You're using an outdated browser. Please upgrade to a modern browser for the best experience.
Immune-Related Adverse Events for Anti-Cancer Immunotherapy
Edit

Immune checkpoint inhibitors (ICIs) have changed how we think about tumor management. Combinations of anti-programmed death ligand-1 (PD-L1) immunotherapy have become the standard of care in many advanced-stage cancers, including as a first-line therapy. Aside from improved anti-tumor immunity, the mechanism of action of immune checkpoint inhibitors (ICIs) exposes a new toxicity profile known as immune-related adverse effects (irAEs). This novel toxicity can damage any organ, but the skin, digestive and endocrine systems are the most frequently afflicted. Most ICI-attributed toxicity symptoms are mild, but some are severe and necessitate multidisciplinary side effect management. Obtaining knowledge on the various forms of immune-related toxicities and swiftly changing treatment techniques to lower the probability of experiencing severe irAEs has become a priority in oncological care.

irAEs ICI anti-PD1 immunotherapy gut microbiome

1. Introduction

Immune checkpoint inhibitors (ICI) and their combination with conventional chemotherapy and/or radiotherapy are currently the standard of care in cancer, and the number of patients receiving ICI as a first-line therapy is rising. Currently, the CTLA-4 inhibitor ipilimumab; PD-1 inhibitors pembrolizumab, nivolumab, and cemiplimab; and PD-L1 inhibitors atezolizumab, avelumab, and durvalumab are approved by the US Food and Drug Administration (FDA) for the treatment of metastatic melanoma, non-small cell lung cancer (NSCLC), Hodgkin’s lymphoma, head and neck squamous cell carcinoma, urothelial carcinoma, renal cell carcinoma, and hepatocellular carcinoma [1]. PD-L1 inhibitors are also approved for the treatment of triple-negative breast cancer and Merkel cell tumors [2].
ICIs have now been in use for a decade and, in addition to their positive effects, there is growing evidence of their side effects. The efficacy of ICIs, including their toxicity, has been demonstrated in murine models to have a significant link to the gut microbiome [3]. All aspects of the gut, including the microbiome, micro-organism composition, and metabolic products, induce effects on host immune defenses [4]; however, in healthy individuals, tolerance and antigenic response are generally in balance. In recent years, numerous independent research groups discovered an intriguing link between the gut flora and ICI efficacy in a clinical setting [5][6]. Multiple investigations and an impartial meta-analysis in malignant melanoma demonstrated a connection between microbial metagenomic and metatranscriptomic markers and ICI efficacy [7][8][9]. Despite the fact, that causality and the biological background of such a connection are still disputed and under scrutiny, multiple studies have shown at least partial or indirect evidence regarding such mechanisms. The most likely supported hypothesis is the “molecular mimicry” phenomenon, where epitopes produced by gut microbial species as part of their native gene expression programs resemble tumor neoantigens, prompting “autoreactive” T cells and powerful anti-tumor immunity [10][11]
Similar to other anti-cancer therapies, ICI comes with its own set of adverse effects; about 70–90% of patients receiving ICI show immune-related adverse events (irAEs) [12][13][14][15]. irAEs are commonly attributed to the inability of the immune system to distinguish between tumors and self-antigens, hence the cross-reactivity [16]. This dysregulation of the immune system’s self-tolerance leads to an over-activation of innate and adaptive immunity that translates to cytokine-mediated inflammation, autoreactive B and T cells, and autoantibodies [17][18]. irAEs can be observed in almost all organs of the human body, more commonly the skin, thyroid, lungs, colon, liver, and pituitary are affected; however, in rare cases even the nervous and cardiac systems can be damaged, with the possibility of a fatal outcome [18][19]. The mechanisms underlying these toxicities are diverse and differ in each organ; in contrast, irAEs can have similar manifestations in the affected organ regardless of the cancer type [20][21][22].

2. Adverse Events and Their Clinical Management in Anti-Cancer Immunotherapy

irAEs are classified by the American International Cancer Institute according to the Common Terminology Criteria for Adverse Events (CTCAE), which is divided into mild (1), moderate (2), severe (3), life-threatening (4), and fatal (5) categories [23]. PD-1 and PD-L1 inhibitors are generally better tolerated than CTLA-4 inhibitors and grade 3 and 4 irAEs are also more common with CTLA-4 inhibitors than with PD-1 inhibitors (31% vs. 10%). There is also a difference in the organ specificity of adverse events, for example, colitis, hypophysitis, and skin rashes occur more frequently with CTLA-4 inhibitors, while pneumonia, hyper- or hypothyroidism, joint pain, and vitiligo are more common with PD-1 inhibitors [23]. Generally, at least one negative event is detected in two-thirds of anti-PD-1/PD-L1 immunotherapies. One-seventh of patients experience a high-grade irAE (3–5) and 0.45% of patients die, most commonly due to pneumonitis [24]. The most common symptoms of treatment-associated irAEs (at any grade) were fatigue, pruritus, diarrhea, hyperthyroidism, hypothyroidism, and vitiligo; while the most common high-grade irAE symptoms were fatigue, liver enzyme elevation (AST and ALT), pneumonitis and diarrhea [13][15][25]. Figure 1 demonstrates the most common and severe irAEs organized by organ systems.
Figure 1. Most common and significant irAEs of PD-1/PD-L1 inhibitors. Figure shows the most common side effects organized by organ system. The most common treatment-associated systemic irAE at any grade was fatigue. Among all irAEs, diarrhea and thyroid dysfunction (hypo- or hyperthyroidism) occurred most frequently. Pneumonitis was the most common high-grade symptom and most frequent cause of irAE-related death.

2.1. Dermatological irAEs

Dermatological toxicity is one of the most common irAEs among all organ systems, occurring in 25–30% of treated patients. Clinically, immune-related cutaneous toxic effects are mainly skin rashes, pruritus, and vitiligo [26][27] the latter being a side effect specific to the treatment of melanoma. It has been shown that the treatment of skin tumors is most frequently associated with the highest incidence of dermatological side effects (7.3 times more frequent than other tumors, [28]). Interestingly, the presence of cutaneous irAEs showed a positive correlation with treatment response and long-term relapse-free survival, according to a recent meta-analysis [27]. Topical agents or low-dose systemic steroids are often sufficient to treat skin-related irAEs [29].

2.2. Gastrointestinal irAEs

Pooled meta-analysis data suggest that diarrhea (13% vs. 33%) and colitis (1.4% vs. 9.1%) are less frequent with PD-1/PD-L1 inhibitors compared to CTLA-4 therapy, but symptoms persist longer (2 months vs. 1.4 months) [30][31][32]. Unlike CTLA-4 therapy, the severity of diarrhea as an adverse effect of PD-1/L1-blockade is not dose-dependent [33]. The most common symptom of colitis is diarrhea, which occurs in almost all patients, followed by abdominal pain (25%), loss of appetite (19%), hematochezia (12.5%) and mucoid stools (10%). GI pathology includes inflammatory infiltrates, villi shortening, and crypt or mucosal fragility. GI irAEs present clinically as described above—colitis, ileitis, decreased transit time, diarrhea, and/or blood in stool (ICI-therapy associated colitis/ileitis, CIC) [34][35]. The risk of colitis is increased by taking NSAIDs and is decreased in the case of vitamin D supplementation [30]. Autoimmune diseases increase the risk of developing CIC, and CIC occurrence in underlying IBD patients receiving anti-CTLA4 therapy was reported at 30% [36][37]. In contrast, IBD patients better tolerate anti-PD1/PD-L1 ICI [31].
Low-grade GI toxicity is usually managed based on clinical symptoms. The gold standard for the diagnosis of CIC is biopsies, but to exclude a more severe situation, such as toxic megacolon or perforation, CT scans are required. An infection can be excluded by obtaining a stool sample for culture [31]. Treatment of colitis in grade 1 is supportive, with antimotility agents (loperamide), hydration, and an appropriate diet to reduce symptoms, which can be continued during therapy. In the case of grade 2, systemic steroid treatment should commence, after the exclusion of infection. In grades 3 and 4, immediate discontinuation of ICI therapy is required, and hospital admission for monitoring is recommended, along with IV administration of 1–2 mg/kg/day methylprednisolone with electrolyte and fluid therapy. If symptoms do not improve after 2–4 days, biological therapy (infliximab and vedolizumab) should be administered. While therapy should not be restarted at grade 4, opinions differ at grade 3, but if symptoms can be managed and reduced to grade 1 or below, PD-1/PD-L1 therapy may be restarted [33]. In grades 3–4 cases, 1–1.5% of patients develop colon perforation, which is treated with emergency colon resection surgery [30].
The most common hepatic irAE is hepatitis, with around 1–2% occurrence in PD-1/PD-L1 monotherapy, but this can be as high as 20% in a combination of ICI therapy with ipilimumab [37]. Blood testing of liver enzymes is essential before therapy initiation, as is monitoring ICI throughout the therapy and before each dose [38]. Treatment in the first two grades is based on continuous monitoring of enzyme levels, and at grade 2, initiation of oral steroid therapy. In grades 3 and 4, glucocorticoids are given in a high dose for the first two days, followed by 1–2 mg/kg/day of oral prednisolone until enzyme levels settle [32]. To date, there are some published case reports of pancreas involvement [39][40] but research has shown that PD-1/PD-L1 inhibitors do not increase the risk of pancreatitis [41].

2.3. Respiratory irAEs

Most fatal adverse events are caused by pneumonitis (35%) [42] compared to other irAEs, but the severe, life-threatening grade is found in less than 2% of cases [38]. The real-world incidence of immune-related pneumonitis in ICI-treated patients is between 2.49% and 13.2% [43][44][45]. The most common symptoms are a persistent, unproductive cough, dyspnea, fever, and chest pain; however, one-third of patients remain asymptomatic [46]. Risk factors include elderly age, smoking, male gender, previous lung disease (emphysema, COPD, or asthma), previous chest irradiation, and combination with other therapies [42]. Diagnosis is based on a CT scan and clinical symptoms. Bronchoscopy or BAL helps with differential diagnosis to rule out an infection [47].
Treatment of ICI-related grade 1 pneumonitis includes the temporary suspension of the drug until the normalization of CT findings. Grade 2 treatments include 1 mg/kg/day of oral prednisolone and in grades 3–4, complete abandonment of ICI therapy is required with 2–4 mg/kg/day IV methylprednisolone for 4–6 weeks and initiation of antibiotic therapy to rule out infection [47].

2.4. Endocrine irAEs

Hypothyroidism, hyperthyroidism, and hyperglycemia are the most common endocrine irAEs. Grade 3 or higher side effects are mostly due to hypoglycemia, adrenal insufficiency and Type 1 DM [25]. Approximately 40–50% of patients experience some change in thyroid function during therapy [46][48][49]. Subclinical hyperthyroidism occurs in 18%, overt hyperthyroidism in 12%, subclinical hypothyroidism in 5% and overt hypothyroidism in 3% of all cases [46]. A total of 7.7% of patients who develop persistent hypothyroidism require levothyroxine administration [48]. Onset of overt thyrotoxicosis occurred after a median of 5 weeks of the receipt of first cycle. Combination ICI therapy, female gender, and younger age were strongly associated with the development of overt thyrotoxicosis [46]. Intriguingly, even in real-world practice, ICI treatment-induced thyroid dysfunction was associated with better outcomes [50][51][52].
Immunotherapeutic monoclonal antibodies can damage healthy tissues directly, for example, anti-CTLA4 treatment directly damages the pituitary [53]. The incidence of hypophysitis is 1–18% in metastatic melanoma patients treated with anti-CTLA4, and 0.5–1.5% for PD-1 inhibitors [54][55]. However, the incidence can rise to up to 13% in the case of combination therapy [56][57]. There have also been cases of PD-1/PD-L1 therapy-induced diabetes (0.2%). Although uncommon, 81% of cases occurred with diabetic ketoacidosis and, unlike thyroiditis, all cases developed fully even with prednisolone usage [58]. Generally, endocrine side effects are always treated with hormone replacement and non-steroidal therapy after the acute phase [59].

2.5. Cardiovascular irAEs

Cardiovascular side effects of ICI therapy pose the greatest challenge to physicians and include myocarditis, pericardial disease, supraventricular arrhythmia, and vasculitis. Anti-PD1 antibodies can directly damage heart tissue, causing concomitant myositis and rhabdomyolysis and robust infiltration of macrophages and T cells [60][61][62][63]. A systematic review reported that the most frequent underlying biological mechanisms were the recruitment of CD4+ and CD8+ T cells, autoantibody-mediated cardiotoxicity and substantial inflammatory cytokine release [64]. Supraventricular arrhythmias are almost always associated with cardiac irAEs and are therefore thought to be secondary events [65]. In a review of several meta-analyses, myocarditis was found to be among the less common irAEs at about 1% prevalence, but has an extremely high mortality (27–46%) compared to others [66]. Risk factors include elderly age (70–80 years), male sex, ethnicity, and pre-existing autoimmune or cardiovascular disease [67][68]. Symptoms usually occur in the form of palpitations, dyspnea, and left ventricular pump dysfunction, as well as chest pain, hypotension, lower limb edema or heart block, but less specific symptoms such as fatigue, malaise, ptosis, diplopia, paresis, nausea, and vomiting can also dominate. In severe cases, hemodynamic instability, cardiogenic shock, and sudden death may happen [66][67].
Close cardiological monitoring and temporary drug suspension are considered in asymptomatic cardiac involvement due to asymptomatic arrhythmias or structural abnormalities of the myocardial wall. If symptoms are present, discontinuation of therapy is recommended, then, depending on improvement, restarting treatment can be considered [38]. If myocarditis is suspected, immediate hospital admission is necessary and high dose of corticosteroid should be administered as the first choice. In severe or life-threatening cases, pulsatile methylprednisolone at 1 g/day for 3–5 days is recommended and, in refractory cases, mycophenolate mofetil or tacrolimus should be used. If the patient has symptoms of chronic heart failure, β-blockers and ACE inhibitor/ATR2 antagonists are administered as treatment [66].

2.6. Musculoskeletal irAEs

Musculoskeletal side effects caused by PD-1/PD-L1 inhibitors occur in between 2 and 12% of cases, with around one in 15 patients requiring rheumatological treatment [69]. The most common presentations are arthralgia, inflammatory arthritis, sicca syndrome, myositis, vasculitis, and polymyalgia rheumatica. Treatment with NSAIDs is recommended for grade 1 arthritis, a low dose (10–20 mg/day) and a high dose (0.5–1 mg/kg/day) of prednisone is recommended for grade 2 and grade 3 arthritis, respectively, with the temporary discontinuation of therapy. Disease-modifying antirheumatic drugs (DMARDs) are recommended as steroid-sparing agents in steroid-resistant cases and for steroid reduction [70].

2.7. Neurological irAEs-Peripheral Nervous System

Neurological irAEs include involvement of the peripheral nervous system (PNS) and the neuromuscular unit and central nervous system afflictions. The most common manifestations of ICI-related peripheral neurological syndromes are myositis, myasthenia gravis (MG), and Guillain–Barré Syndrome (GBS). Immune-related myositis is a rare irAE and has been reported mainly in melanoma patients [71]. Its clinical symptoms differ significantly from those of idiopathic and paraneoplastic inflammatory myopathies such as dermatomyositis (DM) and polymyositis (PM) [72][73]. There have also been reports of dyspnea, dysarthria, and dysphonia. Despite this, the clinical pattern is very consistent, with myalgia being the most prevalent and early symptom, even in the absence of creatine kinase (CK) elevation [73].
ICI-related MG is a well-known neurological irAE that has been extensively documented in case reports and case series. This condition rarely arises without associated myositis, according to growing data [74]. Individuals suspected of having an irMG should receive a thorough evaluation that includes a CK, an electromyogram, and, if possible, a muscular MRI and a biopsy in order to discover associated myositis and alter treatment and follow-up. The presence of irMG and ir-myositis at the same time raises the likelihood of a myasthenic crisis, which may necessitate ventilator support and hospitalization in an intensive care unit. Bulbar symptoms, dysarthria, dysphagia, and dyspnea occur in 50% of the cases [75].
In terms of treatment regarding irMG and irMyositis, pyridostigmine is commonly used, but rarely as a singular therapy (3–9%). Often, immune modulatory treatment is required, either with steroid alone (27–45%) or in combination with immunoglobulin infusion and/or plasma exchange (50–63%) [76][77].
GBS is an uncommon complication among irAEs, occurring in around 0.1–0.3% of patients treated with ICIs [78][79][80]. However, according to an earlier meta-analysis, this peripheral neuropathy was more common (up to 3% for anti-PD-L1 drugs and 7% for anti-PD-1) [81]. In addition, a systemic evaluation of 86 patients treated with ipilimumab or pembrolizumab found that 23% had some form of demyelinating polyradiculoneuropathy [82]. Acute classical GBS begins with subtle paresthesia, followed by leg weakness, then arm, face and oropharyngeal weakness as it spreads proximally (ascending paralysis). Pain is prevalent, manifesting as bilateral sciatica or pain in the large muscles of the upper legs [83].

2.8. Neurological irAEs-Central Nervous System

The most common manifestations of CNS irAEs are encephalitis, meningitis [84][85] transverse myelitis [86][87][88] multiple sclerosis (MS)-like demyelination syndromes [89], vasculitis [90] and cranial neuropathies (CNDs) [91], however, myelitis, MS, vasculitis and CNDs are extremely rare irAEs and evidence of their presence is mainly based on case reports and case series.
The most common and severe irAEs with CNS involvement are aseptic meningitis and encephalitis. In cases of acute or subacute onset of headache, altered mental status, psychiatric symptoms, speech impairments, seizures or neurological deficits with/without fever, immune-related encephalitis (irEncephalitis) should be suspected and be differentiated from infectious encephalitis, CNS metastasis or metabolic encephalopathy [92]. According to Larkin and colleagues, irEncephalitis occurred in 0.16% of patients treated with ICI and accounted for 0.44% of all irAEs [84]. A total of 82% of encephalitis cases occurred alone without the manifestation of other irAEs [85]. The incidence of encephalitis is higher following anti-PD-1/PD-L1 treatment compared to after anti-CTLA-4 therapy and is more associated with combination therapy compared to monotherapy [93]. Given the high fatality rate of CNS irAEs (encephalitis, 6.3–12.8%, and meningitis, 7.4–8.3 [93][94] a prompt recognition of suspected patients is required and treatment with immune-modulating agents should start without delay. Diagnostic tests to confirm neurological irAEs include lumbar puncture, cytology, and tests for infectious diseases with neuroimaging.
irAEs often occur in a non-specific form that is difficult to diagnose and may onset several months after therapy [95]. In addition, the side effects that are more likely to cause death vary from drug to drug. In the case of CTLA-4 inhibitors colitis is more likely; in the case of PD-1/PDL1 inhibitors pneumonitis, hepatitis, and neurotoxicity are more likely; and in the case of combination therapy, colitis, and myocarditis are more likely [19]. Therefore, a complex multidisciplinary approach is essential in the clinical management of ICI treatment to prevent severe and life-threatening irAEs, including early monitoring or even prophylactic supportive care. 

3. The Biological Basis of irAEs in Anti-Cancer Immunotherapies

T cells are the most important players in the immune defense and play an equally important role in irAEs. Mechanisms by which T cells induce toxic effects are diverse and can be organ dependent but most commonly include an imbalance in the ratio of CD8+ and CD4+ cells [96]. Apart from activating tumor cell-specific T cells, systemically delivered ICI can activate previously inactivated self-recognizing T cells, which leads to autoimmune reactions [97]. Furthermore, an increase in cytokine and chemokine production can lead to inflammation, which can have adverse effects in that it promotes [98]. In many cases, ICI reduces regulatory T (Treg) cell proliferation and activity, abolishing T cell response regulation, and thereby increasing CD8+ T cells in the tumor peripheral area, which promotes autoimmune inflammation [99][100]. Treg cells further reduce the cytotoxic activity of T cells by hindering the function of antigen-presenting cells (APCs) and cause T cell exhaustion by an enhanced expression of anti-inflammatory IL-6 [101]. ICI causes diversification of the CD8+T cell repertoire [102] as an increase in highly proliferative and cytotoxic phenotype of CD8+T cells was observed in the colon of patients treated with anti-PD1 ICI. Similarly, an increase in CD8+T cell diversity was observed in patients receiving anti-CTLA4 therapy. This diverse repertoire of immunological actions could interestingly both benefit and harm the patient, as the combination of toxicity and benefits results from a complex and divergent pathway that converges at the diversification of T cells [54][96][103].
CD4+T cells are classified into different T helper types, namely Th1, Th2, Th17, and follicular T cells (Tfh). Both the IFN-γ-producing Th1 cells and IL-17-producing Th17 cells promote autoimmune reactions, leading to colitis, nephritis, liver damage, and skin complications [16]. Furthermore, patients receiving ICI often have a much higher Th17/Th1 cell ratio [104]. This increase in Th17 cells leads to increased inflammation and autoimmune adversities in patients [105]. Melanoma patients receiving anti-PD1 therapy demonstrated an increase in Th-1-mediated IL-6 production which led to severe colitis [106].
ICIs directly affect B cell function by reducing the number of circulating B cells while increasing the production of the CD21-low phenotype. These CD21-low B cells have high IFN-γ production that enhances immune response and might be responsible for B cell exhaustion [107]. B cells produce auto-antibodies in response to ICI against self-reactive T and B cells [108][109] which commonly damage the thyroid and islet cells of the pancreas leading to thyroiditis, hypothyroidism, and diabetes, respectively [50][110].
Other cells of the immune system also partake in irAEs directly or indirectly. Activated neutrophils promote T and B cell-mediated response and lead to distant inflammation and organ damage with their long-lasting effects [111][112]. Likewise, eosinophils promote irAEs by producing inflammation-promoting IL-17, which can elicit an inflammatory response; however, the effects mediated by eosinophils do not seriously affect patient survival [113]. NK cells are classically tumor suppressive; however, their functionality is altered in response to ICI in many cancers [114]. Either the NK cells become hyperactive and produce pro-inflammatory cytokines, or they modulate the immune functions of dendritic cells, T and B cells, and the epithelium, resulting in an exaggerated inflammatory response and damage to hepatocytes [115].
ICI treatment can induce macrophage activation and accumulation in patients with anti-PD1 treatment, which is translated to muscle weakness, atrophy, and myopathy [116]. In response to ICI, exhausted T cells release IFN-γ, which recruits monocyte-derived macrophages. These macrophages acquire cytotoxic abilities and are reported to damage the pancreas leading to diabetes [117]. Furthermore, with an active T cell response and Treg reduction, the infiltration of type 2 macrophages promotes further inflammation and leads to progressive organ damage [118]. In patients receiving ICI, activated monocytes were reported to promote liver inflammation, coagulation and fibrinolysis pathways, and hyperactivation of the innate immune system, leading to dermatitis [94][119][120].
Although the involvement of these immune cells in promoting irAEs is clinically evident, the mechanistic basis and potential cross-talks need further elucidation. Moreover, the role of other factors such as genetics, epigenetics, environment, and predisposition to autoimmune disease cannot be neglected. Equivalently, the gut microbiota is another contributing factor that can alter immune function by cross-reactivity, as an elevated level of microbial antibodies has been detected in ICI-receiving patients [121] which might result in the competition with self or tumor antigens or the recruitment and activation of immune cells [102]. Species of Bacteroides and Burkholderiales provide protection against irAEs; Bacteroides fragilis, for example, reduces irAEs by promoting Treg cell development and production of anti-inflammatory IL-10 [122]. On the contrary, high Firmicutes phylum can promote colitis in melanoma patients by supposedly sequestering Treg cells [123].
The mechanisms by which the immune system reacts to and regulates the effects of ICI are diverse and complex. Several pre-clinical and clinical trials are currently underway to identify specific antibodies, cytokines, and/or pathways to circumvent these mild to severe irAEs and improve the usability of ICI treatments worldwide [124]. Figure 2 shows the immune-modulatory role of ICI treatment in different immune cells.
Figure 2. Immune modulatory role of immune cells in response to ICI blockade. Immunotherapy mediates the functional alteration of immune cells and the microbiome, leading to hyperactivation, reduced proliferation, self-cytotoxicity, autoantibody production and cross-reactivity. This complex modulation of the immune system facilitates a pro-inflammatory and autoreactive immune environment which translates to immunotherapy-related adverse events.

References

  1. Ai, L.; Chen, J.; Yan, H.; He, Q.; Luo, P.; Xu, Z.; Yang, X. Research status and outlook of pd-1/pd-l1 inhibitors for cancer therapy. Drug Des. Dev. Ther. 2020, 14, 3625–3649.
  2. Bagchi, S.; Yuan, R.; Engleman, E.G. Immune Checkpoint Inhibitors for the Treatment of Cancer: Clinical Impact and Mechanisms of Response and Resistance. Annu. Rev. Pathol. Mech. Dis. 2021, 16, 223–249.
  3. Wardill, H.R.; Chan, R.J.; Chan, A.; Keefe, D.; Costello, S.P.; Hart, N.H. Dual contribution of the gut microbiome to immunotherapy efficacy and toxicity: Supportive care implications and recommendations. Support Care Cancer 2022, 30, 6369–6373.
  4. Lee, Y.K.; Mazmanian, S.K. Has the microbiota played a critical role in the evolution of the adaptive immune system? Science 2010, 330, 1768–1773.
  5. Pierrard, J.; Seront, E. Impact of the gut microbiome on immune checkpoint inhibitor efficacy-a systematic review. Curr. Oncol. 2019, 26, 395–403.
  6. Huang, C.; Li, M.; Liu, B.; Zhu, H.; Dai, Q.; Fan, X.; Mehta, K.; Huang, C.; Neupane, P.; Wang, F.; et al. Relating Gut Microbiome and Its Modulating Factors to Immunotherapy in Solid Tumors: A Systematic Review. Front. Oncol. 2021, 11, 642110.
  7. Gopalakrishnan, V.; Spencer, C.N.; Nezi, L.; Reuben, A.; Andrews, M.C.; Karpinets, T.V.; Prieto, P.A.; Vicente, D.; Hoffman, K.; Wei, S.C.; et al. Gut microbiome modulates response to anti–PD-1 immunotherapy in melanoma patients. Science 2018, 359, 97–103.
  8. Matson, V.; Fessler, J.; Bao, R.; Chongsuwat, T.; Zha, Y.; Alegre, M.-L.; Luke, J.J.; Gajewski, T.F. The commensal microbiome is associated with anti–PD-1 efficacy in metastatic melanoma patients. Science 2018, 359, 104–108.
  9. Limeta, A.; Ji, B.; Levin, M.; Gatto, F.; Nielsen, J. Meta-analysis of the gut microbiota in predicting response to cancer immunotherapy in metastatic melanoma. JCI Insight 2020, 5, e140940.
  10. de Kivit, S.; Tobin, M.C.; Forsyth, C.B.; Keshavarzian, A.; Landay, A.L. Regulation of Intestinal Immune Responses through TLR Activation: Implications for Pro- and Prebiotics. Front Immunol. 2014, 5, 60.
  11. Zitvogel, L.; Ayyoub, M.; Routy, B.; Kroemer, G. Microbiome and Anticancer Immunosurveillance. Cell 2016, 165, 276–287.
  12. Arnaud-Coffin, P.; Maillet, D.; Gan, H.K.; Stelmes, J.-J.; You, B.; Dalle, S.; Péron, J. A systematic review of adverse events in randomized trials assessing immune checkpoint inhibitors. Int. J. Cancer 2019, 145, 639–648.
  13. Martins, F.; Sofiya, L.; Sykiotis, G.P.; Lamine, F.; Maillard, M.; Fraga, M.; Shabafrouz, K.; Ribi, C.; Cairoli, A.; Guex-Crosier, Y.; et al. Adverse effects of immune-checkpoint inhibitors: Epidemiology, management and surveillance. Nat. Rev. Clin. Oncol. 2019, 16, 563–580.
  14. Brown, V.T.; Antol, D.D.; Racsa, P.N.; Ward, M.A.; Naidoo, J. Real-World Incidence and Management of Immune-Related Adverse Events from Immune Checkpoint Inhibitors: Retrospective Claims-Based Analysis. Cancer Investig. 2021, 39, 789–796.
  15. Ouyang, T.; Cao, Y.; Kan, X.; Chen, L.; Ren, Y.; Sun, T.; Yan, L.; Xiong, B.; Liang, B.; Zheng, C. Treatment-Related Serious Adverse Events of Immune Checkpoint Inhibitors in Clinical Trials: A Systematic Review. Front. Oncol. 2021, 11, 621639.
  16. Okiyama, N.; Tanaka, R. Immune-related adverse events in various organs caused by immune checkpoint inhibitors. Allergol. Int. 2022, 71, 169–178.
  17. Bardoscia, L.; Pasinetti, N.; Triggiani, L.; Cozzi, S.; Sardaro, A. Biological Bases of Immune-Related Adverse Events and Potential Crosslinks With Immunogenic Effects of Radiation. Front. Pharmacol. 2021, 12, 746853.
  18. Khan, S.; Gerber, D.E. Autoimmunity, checkpoint inhibitor therapy and immune-related adverse events: A review. Semin. Cancer Biol. 2019, 64, 93–101.
  19. Wang, D.Y.; Salem, J.E.; Cohen, J.V.; Chandra, S.; Menzer, C.; Ye, F.; Zhao, S.; Das, S.; Beckermann, K.E.; Ha, L.; et al. Fatal toxic effects associated with immune checkpoint inhibitors: A systematic review and meta-analysis. JAMA Oncol. 2018, 4, 1721–1728.
  20. Conroy, M.; Naidoo, J. Immune-related adverse events and the balancing act of immunotherapy. Nat. Commun. 2022, 13, 1–4.
  21. Esfahani, K.; Meti, N.; Miller, W.H.; Hudson, M. Adverse events associated with immune checkpoint inhibitor treatment for cancer. Can. Med Assoc. J. 2019, 191, E40–E46.
  22. Postow, M.A.; Sidlow, R.; Hellmann, M.D. Immune-Related Adverse Events Associated with Immune Checkpoint Blockade. N. Engl. J. Med. 2018, 378, 158–168.
  23. Ramos-Casals, M.; Brahmer, J.R.; Callahan, M.K.; Flores-Chávez, A.; Keegan, N.; Khamashta, M.A.; Lambotte, O.; Mariette, X.; Prat, A.; Suárez-Almazor, M.E. Immune-related adverse events of checkpoint inhibitors. Nat. Rev. Dis. Prim. 2020, 6, 1–21.
  24. Davar, D.; Kirkwood, J.M. PD-1 Immune Checkpoint Inhibitors and Immune-Related Adverse Events: Understanding the Upside of the Downside of Checkpoint Blockade. JAMA Oncol. 2019, 5, 942–943.
  25. Wang, Y.; Zhou, S.; Yang, F.; Qi, X.; Wang, X.; Guan, X.; Shen, C.; Duma, N.; Aguilera, J.V.; Chintakuntlawar, A.; et al. Treatment-Related Adverse Events of PD-1 and PD-L1 Inhibitors in Clinical Trials: A Systematic Review and Meta-analysis. JAMA Oncol. 2019, 5, 1008–1019.
  26. Wongvibulsin, S.; Pahalyants, V.; Kalinich, M.; Murphy, W.; Yu, K.-H.; Wang, F.; Chen, S.T.; Reynolds, K.; Kwatra, S.G.; Semenov, Y.R. Epidemiology and risk factors for the development of cutaneous toxicities in patients treated with immune-checkpoint inhibitors: A United States population-level analysis. J. Am. Acad. Dermatol. 2021, 86, 563–572.
  27. Zhao, F.; Zhu, J.; Yu, R.; Shao, T.; Chen, S.; Zhang, G.; Shu, Q. Cutaneous adverse events in patients treated with PD-1/PD-L1 checkpoint inhibitors and their association with survival: A systematic review and meta-analysis. Sci. Rep. 2022, 12, 20038.
  28. Phillips, G.S.; Wu, J.; Hellmann, M.D.; Postow, M.A.; Rizvi, N.A.; Freites-Martinez, A.; Chan, D.; Dusza, S.; Motzer, R.J.; Rosenberg, J.E.; et al. Treatment Outcomes of Immune-Related Cutaneous Adverse Events. J. Clin. Oncol. 2019, 37, 2746–2758.
  29. Chadha, S.; Para, A.J.; Choi, J. Management of Immune-Related Cutaneous Adverse Reactions to PD-1 and PD-L1 Inhibitors for the Inpatient Dermatologist. Curr. Dermatol. Rep. 2020, 9, 231–243.
  30. Tang, L.; Wang, J.; Lin, N.; Zhou, Y.; He, W.; Liu, J.; Ma, X. Immune Checkpoint Inhibitor-Associated Colitis: From Mechanism to Management. Front. Immunol. 2021, 12, 800879.
  31. de Malet, A.; Antoni, G.; Collins, M.; Soularue, E.; Marthey, L.; Vaysse, T.; Coutzac, C.; Chaput, N.; Mateus, C.; Robert, C.; et al. Evolution and recurrence of gastrointestinal immune-related adverse events induced by immune checkpoint inhibitors. Eur. J. Cancer 2019, 106, 106–114.
  32. Shivaji, U.N.; Jeffery, L.; Gui, X.; Smith, S.C.L.; Ahmad, O.F.; Akbar, A.; Ghosh, S.; Iacucci, M. Immune checkpoint inhibitor-associated gastrointestinal and hepatic adverse events and their management. Ther. Adv. Gastroenterol. 2019, 12, 1756284819884196.
  33. Som, A.; Mandaliya, R.; Alsaadi, D.; Farshidpour, M.; Charabaty, A.; Malhotra, N.; Mattar, M.C. Immune checkpoint inhibitor-induced colitis: A comprehensive review. World J. Clin. Cases 2019, 7, 405–418.
  34. Das, S.; Johnson, D.B. Immune-related adverse events and anti-tumor efficacy of immune checkpoint inhibitors. J. Immunother. Cancer 2019, 7, 306.
  35. Naqash, A.R.; Kihn-Alarcón, A.J.; Stavraka, C.; Kerrigan, K.; Vareki, S.M.; Pinato, D.J.; Puri, S. The role of gut microbiome in modulating response to immune checkpoint inhibitor therapy in cancer. Ann. Transl. Med. 2021, 9, 1034.
  36. Kähler, K.C.; For the German Dermatologic Cooperative Oncology Group (DeCOG); Eigentler, T.K.; Gesierich, A.; Heinzerling, L.; Loquai, C.; Meier, F.; Meiss, F.; Pföhler, C.; Schlaak, M.; et al. Ipilimumab in metastatic melanoma patients with pre-existing autoimmune disorders. Cancer Immunol. Immunother. 2018, 67, 825–834.
  37. Guo, X.; Li, W.; Hu, J.; Zhu, E.C.; Su, Q. Hepatotoxicity in patients with solid tumors treated with PD-1/PD-L1 inhibitors alone, PD-1/PD-L1 inhibitors plus chemotherapy, or chemotherapy alone: Systematic review and meta-analysis. Eur. J. Clin. Pharmacol. 2020, 76, 1345–1354.
  38. Winer, A.; Bodor, J.N.; Borghaei, H. Identifying and managing the adverse effects of immune checkpoint blockade. J. Thorac. Dis. 2018, 10, S480–S489.
  39. Rogers, C.B.B.; Cuddahy, R.T.; Zawislak, M.C. Management of Acute Pancreatitis Associated With Checkpoint Inhibitors. J. Adv. Pract. Oncol. 2020, 11, 49–62.
  40. Kakuwa, T.; Hashimoto, M.; Izumi, A.; Naka, G.; Takeda, Y.; Sugiyama, H. Pembrolizumab-related pancreatitis with elevation of pancreatic tumour markers. Respirol. Case Rep. 2020, 8, e00525.
  41. Tian, Y.; Zhang, Z.; Yang, X.; Li, D.; Zhang, L.; Li, Z.; Zhang, S.; Mao, Y.; Jin, C.; Zhao, Y. The Risk Ratio of Immune-Related Colitis, Hepatitis, and Pancreatitis in Patients with Solid Tumors Caused by PD-1/PD-L1 Inhibitors: A Systematic Review and Meta-Analysis. Front. Oncol. 2020, 10, 261.
  42. Yin, J.; Wu, Y.; Yang, X.; Gan, L.; Xue, J. Checkpoint Inhibitor Pneumonitis Induced by Anti-PD-1/PD-L1 Therapy in Non-Small-Cell Lung Cancer: Occurrence and Mechanism. Front. Immunol. 2022, 13, 830631.
  43. Cho, J.Y.; Kim, J.; Lee, J.S.; Kim, Y.J.; Kim, S.H.; Lee, Y.J.; Cho, Y.-J.; Yoon, H.I.; Lee, J.H.; Lee, C.-T.; et al. Characteristics, incidence, and risk factors of immune checkpoint inhibitor-related pneumonitis in patients with non-small cell lung cancer. Lung Cancer 2018, 125, 150–156.
  44. Banavasi, H.; Kim, S.; Alkassis, S.; Daoud, A.; Laktineh, A.; Nagasaka, M.; Sukari, A.; Soubani, A.O. Immune checkpoint inhibitor-induced pneumonitis: Incidence, clinical characteristics, and outcomes. Hematol. Stem Cell Ther. 2021.
  45. Tiu, B.C.; Zubiri, L.; Iheke, J.; Pahalyants, V.; Theodosakis, N.; Ugwu-Dike, P.; Seo, J.; Tang, K.; E Sise, M.; Sullivan, R.; et al. Real-world incidence and impact of pneumonitis in patients with lung cancer treated with immune checkpoint inhibitors: A multi-institutional cohort study. J. Immunother. Cancer 2022, 10, e004670.
  46. A Muir, C.; Clifton-Bligh, R.J.; Long, G.V.; A Scolyer, R.; Lo, S.N.; Carlino, M.S.; Tsang, V.H.M.; Menzies, A.M. Thyroid Immune-related Adverse Events Following Immune Checkpoint Inhibitor Treatment. J. Clin. Endocrinol. Metab. 2021, 106, e3704–e3713.
  47. Bala-Hampton, J.E.; Bazzell, A.F.; Dains, J.E. Clinical Management of Pneumonitis in Patients Receiving Anti–PD-1/PD-L1 Therapy. J. Adv. Pract. Oncol. 2018, 9, 422.
  48. Yoon, J.H.; Hong, A.R.; Kim, H.K.; Kang, H.C. Characteristics of immune-related thyroid adverse events in patients treated with PD-1/PD-L1 inhibitors. Endocrinol. Metab. 2021, 36, 413–423.
  49. Chang, L.S.; Barroso-Sousa, R.; Tolaney, S.M.; Hodi, F.S.; Kaiser, U.B.; Min, L. Endocrine Toxicity of Cancer Immunotherapy Targeting Immune Checkpoints. In Endocrine Reviews; Oxford University Press: Oxford, UK, 2018; Volume 40, pp. 17–65.
  50. Osorio, J.C.; Ni, A.; Chaft, J.E.; Pollina, R.; Kasler, M.K.; Stephens, D.; Rodriguez, C.; Cambridge, L.; Rizvi, H.; Wolchok, J.D.; et al. Antibody-mediated thyroid dysfunction during T-cell checkpoint blockade in patients with non-small-cell lung cancer. Ann. Oncol. 2017, 28, 583–589.
  51. Kotwal, A.; Kottschade, L.; Ryder, M. PD-L1 Inhibitor-Induced Thyroiditis Is Associated with Better Overall Survival in Cancer Patients. Thyroid 2020, 30, 177–184.
  52. Baek, H.-S.; Jeong, C.; Shin, K.; Lee, J.; Suh, H.; Lim, D.-J.; Kang, M.I.; Ha, J. Association between the type of thyroid dysfunction induced by immune checkpoint inhibitors and prognosis in cancer patients. BMC Endocr. Disord. 2022, 22, 89.
  53. Iwama, S.; De Remigis, A.; Callahan, M.K.; Slovin, S.F.; Wolchok, J.D.; Caturegli, P. Pituitary Expression of CTLA-4 Mediates Hypophysitis Secondary to Administration of CTLA-4 Blocking Antibody. Sci. Transl. Med. 2014, 6, 230ra45.
  54. Robert, L.; Tsoi, J.; Wang, X.; Emerson, R.; Homet, B.; Chodon, T.; Mok, S.; Huang, R.R.; Cochran, A.J.; Comin-Anduix, B.; et al. CTLA4 Blockade Broadens the Peripheral T-Cell Receptor Repertoire. Clin. Cancer Res. 2014, 20, 2424–2432.
  55. Weber, J.; Mandalà, M.; Del Vecchio, M.; Gogas, H.J.; Arance, A.M.; Cowey, C.L.; Dalle, S.; Schenker, M.; Chiarion-Sileni, V.; Marquez-Rodas, I.; et al. Adjuvant Nivolumab versus Ipilimumab in Resected Stage III or IV Melanoma. N. Engl. J. Med. 2017, 377, 1824–1835.
  56. Barroso-Sousa, R.; Barry, W.T.; Garrido-Castro, A.C.; Hodi, F.S.; Min, L.; Krop, I.E.; Tolaney, S.M. Incidence of Endocrine Dysfunction Following the Use of Different Immune Checkpoint Inhibitor Regimens. JAMA Oncol. 2018, 4, 173–182.
  57. Shoushtari, A.N.; Friedman, C.F.; Navid-Azarbaijani, P.; Postow, M.A.; Callahan, M.K.; Momtaz, P.; Panageas, K.S.; Wolchok, J.D.; Chapman, P.B. Measuring Toxic Effects and Time to Treatment Failure for Nivolumab Plus Ipilimumab in Melanoma. JAMA Oncol. 2018, 4, 98–101.
  58. Ferrari, S.M.; Fallahi, P.; Elia, G.; Ragusa, F.; Ruffilli, I.; Patrizio, A.; Galdiero, M.R.; Baldini, E.; Ulisse, S.; Marone, G.; et al. Autoimmune Endocrine Dysfunctions Associated with Cancer Immunotherapies. Int. J. Mol. Sci. 2019, 20, 2560.
  59. Sullivan, R.J.; Weber, J.S. Immune-related toxicities of checkpoint inhibitors: Mechanisms and mitigation strategies. Nat. Rev. Drug Discov. 2021, 21, 495–508.
  60. Johnson, D.B.; Balko, J.M.; Compton, M.L.; Chalkias, S.; Gorham, J.; Xu, Y.; Hicks, M.; Puzanov, I.; Alexander, M.R.; Bloomer, T.L.; et al. Fulminant Myocarditis with Combination Immune Checkpoint Blockade. New Engl. J. Med. 2016, 375, 1749–1755.
  61. Ganatra, S.; Neilan, T.G. Immune Checkpoint Inhibitor-Associated Myocarditis. Oncol. 2018, 23, 879–886.
  62. Makunts, T.; Saunders, I.M.; Cohen, I.V.; Li, M.; Moumedjian, T.; Issa, M.A.; Burkhart, K.; Lee, P.; Patel, S.P.; Abagyan, R. Myocarditis occurrence with cancer immunotherapy across indications in clinical trial and post-marketing data. Sci. Rep. 2021, 11, 17324.
  63. Matzen, E.; Bartels, L.E.; Løgstrup, B.; Horskær, S.; Stilling, C.; Donskov, F. Immune checkpoint inhibitor-induced myocarditis in cancer patients: A case report and review of reported cases. Cardiooncology 2021, 7, 27.
  64. Rubio-Infante, N.; Ramírez-Flores, Y.A.; Castillo, E.C.; Lozano, O.; García-Rivas, G.; Torre-Amione, G. A Systematic Review of the Mechanisms Involved in Immune Checkpoint Inhibitors Cardiotoxicity and Challenges to Improve Clinical Safety. Front. Cell Dev. Biol. 2022, 10, 851032.
  65. Salem, J.E.; Manouchehri, A.; Moey, M.; Lebrun-Vignes, B.; Bastarache, L.; Pariente, A.; Gobert, A.; Spano, J.P.; Balko, J.M.; Bonaca, M.P.; et al. Cardiovascular toxicities associated with immune checkpoint inhibitors: An observational, retrospective, pharmacovigilance study. Lancet Oncol. 2018, 19, 1579–1589.
  66. Dong, H.; Qi, Y.; Kong, X.; Wang, Z.; Fang, Y.; Wang, J. PD-1/PD-L1 Inhibitor-Associated Myocarditis: Epidemiology, Characteristics, Diagnosis, Treatment, and Potential Mechanism. Front. Pharmacol. 2022, 13, 1082.
  67. Safi, M.; Ahmed, H.; Al-Azab, M.; Xia, Y.L.; Shan, X.; Al-Radhi, M.; Al-Danakh, A.; Shopit, A.; Liu, J. PD-1/PDL-1 Inhibitors and Cardiotoxicity; Molecular, Etiological and Management Outlines. J. Adv. Res. 2021, 29, 45–54.
  68. Varricchi, G.; Marone, G.; Mercurio, V.; Galdiero, M.R.; Bonaduce, D.; Tocchetti, C.G. Immune Checkpoint Inhibitors and Cardiac Toxicity: An Emerging Issue. Curr. Med. Chem. 2018, 25, 1327–1339.
  69. Angelopoulou, F.; Bogdanos, D.; Dimitroulas, T.; Sakkas, L.; Daoussis, D. Immune checkpoint inhibitor-induced musculoskeletal manifestations. Rheumatol. Int. 2020, 41, 33–42.
  70. Chan, K.K.; Bass, A.R. Monitoring and Management of the Patient with Immune Checkpoint Inhibitor-Induced Inflammatory Arthritis: Current Perspectives. J. Inflamm. Res. 2022, 15, 3105–3118.
  71. Cappelli, L.C.; Gutierrez, A.K.; Bingham, C.O.; Shah, A.A. Rheumatic and Musculoskeletal Immune-Related Adverse Events Due to Immune Checkpoint Inhibitors: A Systematic Review of the Literature. Arthritis Care Res. 2017, 69, 1751–1763.
  72. Kao, J.C.; Liao, B.; Markovic, S.N.; Klein, C.; Naddaf, E.; Staff, N.P.; Liewluck, T.; Hammack, J.E.; Sandroni, P.; Finnes, H.; et al. Neurological Complications Associated with Anti–Programmed Death 1 (PD-1) Antibodies. JAMA Neurol. 2017, 74, 1216–1222.
  73. Moreira, A.; Loquai, C.; Pföhler, C.; Kähler, K.C.; Knauss, S.; Heppt, M.V.; Gutzmer, R.; Dimitriou, F.; Meier, F.; Mitzel-Rink, H.; et al. Myositis and neuromuscular side-effects induced by immune checkpoint inhibitors. Eur. J. Cancer 2019, 106, 12–23.
  74. Psimaras, D. Neuromuscular complications of immune checkpoint inhibitors. La Presse Médicale 2018, 47, e253–e259.
  75. Kao, J.C.; Brickshawana, A.; Liewluck, T. Neuromuscular Complications of Programmed Cell Death-1 (PD-1) Inhibitors. Curr. Neurol. Neurosci. Rep. 2018, 18, 63.
  76. Guptill, J.T.; Sanders, D.B.; Evoli, A. Anti-musk antibody myasthenia gravis: Clinical findings and response to treatment in two large cohorts. Muscle Nerve 2011, 44, 36–40.
  77. Gilhus, N.E.; Skeie, G.O.; Romi, F.; Lazaridis, K.; Zisimopoulou, P.; Tzartos, S. Myasthenia gravis—Autoantibody characteristics and their implications for therapy. Nat. Rev. Neurol. 2016, 12, 259–268.
  78. Wilgenhof, S.; Neyns, B. Anti-CTLA-4 antibody-induced Guillain–Barré syndrome in a melanoma patient. Ann. Oncol. 2011, 22, 991–993.
  79. Xu, M.; Nie, Y.; Yang, Y.; Lu, Y.-T.; Su, Q. Risk of Neurological Toxicities Following the Use of Different Immune Checkpoint Inhibitor Regimens in Solid Tumors: A Systematic Review and Meta-Analysis. Neurologist 2019, 24, 75–83.
  80. Fan, Y.; Xie, W.; Huang, H.; Wang, Y.; Li, G.; Geng, Y.; Hao, Y.; Zhang, Z. Association of Immune Related Adverse Events with Efficacy of Immune Checkpoint Inhibitors and Overall Survival in Cancers: A Systemic Review and Meta-analysis. Front. Oncol. 2021, 11, 633032.
  81. Man, J.; Ritchie, G.; Links, M.; Lord, S.J.; Lee, C.K. Treatment-related toxicities of immune checkpoint inhibitors in advanced cancers: A meta-analysis. Asia-Pacific J. Clin. Oncol. 2018, 14, 141–152.
  82. Johansen, A.; Christensen, S.J.; Scheie, D.; Højgaard, J.L.; Kondziella, D. Neuromuscular adverse events associated with anti-PD-1 monoclonal antibodies. Neurology 2019, 92, 663–674.
  83. Willison, H.J.; Jacobs, B.C.; A van Doorn, P. Guillain-Barré syndrome. Lancet 2016, 388, 717–727.
  84. Larkin, J.; Chmielowski, B.; Lao, C.D.; Hodi, F.S.; Sharfman, W.; Weber, J.; Suijkerbuijk, K.P.M.; Azevedo, S.; Li, H.; Reshef, D.; et al. Neurologic Serious Adverse Events Associated with Nivolumab Plus Ipilimumab or Nivolumab Alone in Advanced Melanoma, Including a Case Series of Encephalitis. Oncology 2017, 22, 709–718.
  85. Johnson, D.B.; Manouchehri, A.; Haugh, A.M.; Quach, H.T.; Balko, J.M.; Lebrun-Vignes, B.; Mammen, A.; Moslehi, J.J.; Salem, J.E. Neurologic toxicity associated with immune checkpoint inhibitors: A pharmacovigilance study. J. Immunother. Cancer 2019, 7, 134.
  86. Narumi, Y.; Yoshida, R.; Minami, Y.; Yamamoto, Y.; Takeguchi, S.; Kano, K.; Takahashi, K.; Saito, T.; Sawada, J.; Terui, H.; et al. Neuromyelitis optica spectrum disorder secondary to treatment with anti-PD-1 antibody nivolumab: The first report. BMC Cancer 2018, 18, 95.
  87. Mancone, S.; Lycan, T.; Ahmed, T.; Topaloglu, U.; Dothard, A.; Petty, W.J.; Strowd, R.E. Severe neurologic complications of immune checkpoint inhibitors: A single-center review. J. Neurol. 2018, 265, 1636–1642.
  88. Shimada, T.; Hoshino, Y.; Tsunemi, T.; Hattori, A.; Nakagawa, E.; Yokoyama, K.; Hattori, N. Neuromyelitis optica spectrum disorder after treatment with pembrolizumab. Mult. Scler. Relat. Disord. 2019, 37, 101447.
  89. Garcia, C.R.; Jayswal, R.; Adams, V.; Anthony, L.B.; Villano, J.L. Multiple sclerosis outcomes after cancer immunotherapy. Clin. Transl. Oncol. 2019, 21, 1336–1342.
  90. Läubli, H.; Hench, J.; Stanczak, M.; Heijnen, I.; Papachristofilou, A.; Frank, S.; Zippelius, A.; Stenner-Liewen, F. Cerebral vasculitis mimicking intracranial metastatic progression of lung cancer during PD-1 blockade. J. Immunother. Cancer 2017, 5, 46.
  91. Bruno, F.; Palmiero, R.A.; Ferrero, B.; Franchino, F.; Pellerino, A.; Milanesi, E.; Soffietti, R.; Rudà, R. Pembrolizumab-Induced Isolated Cranial Neuropathy: A Rare Case Report and Review of Literature. Front. Neurol. 2021, 12, 669493.
  92. Watanabe, Y.; Kikuchi, R.; Iwai, Y.; Ito, M.; Tsukamoto, H.; Yamazaki, K.; Nakamura, H.; Aoshiba, K. Varicella zoster virus encephalitis mimicking nivolumab-induced autoimmune neuropathy in a patient with lung cancer. J. Thorac. Oncol. 2019, 14, e163–e165.
  93. Johnson, D.B.; McDonnell, W.J.; Gonzalez-Ericsson, P.I.; Al-Rohil, R.N.; Mobley, B.C.; Salem, J.-E.; Wang, D.Y.; Sanchez, V.; Wang, Y.; Chastain, C.A.; et al. A case report of clonal EBV-like memory CD4+ T cell activation in fatal checkpoint inhibitor-induced encephalitis. Nat. Med. 2019, 25, 1243–1250.
  94. Sato, K.; Mano, T.; Iwata, A.; Toda, T. Neurological and related adverse events in immune checkpoint inhibitors: A pharmacovigilance study from the Japanese Adverse Drug Event Report database. J. Neurooncol. 2019, 145, 1–9.
  95. Owen, C.N.; Bai, X.; Quah, T.; Lo, S.N.; Allayous, C.; Callaghan, S.; Martínez-Vila, C.; Wallace, R.; Bhave, P.; Reijers, I.L.M.; et al. Delayed immune-related adverse events with anti-PD-1-based immunotherapy in melanoma. Ann. Oncol. 2021, 32, 917–925.
  96. Oh, D.Y.; Cham, J.; Zhang, L.; Fong, G.; Kwek, S.S.; Klinger, M.; Faham, M.; Fong, L. Immune Toxicities Elicted by CTLA-4 Blockade in Cancer Patients Are Associated with Early Diversification of the T-cell RepertoireIpilimumab-Induced T-cell Diversity Predicts IRAEs. Cancer Res. 2017, 77, 1322–1330.
  97. Kumar, P.; Saini, S.; Prabhakar, B.S. Cancer immunotherapy with check point inhibitor can cause autoimmune adverse events due to loss of Treg homeostasis. Semin. Cancer Biol. 2019, 64, 29–35.
  98. Denk, D.; Greten, F.R. Inflammation: The incubator of the tumor microenvironment. Trends Cancer 2022, 8, 901–914.
  99. Milling, L.; Zhang, Y.; Irvine, D.J. Delivering safer immunotherapies for cancer. Adv. Drug Deliv. Rev. 2017, 114, 79–101.
  100. 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.
  101. Zhou, X.; Chen, X.; Han, L.; Liu, H. Mechanisms underlying immune-related adverse events during checkpoint immunotherapy. Clin. Sci. 2022, 136, 771–785.
  102. Young, A.; Quandt, Z.; Bluestone, J.A. The Balancing Act between Cancer Immunity and Autoimmunity in Response to Immunotherapy. Cancer Immunol. Res. 2018, 6, 1445–1452.
  103. Luoma, A.M.; Suo, S.; Williams, H.L.; Sharova, T.; Sullivan, K.; Manos, M.; Bowling, P.; Hodi, F.S.; Rahma, O.; Sullivan, R.J.; et al. Molecular Pathways of Colon Inflammation Induced by Cancer Immunotherapy. Cell 2020, 182, 655–671.e622.
  104. Kim, K.H.; Hur, J.Y.; Cho, J.; Ku, B.M.; Koh, J.; Koh, J.Y.; Sun, J.-M.; Lee, S.-H.; Ahn, J.S.; Park, K.; et al. Immune-related adverse events are clustered into distinct subtypes by T-cell profiling before and early after anti-PD-1 treatment. Oncoimmunology 2020, 9, 1722023.
  105. Von Euw, E.; Chodon, T.; Attar, N.; Jalil, J.; Koya, R.C.; Comin-Anduix, B.; Ribas, A. CTLA4 blockade increases Th17 cells in patients with metastatic melanoma. J. Transl. Med. 2009, 7, 35.
  106. Yoshino, K.; Nakayama, T.; Ito, A.; Sato, E.; Kitano, S. Severe colitis after PD-1 blockade with nivolumab in advanced melanoma patients: Potential role of Th1-dominant immune response in immune-related adverse events: Two case reports. BMC Cancer 2019, 19, 1019.
  107. Das, R.; Bar, N.; Ferreira, M.; Newman, A.M.; Zhang, L.; Bailur, J.K.; Bacchiocchi, A.; Kluger, H.; Wei, W.; Halaban, R.; et al. Early B cell changes predict autoimmunity following combination immune checkpoint blockade. J. Clin. Investig. 2018, 128, 715–720.
  108. de Moel, E.C.; Rozeman, E.A.; Kapiteijn, E.H.; Verdegaal, E.M.; Grummels, A.; Bakker, J.A.; Huizinga, T.W.; Haanen, J.B.; Toes, R.E.; van der Woude, D. Autoantibody Development under Treatment with Immune-Checkpoint InhibitorsAutoantibodies under Immune-Checkpoint Inhibitors. Cancer Immunol. Res. 2019, 7, 6–11.
  109. Esfahani, K.; Elkrief, A.; Calabrese, C.; Lapointe, R.; Hudson, M.; Routy, B.; Miller, W.H.; Calabrese, L. Moving towards personalized treatments of immune-related adverse events. Nat. Rev. Clin. Oncol. 2020, 17, 504–515.
  110. Stamatouli, A.M.; Quandt, Z.; Perdigoto, A.L.; Clark, P.L.; Kluger, H.; Weiss, S.A.; Gettinger, S.; Sznol, M.; Young, A.; Rushakoff, R.; et al. Collateral Damage: Insulin-Dependent Diabetes Induced with Checkpoint Inhibitors. Diabetes 2018, 67, 1471–1480.
  111. Kolaczkowska, E.; Kubes, P. Neutrophil recruitment and function in health and inflammation. Nat. Rev. Immunol. 2013, 13, 159–175.
  112. Sibille, A.; Alfieri, R.; Malaise, O.; Detrembleur, N.; Pirotte, M.; Louis, R.; Duysinx, B. Granulomatosis with polyangiitis in a patient on programmed death-1 inhibitor for advanced non-small-cell lung cancer. Front. Oncol. 2019, 9, 478.
  113. Waldman, A.D.; Fritz, J.M.; Lenardo, M.J. A guide to cancer immunotherapy: From T cell basic science to clinical practice. Nat. Rev. Immunol. 2020, 20, 651–668.
  114. Khan, M.; Arooj, S.; Wang, H. NK Cell-Based Immune Checkpoint Inhibition. Front. Immunol. 2020, 11, 167.
  115. Jiang, Y.; Qin, S.; Wei, X.; Liu, X.; Guan, J.; Zhu, H.; Chang, G.; Chen, Y.; Lu, H.; Qian, J.; et al. Highly activated TRAIL+ CD56bright NK cells are associated with the liver damage in HBV-LC patients. Immunol. Lett. 2021, 232, 9–19.
  116. Matas-García, A.; Milisenda, J.C.; Selva-O’Callaghan, A.; Prieto-González, S.; Padrosa, J.; Cabrera, C.; Reguart, N.; Castrejón, N.; Solé, M.; Ros, J.; et al. Emerging PD-1 and PD-1L inhibitors-associated myopathy with a characteristic histopathological pattern. Autoimmun. Rev. 2019, 19, 102455.
  117. Hu, H.; Zakharov, P.N.; Peterson, O.J.; Unanue, E.R. 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. USA 2020, 117, 31319–31330.
  118. Mihic-Probst, D.; Reinehr, M.; Dettwiler, S.; Kolm, I.; Britschgi, C.; Kudura, K.; Maggio, E.M.; Lenggenhager, D.; Rushing, E.J. The role of macrophages type 2 and T-regs in immune checkpoint inhibitor related adverse events. Immunobiology 2020, 225, 152009.
  119. Curry, J.L.; Reuben, A.; Szczepaniak-Sloane, R.; Ning, J.; Milton, D.R.; Lee, C.H.; Hudgens, C.; George, S.; Torres-Cabala, C.; Johnson, D.; et al. Gene expression profiling of lichenoid dermatitis immune-related adverse event from immune checkpoint inhibitors reveals increased CD14 + and CD16 + monocytes driving an innate immune response. J. Cutan. Pathol. 2019, 46, 627–636.
  120. Gudd, C.L.; Au, L.; Triantafyllou, E.; Shum, B.; Liu, T.; Nathwani, R.; Kumar, N.; Mukherjee, S.; Dhar, A.; Woollard, K.J.; et al. Activation and transcriptional profile of monocytes and CD8+ T cells are altered in checkpoint inhibitor-related hepatitis. J. Hepatol. 2021, 75, 177–189.
  121. Soularue, E.; Lepage, P.; Colombel, J.F.; Coutzac, C.; Faleck, D.; Marthey, L.; Collins, M.; Chaput, N.; Robert, C.; Carbonnel, F. Enterocolitis due to immune checkpoint inhibitors: A systematic review. Gut 2018, 67, 2056–2067.
  122. Al-Qadami, G.; Van Sebille, Y.; Le, H.; Bowen, J. Gut microbiota: Implications for radiotherapy response and radiotherapy-induced mucositis. Expert Rev. Gastroenterol. Hepatol. 2019, 13, 485–496.
  123. Chaput, N.; Lepage, P.; Coutzac, C.; Soularue, E.; Le Roux, K.; Monot, C.; Boselli, L.; Routier, E.; Cassard, L.; Collins, M.; et al. Baseline gut microbiota predicts clinical response and colitis in metastatic melanoma patients treated with ipilimumab. Ann. Oncol. 2019, 30, 2012.
  124. Berg, M.-H.H.; del Rincón, S.V.; Miller, W.H. Potential therapies for immune-related adverse events associated with immune checkpoint inhibition: From monoclonal antibodies to kinase inhibition. J. Immunother. Cancer 2022, 10, e003551.
More
Upload a video for this entry
Information
Subjects: Oncology
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , , , , , , ,
View Times: 671
Revisions: 2 times (View History)
Update Date: 07 Feb 2023
Academic Video Service