra45. [DOI] [PubMed] [Google Scholar]
- 43. Kotwal A, Gustafson MP, Bornschlegl S, et al. Immune checkpoint inhibitor-induced thyroiditis is associated with increased intrathyroidal T lymphocyte subpopulations. Thyroid 2020;30:1440–50. 10.1089/thy.2020.0075. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44. 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;5:376–83. 10.1001/jamaoncol.2018.5860. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45. Mathias K, Rouhani S, Olson D, Bass AR, Gajewski TF, Reid P. Association between rheumatic autoantibodies and immune-related adverse events. Oncologist 2023;28:440–8. 10.1093/oncolo/oyac252. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46. Ghosh N, Postow M, Zhu C, et al. Lower baseline autoantibody levels are associated with immune-related adverse events from immune checkpoint inhibition. J Immunother Cancer 2022;10:e004008. 10.1136/jitc-2021-004008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47. Alserawan L, Anguera G, Zamora Atenza C, et al. Association between changes in the patterns of antinuclear autoantibodies during immune checkpoint inhibition therapy and the development of severe immune related adverse events. Int J Mol Sci 2022;23:12641. 10.3390/ijms232012641. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48. Hassel JC, Zucht H-D, Mangana J, et al. Autoantibodies as predictors for survival and immune-related adverse events in checkpoint inhibition therapy of metastasized melanoma. J Clin Oncol 2020;38:10011. 10.1200/JCO.2020.38.15_suppl.10011. [DOI] [Google Scholar]
- 49. Hodi FS, O'Day SJ, McDermott DF, et al. Improved survival with ipilimumab in patients with metastatic melanoma. N Engl J Med 2010;363:711–23. 10.1056/NEJMoa1003466. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50. Haanen J, Ernstoff MS, Wang Y, et al. Autoimmune diseases and immune-checkpoint inhibitors for cancer therapy: review of the literature and personalized risk-based prevention strategy. Ann Oncol 2020;31:724–44. 10.1016/j.annonc.2020.03.285. [DOI] [PubMed] [Google Scholar]
- 51. Abdel-Wahab N, Shah M, Lopez-Olivo MA, Suarez-Almazor ME. Use of immune checkpoint inhibitors in the treatment of patients with cancer and preexisting autoimmune disease: a systematic review. Ann Intern Med 2018;168:121–30. 10.7326/M17-2073. [DOI] [PubMed] [Google Scholar]
- 52. Märkl F, Huynh D, Endres S, Kobold S. Utilizing chemokines in cancer immunotherapy. Trends Cancer 2022;8:670–82. 10.1016/j.trecan.2022.04.001. [DOI] [PubMed] [Google Scholar]
- 53. Kang JH, Bluestone JA, Young A. Predicting and preventing immune checkpoint inhibitor toxicity: targeting cytokines. Trends Immunol 2021;42:293–311. 10.1016/j.it.2021.02.006. [DOI] [PubMed] [Google Scholar]
- 54. Stefania S, Colia R, Cinzia R, Corrado A, Cantatore FP. Off-label use of anti-IL-1 drugs in rheumatic diseases. Int J Immunopathol Pharmacol 2021;35:1–19. 10.1177/20587384211006584. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55. Wang Y, Zou J, Li Y, et al. Serological biomarkers predict immune-related adverse events and clinical benefit in patients with advanced gastrointestinal cancers. Front Immunol 2022;13:987568. 10.3389/fimmu.2022.987568. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56. Schindler H, Lusky F, Daniello L, et al. Serum cytokines predict efficacy and toxicity, but are not useful for disease monitoring in lung cancer treated with PD-(L)1 inhibitors. Front Oncol 2022;12:1010660. 10.3389/fonc.2022.1010660. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57. Zhou Y, Medik YB, Patel B, et al. Intestinal toxicity to CTLA-4 blockade driven by IL-6 and myeloid infiltration. J Exp Med 2023;220:e20221333. 10.1084/jem.20221333. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58. Valpione S, Pasquali S, Campana LG, et al. Sex and interleukin-6 are prognostic factors for autoimmune toxicity following treatment with anti-CTLA4 blockade. J Transl Med 2018;16:1–10. 10.1186/s12967-018-1467-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59. Husain B, Kirchberger MC, Erdmann M, et al. Inflammatory markers in autoimmunity induced by checkpoint inhibitors. J Cancer Res Clin Oncol 2021;147:1623–30. 10.1007/s00432-021-03550-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60. Tyan K, Baginska J, Brainard M, et al. Cytokine changes during immune-related adverse events and corticosteroid treatment in melanoma patients receiving immune checkpoint inhibitors. Cancer Immunol Immunother 2021;70:2209–21. 10.1007/s00262-021-02855-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61. Tarhini AA, Zahoor H, Lin Y, et al. Baseline circulating IL-17 predicts toxicity while TGF-β1 and IL-10 are prognostic of relapse in ipilimumab neoadjuvant therapy of melanoma. J Immunother Cancer 2015;3:1–6. 10.1186/s40425-015-0081-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62. Wang H, Zhou F, Zhao C, et al. Interleukin-10 is a promising marker for immune-related adverse events in patients with non-small cell lung cancer receiving immunotherapy. Front Immunol 2022;13:840313. 10.3389/fimmu.2022.840313. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63. Möhn N, Mahjoub S, Duzzi L, et al. Monocyte chemoattractant protein 1 as a potential biomarker for immune checkpoint inhibitor-associated neurotoxicity. Cancer Med 2023;12:9373–83. 10.1002/cam4.5695. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64. Khan S, Khan SA, Luo X, et al. Immune dysregulation in cancer patients developing immune-related adverse events. Br J Cancer 2019;120:63–8. 10.1038/s41416-018-0155-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65. Tsukamoto H, Komohara Y, Tomita Y, et al. Aging-associated and CD4 T-cell-dependent ectopic CXCL13 activation predisposes to anti-PD-1 therapy-induced adverse events. Proc Natl Acad Sci U S A 2022;119:e2205378119. 10.1073/pnas.2205378119. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66. Inoue Y, Inui N, Karayama M, et al. Serum immune modulators associated with immune-related toxicities and efficacy of atezolizumab in patients with non-small cell lung cancer. J Cancer Res Clin Oncol 2022;149:2963–74. 10.1007/s00432-022-04193-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67. Oyanagi J, Koh Y, Sato K, et al. Predictive value of serum protein levels in patients with advanced non-small cell lung cancer treated with nivolumab. Lung Cancer (Amsterdam, Netherlands) 2019;132:107–13. 10.1016/j.lungcan.2019.03.020. [DOI] [PubMed] [Google Scholar]
- 68. Fujimura T, Sato Y, Tanita K, et al. Serum levels of soluble CD163 and CXCL5 may be predictive markers for immune-related adverse events in patients with advanced melanoma treated with nivolumab: a pilot study. Oncotarget 2018;9:15542–51. 10.18632/oncotarget.24509. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69. Bins S, Basak EA, el Bouazzaoui S, et al. Association between single-nucleotide polymorphisms and adverse events in nivolumab-treated non-small cell lung cancer patients. Br J Cancer 2018;118:1296–301. 10.1038/s41416-018-0074-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70. Kobayashi M, Numakura K, Hatakeyama S, et al. Severe immune-related adverse events in patients treated with nivolumab for metastatic renal cell carcinoma are associated with PDCD1 polymorphism. Genes 2022;13:1204. 10.3390/genes13071204. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71. Abdel-Wahab N, Diab A, Yu RK, et al. Genetic determinants of immune-related adverse events in patients with melanoma receiving immune checkpoint inhibitors. Cancer Immunol Immunother 2021;70:1939–49. 10.1007/s00262-020-02797-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72. Wölffer M, Battke F, Schulze M, et al. Biomarkers associated with immune-related adverse events under checkpoint inhibitors in metastatic melanoma. Cancer 2022;14:302. 10.3390/cancers14020302. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73. Jing Y, Liu J, Ye Y, et al. Multi-omics prediction of immune-related adverse events during checkpoint immunotherapy. Nat Commun 2020;11:4946. 10.1038/s41467-020-18742-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74. Shahabi V, Berman D, Chasalow SD, et al. Gene expression profiling of whole blood in ipilimumab-treated patients for identification of potential biomarkers of immune-related gastrointestinal adverse events. J Transl Med 2013;11:75. 10.1186/1479-5876-11-75. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75. Ye W, Olsson-Brown A, Watson RA, et al. Checkpoint-blocker-induced autoimmunity is associated with favourable outcome in metastatic melanoma and distinct T-cell expression profiles. Br J Cancer 2021;124:1661–9. 10.1038/s41416-021-01310-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76. Friedlander P, Wood K, Wassmann K, Christenfeld AM, Bhardwaj N, Oh WK. A whole-blood RNA transcript-based gene signature is associated with the development of CTLA-4 blockade-related diarrhea in patients with advanced melanoma treated with the checkpoint inhibitor tremelimumab. J Immunother Cancer 2018;6:90. 10.1186/s40425-018-0408-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77. Dendrou CA, Petersen J, Rossjohn J, Fugger L. HLA variation and disease. Nat Rev Immunol 2018;18:325–39. 10.1038/nri.2017.143. [DOI] [PubMed] [Google Scholar]
- 78. 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. 10.1016/j.ejca.2018.11.009. [DOI] [PubMed] [Google Scholar]
- 79. Chang H, Shin YW, Keam B, Kim M, Im SA, Lee ST. HLA-B27 association of autoimmune encephalitis induced by PD-L1 inhibitor. Ann Clin Transl Neurol 2020;7:2243–50. 10.1002/acn3.51213. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80. Akturk HK, Couts KL, Baschal EE, et al. Analysis of human leukocyte antigen DR alleles, immune-related adverse events, and survival associated with immune checkpoint inhibitor use among patients with advanced malignant melanoma. JAMA Netw Open 2022;5:e2246400. 10.1001/jamanetworkopen.2022.46400. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81. Abed A, Law N, Calapre L, et al. Human leucocyte antigen genotype association with the development of immune-related adverse events in patients with non-small cell lung cancer treated with single agent immunotherapy. Eur J Cancer 2022;172:98–106. 10.1016/j.ejca.2022.05.021. [DOI] [PubMed] [Google Scholar]
- 82. Jiang N, Yu Y, Zhang M, et al. Association between germ-line HLA and immune-related adverse events. Front Immunol 2022;13:952099. 10.3389/fimmu.2022.952099. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83. Boldin MP, Taganov KD, Rao DS, et al. miR-146a is a significant brake on autoimmunity, myeloproliferation, and cancer in mice. J Exp Med 2011;208:1189–201. 10.1084/jem.20101823. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84. Marschner D, Falk M, Javorniczky NR, et al. MicroRNA-146a regulates immune-related adverse events caused by immune checkpoint inhibitors. JCI Insight 2020;5:e132334. 10.1172/jci.insight.132334. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85. Ivanova E, Asadullina D, Rakhimov R, et al. Exosomal miRNA-146a is downregulated in clear cell renal cell carcinoma patients with severe immune-related adverse events. Noncoding RNA Res 2022;7:159–63. 10.1016/j.ncrna.2022.06.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86. Xia W, Chen H, Chen D, Ye Y, Xie C, Hou M. PD-1 inhibitor inducing exosomal miR-34a-5p expression mediates the cross talk between cardiomyocyte and macrophage in immune checkpoint inhibitor–related cardiac dysfunction. J Immunother Cancer 2020;8:e001293. 10.1136/jitc-2020-001293. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87. Wang Y, Jenq RR, Wargo JA, Watowich SS. Microbiome influencers of checkpoint blockade-associated toxicity. J Exp Med 2023;220:e20220948. 10.1084/jem.20220948. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88. Liu X, Tang H, Zhou Q, et al. Gut microbiota composition in patients with advanced malignancies experiencing immune-related adverse events. Front Immunol 2023;14:1109281. 10.3389/fimmu.2023.1109281. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89. Hakozaki T, Richard C, Elkrief A, et al. The gut microbiome associates with immune checkpoint inhibition outcomes in patients with advanced non-small cell lung cancer. Cancer Immunol Res 2020;8:1243–50. 10.1158/2326-6066.CIR-20-0196. [DOI] [PubMed] [Google Scholar]
- 90. Cortellini A, Bersanelli M, Santini D, et al. Another side of the association between body mass index (BMI) and clinical outcomes of cancer patients receiving programmed cell death protein-1 (PD-1)/programmed cell death-ligand 1 (PD-L1) checkpoint inhibitors: a multicentre analysis of immune-related adverse events. Eur J Cancer 2020;128:17–26. 10.1016/j.ejca.2019.12.031. [DOI] [PubMed] [Google Scholar]
- 91. Leiter A, Carroll E, De Alwis S, et al. Metabolic disease and adverse events from immune checkpoint inhibitors. Eur J Endocrinol 2021;184:857–65. 10.1530/EJE-20-1362. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92. Greten FR, Grivennikov SI. Inflammation and cancer: triggers, mechanisms, and consequences. Immunity 2019;51:27–41. 10.1016/j.immuni.2019.06.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93. Das S, Johnson DB. Immune-related adverse events and anti-tumor efficacy of immune checkpoint inhibitors. J Immunother Cancer 2019;7:306. 10.1186/s40425-019-0805-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94. Ferguson R, Chat V, Morales L, et al. Germline immunomodulatory expression quantitative trait loci (ieQTLs) associated with immune-related toxicity from checkpoint inhibition. Eur J Cancer (Oxford England: 1990) 2023:112923. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95. Taylor CA, Watson RA, Tong O, et al. IL7 genetic variation and toxicity to immune checkpoint blockade in patients with melanoma. Nat Med 2022;28:2592–600. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96. Refae S, Gal J, Ebran N, et al. Germinal Immunogenetics predict treatment outcome for PD-1/PD-L1 checkpoint inhibitors. Invest New Drugs. 2020;38:160–71. [DOI] [PubMed] [Google Scholar]
- 97. Xin Z, You L, Na F, et al. Immunogenetic variations predict immune-related adverse events for PD-1/PD-L1 inhibitors. Eur J Cancer (Oxford England: 1990) 2023;184:124–36. [DOI] [PubMed] [Google Scholar]