Skip to main content
Wiley Open Access Collection logoLink to Wiley Open Access Collection
. 2025 Jan 27;45(2):e16198. doi: 10.1111/liv.16198

Immune‐Mediated Liver Injury From Checkpoint Inhibitor: An Evolving Frontier With Emerging Challenges

Lily Dara 1,, Eleonora De Martin 2
PMCID: PMC11771569  PMID: 39868913

ABSTRACT

Over the past decade, immune checkpoint inhibitors (ICIs) have transformed the treatment of cancer, though they come with the risk of immune‐related adverse (irAEs) events such as hepatotoxicity or Immune‐mediated Liver Injury from Checkpoint Inhibitors (ILICI). ILICI is a serious irAE that, when severe, requires cessation of ICI and initiation of immunosuppression. Cytotoxic T Lymphocytes (CTLs) play a central role in ILICI; however, they are just part of the picture as immunotherapy broadly impacts all aspects of the immune microenvironment and can directly and indirectly activate innate and adaptive immune cells. Clinically, as our understanding of this entity grows, we encounter new challenges. The presentation of ILICI is heterogeneous with respect to latency, pattern of injury (hepatitis vs. cholangitis) and severity. This review focuses on our knowledge regarding risk factors, presentation and treatment of ILICI including ILICI refractory to steroids. An emerging topic, the possibility of rechallenge while accepting some risk, in patients who experience ILICI but require immunotherapy, is also discussed. This review provides an update on the current knowns and unknowns in ILICI and highlights several knowledge gaps where studies are needed.

Keywords: immune checkpoint inhibitors, immunosuppressive therapy, immunotherapy‐induced liver injury, liver toxicity


Summary.

  • ICI‐induced liver injury is due to a disruption of immune tolerance but the mechanism is still largely unknown.

  • Several risk factors have been identified such as female sex, young age, history of ICI therapy and ICI combination therapy.

  • ILICI is heterogeneous with respect to the time interval between initiation of ICI and onset of toxicity, biological characteristics (hepatitis vs. cholangitis) and severity.

  • Corticosteroid therapy should be tailored to the severity of the hepatitis. In steroid‐refractory hepatitis, MMF is a successful second‐line therapy, while third‐line therapy remains controversial.

  • Reintroduction of immunotherapy after ILICI is possible in certain cases and is patient and circumstance dependent and should be discussed by a multidisciplinary team to assess risk vs. benefit.

Abbreviations

AIH

autoimmune hepatitis

ALP

alkaline phosphatase

ALT

alanine transferase

AST

aspartate aminotransferase

CNI

calcineurin inhibitor

CTCAE

Common Terminology Criteria for Adverse Events

CTLA‐4

cytotoxic T‐lymphocyte‐associated protein‐4

CTLs

cytotoxic T‐cells

DILI

drug‐induced liver injury

FGL1

fibrinogen‐like protein‐1

HCC

hepatocellular carcinoma

ICIs

immune checkpoint inhibitors

IFN

interferon

IL

interleukin

ILICI

immunotherapy‐induced liver injury

irAEs

immune‐related Adverse Events

IVIG

intravenous immunoglobulin.

JAK

Janus kinase

LAG‐3

lymphocyte‐activation gene‐3

MALFD

metabolic dysfunction‐associated liver disease

MMF

mycophenolate mofetil

MRCP

magnetic resonance cholangiopancreatography

NK

natural killer

PD‐1

programmed cell death protein‐1

PDL‐1

PD ligand 1

PSC

primary sclerosing cholangitis

TIGIT

T‐cell immunoglobulin—ITIM domain

TIM3

T‐cell immunoglobulin and mucin domain‐containing protein 3

TNF

tumour necrosis factor

UDCA

Ursodeoxycholic Acid

ULN

upper limits of normal

VISTA

V‐domain Ig suppressor of T‐cell activation

VSIG‐1

V‐Set and immunoglobulin

1. Introduction

The liver is an immunotolerant organ. Due to constant exposure to foreign antigens, it has evolved several mechanisms to dampen the immune response. These mechanisms are many fold, involve hepatocytes, innate, adaptive, and non‐parenchymal liver cells, and importantly, they include the immune checkpoints. The immune checkpoints are ligand–receptor pairs that regulate the immune response and exert control over immune cells to prevent their aberrant activation. As such, they have a critical role in maintaining self‐tolerance and preventing autoimmunity. Malignant cells exploit these checkpoints to evade the immune system. The expression of immune checkpoints can lead to T‐cell exhaustion, a decline in T‐cell proliferation and reduced T‐cell function, allowing tumors to grow. Therefore, immune checkpoint inhibitors (ICIs) have been developed to block these immune‐regulatory pathways and augment immune responses. Since the liver relies on these self‐tolerance mechanisms to maintain its state of immune privilege, an unintended side effect of ICIs is immune‐mediated liver injury from checkpoint inhibitors, herein, ILICI.

The best studied immune checkpoints include the programmed cell death protein‐1 (PD‐1) and PD ligand 1 (PDL‐1) and the cytotoxic T‐lymphocyte‐associated protein‐4 (CTLA‐4) pathways [1]. There are currently over 10 FDA‐approved ICIs but given resistance and the side effects of these antibodies, there are more than 100 immunotherapy targets under development as next‐generation immunomodulators [2, 3]. Some of these targets include T‐cell immunoglobulin and mucin domain‐containing protein 3 (TIM3), lymphocyte‐ activation gene‐3 (LAG‐3), and T‐cell immunoglobulin and ITIM domain (TIGIT), fibrinogen‐like protein‐1 (FGL1), V‐Set and immunoglobulin domain containing (VSIG‐1), and V‐domain Ig suppressor of T‐cell activation (VISTA), among others.

ICIs are currently critical tools in the oncology toolbox for treatment of various malignancies. It is estimated that over 50% of cancers will be exposed to immunotherapy due to their high efficacy and better tolerability compared to conventional and targeted chemotherapy. Despite their undeniable effectiveness, the consequences and immune‐related Adverse Events (irAEs) such as ILICI are currently inevitable. Therefore, it is critical to understand, prevent, diagnose, and treat these unintended side effects. And to complicate matters further, unlike other forms of drug‐induced liver injury (DILI) where discontinuing the drug is the best solution, in cancer patients where immunotherapy is working, the benefits are significant. Therefore, finding solutions towards safe continuation of the drug is imperative.

2. Mechanism of ILICI

Immunotherapy‐induced liver injury is thought to be due to the activation of cytotoxic T‐cells (CTLs) that inadvertently target the liver. However, immunotherapy can have very broad effects on the immune microenvironment and can directly or indirectly effect cells other than CTLs, such as B cells, FoxP3+ Tregs, T helper cells, and even innate immune cells such as macrophages and dendritic cells [4] (Figure 1). In addition, ICIs modify the tumor microenvironment and circulating chemokines and cytokine levels, in fact, baseline and post‐treatment cytokine levels have been associated with the development of irAEs such as ILICI [5]. Various cytokines have been shown to be upregulated in patients and mice with ILICI, and these include interleukin (IL)‐6, IL1b, interferon (IFN) ‐γ, tumour necrosis factor (TNF)‐α and chemokines including CXCL9, CXCL10, CXCL11 and CXCL13 [5].

FIGURE 1.

FIGURE 1

Immune Checkpoint Inhibitors (ICIs) alter the immune microenvironment in various ways and by affecting multiple immune cell types.

In some tumors, neoepitope formation and chronic inflammation, break immune tolerance to tissue‐specific self‐antigens leading to serious irAEs [6]. One of the current leading hypotheses for immunotherapy side effects is the concept of epitope spreading. It is hypothesised that the lysis of tumors causes the release of proteins, forming of neoantigens which then drives irAEs [7]. Epitope spreading is the diversification of the initial T‐cell response against these novel epitopes and neoantigens that diverge from the original epitopes targeted. Skin, intestines and the liver are among the most commonly affected organs. Why T‐cells would specifically target neoantigens in certain tissues and organs and not others is not clear, as many neoantigens are present across multiple tissues and cell types. ICIs induce diversification of T‐cell clones and expansion of CTLs, and studies have shown similarities between tumour infiltrating CTLs and tissues affected by irAEs suggesting a mechanistic role for epitope spreading in irAEs [7].

B cells are in constant crosstalk with other cells within the tumor immune microenvironment. CD20+ B cells and their crosstalk with CD8+ T‐cells play a crucial role in the anti‐tumor immune response through costimulatory signalling and by promoting cytotoxic T‐cell survival and proliferation independently of antigen presentation through CD27/CD70 interactions [8]. B cells also respond to immunotherapy, and activation of B cells portends a favourable anti‐tumour response. Blockade of immune checkpoints has been associated with distinct changes in the B cell compartment, increase in CD21lo B cells and increase in plasmablasts that correlate with the development of irAEs [9, 10]. CTLA4 is a critical checkpoint in B cells, and its loss results in immune dysregulation and autoimmunity [4]. PD‐1 is also expressed in B cells and the blockade of the PD‐1 pathways increases their proliferation, activation and the production of inflammatory cytokines [9, 11]. The physiologic importance of CTLA4 and PD‐1 in keeping B cells in check suggests that the effect of ICIs on B cells likely contributes to the development of irAEs and ILICI. Although small studies have reported autoantibody formation during irAEs, the significance of these and the role of B cells during these events requires further study [12].

Tregs suppress anti‐tumour immunity, and therefore checkpoint blockade helps overcome this by attenuating Treg function and decreasing the Treg/T helper (Th) ratio [13]. This suppression of T reg numbers and function promotes autoimmunity, and in preclinical models of irAEs there is a negative correlation between Treg numbers and side effects. In concert with this, ICIs promote increased inflammatory CD4+ Th17 cells and Th1 proliferation resulting in increased IL‐2, IFN, and TNF production to activate CTLs, as well as innate immune cells such as macrophages and natural killer (NK) cells [7, 13].

An increase in the Th17 population and IL‐17 has also been reported post ICI therapy and with certain irAEs [14, 15]. These proinflammatory cytokines can subsequently activate an innate immune response by recruiting NK cells and macrophages contributing to liver injury. Macrophages play a role in ILICI, and this has been shown in both humans and mice models [16, 17]. Liver biopsies from patients with ILICI revealed the presence of both CD8+ CTLs and CCR2 + CD68+ macrophages in proximity to areas of injury, suggesting that immune cells work in concert and the adaptive innate crosstalk is important in the pathogenesis of ILICI [16]. This crosstalk was confirmed in a mouse model of ILICI where macrophage and T‐cell interactions leading to NLRP3 inflammasome activation were shown to drive hepatocyte apoptosis [17]. Indeed, while the effector cells causing liver injury seem to be the CD8+ T‐cells, as depletion of these cells significantly dampens injury, there still exists a macrophage infiltrate in the liver. This suggests that macrophage activation and recruitment to the liver are independent of CD8+ T‐cell infiltration [17]. There are still many unknowns in the mechanism of ILICI, and it is important to decipher the contribution of the various cell types and signalling pathways to move us towards finding therapeutic solutions.

3. Risk Factors for ICI Toxicity

The identification of risk factors for irAEs presents a significant challenge due to the high variability of these events. The use of two ICIs increases the risk of ILICI [1]. It is well known that females are more susceptible to autoimmune disease. Indeed male sex portends a more favourable response to immunotherapy, and female sex has been associated with a higher incidence of irAEs [18, 19, 20]. Analysis of over 23 000 patients from 202 trials experiencing adverse events revealed a 49% increased risk among women receiving immunotherapy (OR 1.49; 95% CI,1.24 to 1.78; p = 0.001) [18]. This finding was also reported in a subgroup of patients treated with immunosuppressive therapy for an autoimmune disease who received ICI. The results indicated that female patients were more likely to develop irAEs, although in this small study the risk of exacerbation of existing autoimmune disease was lower [21]. While larger studies have shown this in diverse cohorts, some small studies have not been able to replicate this finding [22]. Not surprisingly, female sex has been shown to be a predictor of ILICI in large cohorts and real‐world studies [23, 24]. Given the well‐known effect of sex differences in immune‐oncology and the higher incidence of autoimmune disease in females, the hypothesis of a role of sex as a risk factor for toxicity appears plausible [20]. However, much of this data is retrospectively extrapolated from randomized controlled trials and cohort studies, highlighting the need for further prospectively collected data to reach definitive conclusions.

Age has also been suggested as a risk factor for irAEs. A meta‐analysis of 13 studies found that younger age was significantly associated with the risk of any grade and of grade ≥ 3 ILICI [25]. Other identified risk factors were the use of two ICIs (dual checkpoint blockade), a history of prior ICI exposure, higher aspartate aminotransferase (AST), higher baseline alanine transferase (ALT) and lower baseline alkaline phosphatase (ALP), malignant melanoma, and high lymphocyte count [24, 25, 26].

Regarding the association between underlying cancer and incidence of irAEs, it is important to note that in large meta‐analyses, the highest mean all‐grade adverse events incidence was observed in melanoma. However, the mean incidences of all‐grade and grade 3 or higher adverse events were similar across different cancer types [27]. While this holds true for different cancer types, it likely does not apply to patients with hepatocellular carcinoma (HCC) and, consequently, underlying liver disease. Indeed, HCC and pre‐existing hepatic disease appear to be associated with a higher rate of ILICI compared to the treatment of non‐hepatic cancers [28, 29]. Furthermore, other studies on patients with underlying liver disease have indicated that chronic liver diseases such as hepatitis B and metabolic dysfunction‐associated liver disease (MALFD) may also be risk factors for ILICI [30, 31].

The diagnosis of pre‐existing autoimmune disease is associated with an increased risk for disease flares and irAEs occurrence, although this is highly variable according to the type of the disease and the ICI administered [32]. In general, the greater the immunosuppressive therapy received prior to the introduction of ICIs, the greater the risk of irAEs [32]. The risk of ILICI in patients with autoimmune disease has not been extensively studied. In a small European cohort of 22 patients with hepatic autoimmune diseases, treated with immunotherapy for various malignancies, 36% (8/22) experienced grades 1–2 irAEs, three of which were ILICI, and none experienced ≥ grade 3 injury [33]. In addition, a recent systematic review of 22 studies in patients with Child‐Pugh A and Child‐Pugh B cirrhosis found a similar incidence of treatment‐related adverse events regardless of Child‐Pugh classification [34].

As ICIs break tolerance, it is also feasible to hypothesize that they can predispose patients to idiosyncratic DILI from concomitant meds. Indeed, in one study, a combination of ipilimumab plus dacarbazine resulted in significantly more irAEs compared to ipilimumab alone (56.3% vs. 27.5%, p < 0.001) [35]. In addition to dacarbazine recently multiple reports of DILI following the addition of a small‐molecule inhibitor such as enzyme inhibitors and tyrosine kinase inhibitors to ICIs have been published. Although these are early reports, they point to the complexity of determining causality when multiple drugs are used concomitantly with or even following ICI therapy [36, 37, 38, 39, 40].

Recently, the Drug‐Induced Liver Injury Network (DILIN) published its experience on 57 cases of ILICI, adjudicated by an expert panel of hepatologists as high likelihood [41]. In this cohort, younger age was associated with a hepatocellular pattern of injury. In terms of genetic risk factors, the HLA alleles for autoimmune hepatitis (AIH) were not overrepresented in the ILICI patients. However, two host immune response genes (EDIL3 and SAMA5A) and three other genes (GABRP, SMAD3 and SLCO1B1) were associated with ILICI (OR 2.08–2.4, p < 0.01) [41].

Lastly, the identification of risk factors should not preclude the use of these drugs but rather guide the clinician in identifying higher‐risk patients for hepatotoxicity events and allow for personalized monitoring during therapy.

4. Clinical Presentation

The presentation of ICI‐induced liver injury is heterogeneous in terms of the time interval between initiation of therapy and onset of toxicity, in terms of clinical and biological features (hepatitis vs. cholangitis), and in terms of severity. An accurate specific assessment, along with the causality assessment for all DILI, is paramount for diagnosis and management (Table 1). Hepatic toxicity may occur after a single injection or several weeks after discontinuation of therapy, although the latter is very rare [42, 43]. The median interval between ICI and liver injury is between 3 and 14 weeks, with some variation depending on the drug used, according to some studies: shorter in patients treated with anti‐CTLA4 compared to those treated with anti‐PD1/anti‐PDL1 [44, 45, 46, 47]. The median latency of DILI in patients with hepatocellular injury in the well‐characterised DILIN cohort was 2–3 months, while latency was closer to 6 months in patients with cholestatic ILICI [41]. Although it is possible for ILICI to manifest after 6 months and even one year of therapy, the likelihood is much lower [24]. The late onset is explained by the prolonged activity of these drugs which exceeds their half‐life [48].

TABLE 1.

Key clinical considerations in immune‐mediated liver injury from checkpoint inhibitors (ILICI).

Key points Comments
Rule out common causes of liver injury Similar to other forms of DILI, viral, metabolic, alcohol, stone disease should be ruled out [72, 73]
Concomitant medications and recent pharmacological history Previous exposure to ICI increases the risk of toxicity and the delay between last ICI dose, and new ICI introduction is of great importance due to the potential prolonged activity of the first drug [49]
Pre‐existing liver disease Increased risk of toxicity in the setting of HCC and/or cirrhosis [31, 32, 33, 34]
Type of ICI Association with different liver injury patterns such as cholestatic pattern being more common with anti‐PD1s such as pembrolizumab [54, 55, 56, 57, 58, 59, 60, 61]
Time interval between ICI injection and liver injury onset Late onset (even after ICI discontinuation) does not exclude ICI given long half‐life and receptor occupancy [44]
ICI in conjunction with traditional antineoplastic agents ICIs break immune tolerance and can predispose to DILI from other drugs such as dacarbazine [42, 43]
ICI in conjunction with small‐molecule inhibitors (SMI) Concomitant or subsequent treatment with small‐molecule inhibitors such as Tyrosine Kinase Inhibitors (TKI) is associated with hepatotoxicity. Whether this is due to a break in immune tolerance predisposing to TKI/SMI hepatotoxicity or ILICI or an additive or synergistic effect due to dual therapy is not yet understood [38, 39, 40, 41, 42]
Signs and symptoms Clinical symptoms may be associated with a more severe disease (e.g. jaundice, fever, oedema/ascites) and require different managements. Furthermore, they are associated with a higher risk of steroid‐refractory hepatitis [87]
Hepatocellular versus cholestatic pattern of DILI Distinction between hepatocellular and cholestatic patterns can lead to consideration of different options. Namely UDCA has been used in some centres in Europe for cholestatic ILICI although this is not recommended by most guidelines [53]
Severity of injury Relevant for the introduction, timing and dose of immunosuppressive therapy, as well as consideration of ICI reintroduction or rechallenge [1, 47, 75, 76, 77]
Imaging Can avoid misdiagnosis by detecting disease progression, tumoural hepatic thrombosis and biliary obstruction. Also can help in follow‐up [62, 63]

Patients may be asymptomatic or present with non‐specific symptoms such as fever, nausea/vomiting, abdominal/back pain, arthralgia/myalgia and rash [47, 49], sometimes with jaundice and less commonly with signs of portal hypertension such as edema and/or ascites. This may be due to vascular changes in the liver parenchyma, nodular regenerative hyperplasia [50] or sinusoidal obstructive syndrome (formerly known as veno‐occlusive disease) [51] with clear features of portal hypertension alone, or generalized edema associated with polyserositis [52].

Like other types of DILI, the pattern can be hepatocellular, cholestatic or mixed; however, the hepatocellular pattern of injury is much more common (Table 2) [1, 53].

TABLE 2.

Hepatocellular versus cholestatic pattern of liver injury in ILICI.

Pattern of liver injury (R value = (ALT/ULN)/(ALP/ULN))
Hepatocellular (cytolytic) (R ≥ 5)

Radiology

  • Normal liver

  • Hepatomegaly

  • Portal hypertension

  • Periportal lymphadenopathy

Histology

  • Lobular hepatitis with polymorphic infiltration

  • Spotty or confluent necrosis predominantly in the centrilobular zone

  • Endothelialitis

  • Granulomatous hepatitis

  • Fibrin ring granuloma

Cholestatic (R ≤ 2)
  • Normal liver

  • Cholangitis with mixed inflammatory infiltrate

  • Dilatation and strictures segmental or diffuse (sclerosing cholangitis aspect)

  • Lymphocytic cholangitis

  • Bile duct dystrophy and proliferation

  • Thickening of the bile ducts and gallbladder wall

  • Concentric fibrosis and complete obliteration of the bile duct

  • Vanishing bile duct syndrome

  • Granulomatous cholangitis

  • Fibrin ring granuloma

  • Cholestasis

Mixed (2 < R < 5)
  • All features mentioned above can be present

  • All features mentioned above can be present

Secondary Sclerosing Cholangitis (SSC) is a manifestation of ILICI and may involve small and/or large bile ducts or both and can present with periductal fibrosis on liver biopsy, cholangitis and intraepithelial inflammatory injury, and even vanishing bile ducts [54]. In laboratories, these patients have a very elevated ALP often in the thousands. There is hypertrophy of the bile duct wall and an absence of obstructive pathology [55, 56, 57]. Biliary strictures and areas of dilation are seen intra‐ and extra hepatically on imaging studies such as magnetic resonance cholangiopancreatography (MRCP) resembling primary sclerosing cholangitis (PSC) [54, 55, 56, 57, 58, 59, 60, 61]. This cholestatic pattern of DILI which mimicks PSC has been associated in particular with anti‐PD1 therapy, such as pembrolizumab and nivolumab, and responds poorly to immune suppression (discussed below) [54, 61, 62]. The hepatocellular pattern of liver injury appears to be more frequently associated with combination therapy (dual blockade) and anti‐CTLA4 agents [53]. The distinction seems important for differential diagnosis and management. Patients may experience mild to severe abnormalities in liver tests. In addition, jaundice and hyperbilirubinemia are seen in up to a quarter of patients with SSC which is associated with a more severe outcome [63].

Mortality from ILICI has been reported although this is generally rare [64, 65, 66]. Systematic reviews estimate the incidence of death from fulminant hepatitis to be between 0.5% and 0.07% [67, 68]. Wang et al. found 613 fatal ICI toxic events from 2009 through January 2018 in VigiLyze internationally, 22% of which were attributed to hepatitis [68]. In another study utilising the World Health Organisation's large VigiBase database of individual safety case reports, death from fatal hepatitis occurred in 19% (94/490) of cases, with only age ≥ 65 years being a risk factor [44]. Indeed, some patients exhibited evidence of multiple organ toxicity. Therefore, in the setting of underlying cancer, it is not readily apparent whether the ultimate cause of death in these studies was fulminant hepatitis and liver‐related mortality.

The diagnosis of ILICI, like other forms of hepatotoxicity, is a diagnosis of exclusion and requires ruling out other causes. Causality assessment also involves considering the clinical presentation, latency and careful study of other medications administered and is beyond the scope of this review [69, 70, 71, 72]. Importantly, the very long half‐life of these drugs and the fact that active drug levels persist in patients’ weeks after their last dose should be taken into account during DILI causality assessment.

5. Treatment

Treatment of ILICI depends on the severity of the presentation. Severity is categorized by the degree of elevation over upper limits of normal (ULN) and is most commonly categorized from 1 to 4 according to the Common Terminology Criteria for Adverse Events (CTCAE) classification, with severe ILICI encompassing grades 3–4 [1, 73]. Although this classification has recently been challenged, the utilisation of an updated Roussel Uclaf Causality Assessment Method has been proposed to show a lower incidence of severe cases, resulting in less steroid usage [74]. Mild transaminase elevation and low‐grade ILICI require discontinuation of the ICI and strict follow‐up, but not necessarily high‐dose corticosteroids, as spontaneous improvement has been observed [45, 47, 75]. Despite the fact that a precise cut‐off of liver tests for prognosis has not yet been identified, the kinetics of liver test evolution should guide treatment decisions.

Indeed, high doses of steroids are required in cases of hyperbilirubinemia or impending liver failure. Although there has been some recent controversy as a recent retrospective study on 215 patients with Grade ≥ 3, ILICI showed that there was no significant benefit from doses above 1.5 mg/kg/day [76]. The benefits of high‐dose steroids must be balanced against the risk of side effects such as infections and glycemic control [76, 77]. In general, steroids for irAEs are associated with improved overall survival, however, the long‐term impact of high doses of steroids on tumor progression is still controversial [78]. Early steroid use, within 60 days of initiating ICI, has been shown to be detrimental and is independently associated with lower survival benefit regardless of ICI continuation or discontinuation [78, 79]. This could be interpreted as evidence for the fact that systemic immunosuppression counteracts the beneficial effect of immune activation by immunotherapy. However, when interpreting these findings, variables such as steroid dosage and cancer stage should be considered. To avoid high doses of corticosteroids, budesonide may appear to be an attractive option [80]; however, its use in patients with acute flares of autoimmune hepatitis is inferior to prednisone [81] and cohort studies regarding its efficacy for ILICI are lacking.

For cases of mild cholestatic ILICI, Ursodeoxycholic Acid (UDCA) has been used and may help in improving serum liver tests, although still controversial and not routinely recommended [63, 71, 73, 82, 83]. The use of UDCA has not been established as a treatment for cholestatic DILI due to the lack of adequate evidence. However, it does not appear to be harmful [71, 84]. In general, UDCA is well tolerated, and there is minimal risk associated with its use in ILICI; however, large, randomized controlled trials are needed to determine its efficacy, optimal dose and duration of therapy [46, 53, 71]. Due to the risk of SSC, an MRCP is recommended for those with severe cholestatic liver injury or progressive jaundice to look for these rare cases that have recently been recognized as an important clinical entity due to the therapeutic challenges they pose. Pembrolizumab and nivolumab in particular have been implicated in this form of cholestatic ILICI, although other ICIs, and in particular PD‐1 inhibitors, can cause SSC [54, 55, 56, 57]. Corticosteroids in combination with UDCA have been used to treat cholestatic ILICI although the results are mixed, and many patients with ICI‐mediated cholangitis do not respond to steroids [54].

Risk factors for steroid‐refractory cases of hepatitis include previous liver comorbidity, hyperbilirubinemia, and general symptoms associated with hepatitis such as fever, jaundice, ascites and/or encephalopathy [85]. There are no randomized controlled trials or high‐quality studies with level‐one evidence, or FDA‐approved medications indicated for management of steroid‐refractory ILICI. The information summarized herein are a compilation of published case reports, case series, or retrospective experiences from various centres (Table 3).

TABLE 3.

Immunosuppressive therapy reported in the literature for ILICI. a

Line of therapy Drug Comments
First line Corticosteroids

Steroids remain first line; however, there is no consensus regarding time of administration (according to severity), route, doses and time of escalating doses. In general, once liver enzymes decreased to less than 5× ULN, a slow taper over 30 days with close lab follow‐up is recommended [45, 74, 77, 83, 84]

UDCA

Either alone or with corticosteroids for cholestatic or mixed liver injury pattern recommended by the European Society of Medical Oncology (ESMO), not in the US. There is no consensus in its use, but it is considered a safe drug [45, 83, 94]

Second line (recommended as second line by most guidelines) MMF

AZA has been used [88, 90] but there are more data with MMF [24, 52, 85, 86, 87, 88, 89, 90, 91]

Third line (several options exist and have been reported in the literature, but experience is limited) Tacrolimus

Calcineurin inhibitor. Several case reports showed its efficacy. Cyclosporine is used in other irAEs, not much data in ILICI [92, 93, 94, 95, 96, 97, 98]

ATG

T‐cell‐depleting agent. Successfully used in several case reports [103, 104, 105]

Tocilizumab

Interleukin‐6 (IL‐6) receptor blockers. Emerging strategy with several successful case reports [106, 107, 108, 109, 110]

Plasma exchange +/− IVIG

Technique to remove circulating autoantibodies and other humoral factors +/− polyclonal serum IgG, anti‐inflammatory and immunomodulatory treatment.

Emerging strategy with several successful case reports [84, 112, 113, 114, 115]

Tofacitinib

Janus kinase (JAK) inhibitor. Just one report. More frequently used for other irAEs [102]

a

Many of the reports reviewed here are single cases or small case series where an agent was used to treat ILICI. There are no large prospective studies on this topic. Clinicians should use their own judgement when treating patients, and risk–benefit analysis should be undertaken in a multidisciplinary fashion before the off‐label use of these medications.

Patients who are refractory to corticosteroids should be treated with second‐line immunosuppressive therapy, the most commonly used drug being mycophenolate mofetil (MMF), which is successful in 82% of patients with hepatocellular injury [54, 85, 86, 87]. A recent meta‐analysis of 30 studies that included 1120 patients showed that the pooled ILICI response rate was 79% (95% CI 0.73–0.84) for treatment with corticosteroids and 93% (95% CI 0.79–1.0) for treatment with MMF [87].

Azathioprine has been reported as a successful therapy for steroid‐resistant ILICI presenting as hepatitis [88] but is less commonly reported in the literature in comparison to MMF. Several third‐line therapies have been described in case reports with some differences depending on the organ affected by the toxicity [89]. Calcineurin inhibitors (CNIs), tacrolimus and cyclosporine are commonly used in the liver transplant setting and refractory autoimmune hepatitis (AIH) and have the advantage of being familiar and easy to use for hepatologists. CNIs inhibit the expression of IL‐2 and prevent the differentiation and maturation of T‐cells. Tacrolimus has been shown to be safe and effective in managing steroid‐refractory ILICI in several case reports [90, 91, 92, 93, 94]. Cyclosporine has been effectively used in case reports of ILICI and several non‐liver irAEs including ICI‐induced dermatitis and anaemia [95, 96, 97]. Anti‐TNF antibodies that are commonly used on colitis from ICIs have also been used for the treatment of ILICI [98, 99], but due to potential hepatotoxicity and fear of liver decompensation, they are currently avoided in ILICI until further safety studies can be conducted. The Janus kinase (JAK) inhibitor, tofacitinib, which has been used for ICI‐induced colitis and myocarditis, has been used successfully in a few cases of steroid‐refractory hepatitis [100]. The use of T‐cell‐depleting agent, antithymocyte globulin, ATG has also been reported in patients with steroid side effects (steroid‐related psychosis), or those refractory to both steroids and MMF or insufficient response to MMF [101, 102, 103, 104]. Please note that while these case reports indicate these drugs are successful in controlling severe liver inflammation in some cases, the long‐term consequences of these therapies on the underlying cancer require study.

Two additional strategies seem to be emerging more recently as third‐line therapy: the use of tocilizumab or plasma exchange +/− intravenous immunoglobulin (IVIG). Tocilizumab, a monoclonal antibody that blocks the IL‐6 receptor, has been reported as effective in controlling hepatitis in several case reports [105, 106, 107, 108]. The safety and efficacy of tocilizumab which is given at a dose of 4 mg/kg IV over one hour have also been reported for the treatment of other organ irAEs or for the prevention of flares in patients with rheumatic and autoimmune diseases [109]. Another third‐line option that has been used successfully in steroid and MMF‐refractory irAEs is plasma exchange +/− IVIG [110]. A few case reports have described the successful use of this treatment in acute liver failure [111, 112, 113]. It is important to consider that there may be an element of publication bias where the unsuccessful use of these drugs is less commonly reported, and it is hard to know the true percentage of success of third‐line immunosuppressive therapies in the absence of controlled studies. Given the lack of data and unknown long‐term safety of these interventions and the effect they may have on underlying cancer, prospective randomized studies are needed to make firm recommendations as to the best option for steroid‐refractory disease [114]. We urge our colleagues to consider these options in steroid‐refractory disease, in a multidisciplinary manner, and in conversation with patients.

6. Rechallenge

Given the vast survival benefit of immunotherapy and the association between certain irAEs and tumour response to ICIs, oncologists often deem reintroduction of immune checkpoint inhibitors necessary after hepatitis resolves. The question of whether to resume ICIs after a bout of severe ILICI is patient and situation dependent and should be addressed in a multidisciplinary manner. The resumption of an ICI after experiencing toxicity, termed ‘rechallenge’, is a difficult decision, as solid biomarkers for relapse of toxicity have yet to be identified. What is clear, however, is that some form of rechallenge with alternating agent, dose or regimen is a feasible option, even in patients with grade 3 or above liver injury. In most studies, rechallenge is associated with a 20 to 35% ILICI recurrence rate, the majority of which are non‐severe [24, 53, 115, 116]. In the context of reintroducing therapy, two clinically relevant questions emerge: first, which ICI can be reintroduced, and second, whether prophylaxis with steroids is indicated to prevent relapse of liver injury. The use of an anti‐CTLA4 following DILI from an anti‐PD1/PDL1 or combination therapy appears to be associated with an elevated risk of recurrence for hepatitis and other irAEs [113, 117, 118, 119]. In general, anti‐CTLA4 inhibition is more potent and associated with more irAEs than anti‐PD1. However, the cholestatic phenotype of ILICI is more commonly seen in PD1/PDL‐1 inhibitions; therefore, the choice of ICI to rechallenge with will depend on many factors. The underlying liver disease, the presence of underlying autoimmune disorder, previous biliary injury, as well as progression of cancer, have been identified as risk factors for hepatitis recurrence [53, 115]. The prophylactic use of budesonide during rechallenge has been proposed [120]; however, there is no evidence that pre‐emptive corticosteroids have any impact on the recurrence rate of irAEs and as such cannot be recommended [53, 115].

7. Conclusion

We are just scratching the surface of how the immune system can be utilized to neutralize cancer. The future of cancer immunotherapy is poised to revolutionize how we think about cancer care, treatment, and prognosis. Novel preventive and therapeutic vaccines, cutting‐edge cell therapies, novel inhibitors that target new checkpoints, and a new era of treatment combinations will pave the future [3]. Importantly, most if not all of these treatments will alter liver immune homeostasis by affecting multiple types of immune cells and will have the unintended side effect of hepatotoxicity. Therefore, understanding ILICI mechanisms, prompt diagnosis, and novel treatment approaches focused on the liver are imperative in mitigating side effects and allowing patients to stay on their life‐saving immunotherapy.

Corticosteroids are reported to be successful in the majority of patients with ILICI, and MMF is the most commonly used second‐line therapy in steroid‐refractory hepatitis. Although there are no high‐quality prospective studies to demonstrate MMF's efficacy, it has been the most studied in retrospective series, likely due to the comfort and experience of hepatologists with its use. There are few case series and isolated case reports using third‐line agents such as tacrolimus, cyclosporine, ATG, tocilizumab, plasma exchange, and tofacitinib successfully. Data regarding the use of these medications are limited, and none are FDA approved for this indication. In addition, all of these therapeutics alter the immune balance towards immune suppression, and their long‐term use can have potential consequences on the underlying cancer, which has not been studied. More recently, ILICI with a cholestatic phenotype resembling secondary sclerosing cholangitis on imaging and biopsy has been described. This phenotype of ILICI which is more common with anti‐PD1 therapy and has been classically described with nivolumab and pembrolizumab is more resistant to steroids.

Finally, ICIs are rapidly becoming standard of care for cancer therapy. It is estimated that more than half of all tumours will at some point be given ICIs. Given their undeniable impact on improving overall survival in cancer, there is a legitimate debate about balancing risk and benefit with potential rechallenge with these medications. Consideration of rechallenge should be on a case‐by‐case basis, in a multidisciplinary manner, with alteration of regimen, dose (or both), close monitoring and follow‐up, and after careful consideration of the potential benefits and risks.

Author Contributions

L.D. and E.D.M. equally contributed in conceptualisation, drafting and critical revision of the manuscript. All authors approved the final version of the manuscript.

Conflicts of Interest

The authors declare no conflicts of interest.

Acknowledgements

We would like to thank our colleague, Ms. Helia Alavifard, for her help in crafting Figure 1.

Funding: The authors received no specific funding for this work.

Handling Editor: Luca Valenti

Contributor Information

Lily Dara, Email: lily.dara@usc.edu.

Eleonora De Martin, Email: eleonora.demartin@aphp.fr.

References

  • 1. Regev A., Avigan M. I., Kiazand A., et al., “Best Practices for Detection, Assessment and Management of Suspected Immune‐Mediated Liver Injury Caused by Immune Checkpoint Inhibitors During Drug Development,” Journal of Autoimmunity 114 (2020): 102514. [DOI] [PubMed] [Google Scholar]
  • 2. Mazzarella L., Duso B. A., Trapani D., et al., “The Evolving Landscape of 'next‐generation' Immune Checkpoint Inhibitors: A Review,” European Journal of Cancer 117 (2019): 14–31. [DOI] [PubMed] [Google Scholar]
  • 3. Kreidieh F. Y., Tawbi H. A., Alexaki A., Borghaei H., and Kandalaft L. E., “Novel Immunotherapeutics: Perspectives on Checkpoints, Bispecifics, and Vaccines in Development,” American Society of Clinical Oncology Educational Book / ASCO American Society of Clinical Oncology Meeting 43 (2023): e391278. [DOI] [PubMed] [Google Scholar]
  • 4. Yang Y., Li X., Ma Z., et al., “CTLA‐4 Expression by B‐1a B Cells Is Essential for Immune Tolerance,” Nature Communications 12, no. 1 (2021): 525. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5. Lim S. Y., Lee J. H., Gide T. N., et al., “Circulating Cytokines Predict Immune‐Related Toxicity in Melanoma Patients Receiving Anti‐PD‐1‐Based Immunotherapy,” Clinical Cancer Research 25, no. 5 (2019): 1557–1563. [DOI] [PubMed] [Google Scholar]
  • 6. Lo J. A., Kawakubo M., Juneja V. R., et al., “Epitope Spreading Toward Wild‐Type Melanocyte‐Lineage Antigens Rescues Suboptimal Immune Checkpoint Blockade Responses,” Science Translational Medicine 13, no. 581 (2021): 1–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7. Shojaie L., Ali M., Iorga A., and Dara L., “Mechanisms of Immune Checkpoint Inhibitor‐Mediated Liver Injury,” Acta Pharmaceutica Sinica B 11, no. 12 (2021): 3727–3739. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8. Deola S., Panelli M. C., Maric D., et al., “Helper B Cells Promote Cytotoxic T Cell Survival and Proliferation Independently of Antigen Presentation Through CD27/CD70 Interactions,” Journal of Immunology 180, no. 3 (2008): 1362–1372. [DOI] [PubMed] [Google Scholar]
  • 9. Dhodapkar K. M., Duffy A., and Dhodapkar M. V., “Role of B Cells in Immune‐Related Adverse Events Following Checkpoint Blockade,” Immunological Reviews 318, no. 1 (2023): 89–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10. Das R., Bar N., Ferreira M., et al., “Early B Cell Changes Predict Autoimmunity Following Combination Immune Checkpoint Blockade,” Journal of Clinical Investigation 128, no. 2 (2018): 715–720. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11. Nishimura H., Minato N., Nakano T., and Honjo T., “Immunological Studies on PD‐1 Deficient Mice: Implication of PD‐1 as a Negative Regulator for B Cell Responses,” International Immunology 10, no. 10 (1998): 1563–1572. [DOI] [PubMed] [Google Scholar]
  • 12. de Moel E. C., Rozeman E. A., Kapiteijn E. H., et al., “Autoantibody Development Under Treatment With Immune‐Checkpoint Inhibitors,” Cancer Immunology Research 7, no. 1 (2019): 6–11. [DOI] [PubMed] [Google Scholar]
  • 13. Kumar P., Saini S., and Prabhakar B. S., “Cancer Immunotherapy With Check Point Inhibitor Can Cause Autoimmune Adverse Events due to Loss of Treg Homeostasis,” Seminars in Cancer Biology 64 (2020): 29–35. [DOI] [PubMed] [Google Scholar]
  • 14. Lechner M. G., Cheng M. I., Patel A. Y., et al., “Inhibition of IL‐17A Protects Against Thyroid Immune‐Related Adverse Events While Preserving Checkpoint Inhibitor Antitumor Efficacy,” Journal of Immunology 209, no. 4 (2022): 696–709. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15. von Euw E., Chodon T., Attar N., et al., “CTLA4 Blockade Increases Th17 Cells in Patients With Metastatic Melanoma,” Journal of Translational Medicine 7 (2009): 35. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16. Gudd C. L., Au L., Triantafyllou E., et al., “Activation and Transcriptional Profile of Monocytes and CD8+ T Cells Are Altered in Checkpoint Inhibitor‐Related Hepatitis,” Journal of Hepatology 75 (2021): 177–189. [DOI] [PubMed] [Google Scholar]
  • 17. Shojaie L., Bogdanov J. M., Alavifard H., et al., “Innate and Adaptive Immune Cell Interaction Drives Inflammasome Activation and Hepatocyte Apoptosis in Murine Liver Injury From Immune Checkpoint Inhibitors,” Cell Death and Disease 15, no. 2 (2024): 1–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Unger J. M., Vaidya R., Albain K. S., et al., “Sex Differences in Risk of Severe Adverse Events in Patients Receiving Immunotherapy, Targeted Therapy, or Chemotherapy in Cancer Clinical Trials,” Journal of Clinical Oncology: Official Journal of the American Society of Clinical Oncology 40, no. 13 (2022): 1474–1486. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19. Wahli M. N., Hayoz S., Hoch D., et al., “The Role of Immune Checkpoint Inhibitors in Clinical Practice: An Analysis of the Treatment Patterns, Survival and Toxicity Rates by Sex,” Journal of Cancer Research and Clinical Oncology 149, no. 7 (2023): 3847–3858. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20. Xiao T., Lee J., Gauntner T. D., Velegraki M., Lathia J. D., and Li Z., “Hallmarks of Sex Bias in Immuno‐Oncology: Mechanisms and Therapeutic Implications,” Nature Reviews. Cancer 24, no. 5 (2024): 338–355. [DOI] [PubMed] [Google Scholar]
  • 21. Liu M., Christ L., Richters A., and Ozdemir B. C., “Toxicity, Disease Management and Outcome of Treatment With Immune Checkpoint Inhibitors by Sex in Patients With Cancer and Preexisting Autoimmune Disease,” Oncology Letters 26, no. 3 (2023): 377. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22. Kartolo A., Sattar J., Sahai V., Baetz T., and Lakoff J. M., “Predictors of Immunotherapy‐Induced Immune‐Related Adverse Events,” Current Oncology 25, no. 5 (2018): e403–e410. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23. Miah A., Tinoco G., Zhao S., et al., “Immune Checkpoint Inhibitor‐Induced Hepatitis Injury: Risk Factors, Outcomes, and Impact on Survival,” Journal of Cancer Research and Clinical Oncology 149, no. 5 (2023): 2235–2242. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24. Atallah E., Welsh S. J., O'Carrigan B., et al., “Incidence, Risk Factors and Outcomes of Checkpoint Inhibitor‐Induced Liver Injury: A 10‐Year Real‐World Retrospective Cohort Study,” JHEP Rep 5, no. 10 (2023): 100851. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25. Pan J., Liu Y., Guo X., et al., “Risk Factors for Immune‐Mediated Hepatotoxicity in Patients With Cancer Treated With Immune Checkpoint Inhibitors: A Systematic Review and Meta‐Analysis,” Expert Opinion on Drug Safety 21, no. 10 (2022): 1275–1287. [DOI] [PubMed] [Google Scholar]
  • 26. Kawano M., Yano Y., Yamamoto A., et al., “Risk Factors for Immune Checkpoint Inhibitor‐Induced Liver Injury and the Significance of Liver Biopsy,” Diagnostics (Basel) 14, no. 8 (2024): 1–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27. Wang Y., Zhou S., Yang F., et al., “Treatment‐Related Adverse Events of PD‐1 and PD‐L1 Inhibitors in Clinical Trials: A Systematic Review and Meta‐Analysis,” JAMA Oncology 5, no. 7 (2019): 1008–1019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28. Celsa C., Cabibbo G., Fulgenzi C. A. M., et al., “Characteristics and Outcomes of Immunotherapy‐Related Liver Injury in Patients With Hepatocellular Carcinoma Versus Other Advanced Solid Tumours,” Journal of Hepatology 80, no. 3 (2024): 431–442. [DOI] [PubMed] [Google Scholar]
  • 29. Mallepally N., Ali M., Ennin E., Shojaie L., et al., “The Risk of Immune‐Mediated Liver Injury From Checkpoint Inhibitors (ILICI) in HCC Is Higher Than in Non‐HCC Cancers Receiving the Same Immunotherapy Regimen,” Hepatology 76 (2022): S1–S1564. [Google Scholar]
  • 30. Parlati L., Sakka M., Retbi A., et al., “Burden of Grade 3 or 4 Liver Injury Associated With Immune Checkpoint Inhibitors,” JHEP Rep 5, no. 12 (2023): 100880. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31. Sawada K., Hayashi H., Nakajima S., Hasebe T., Fujiya M., and Okumura T., “Non‐alcoholic Fatty Liver Disease Is a Potential Risk Factor for Liver Injury Caused by Immune Checkpoint Inhibitor,” Journal of Gastroenterology and Hepatology 35, no. 6 (2020): 1042–1048. [DOI] [PubMed] [Google Scholar]
  • 32. Tison A., Garaud S., Chiche L., Cornec D., and Kostine M., “Immune‐Checkpoint Inhibitor Use in Patients With Cancer and Pre‐Existing Autoimmune Diseases,” Nature Reviews Rheumatology 18, no. 11 (2022): 641–656. [DOI] [PubMed] [Google Scholar]
  • 33. Kocheise L., Piseddu I., Vonderlin J., et al., “PD‐1/PD‐L1 Immune Checkpoint Therapy Demonstrates Favorable Safety Profile in Patients With Autoimmune and Cholestatic Liver Disease,” Frontiers in Immunology 14 (2023): 1326078. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34. Xie E., Yeo Y. H., Scheiner B., et al., “Immune Checkpoint Inhibitors for Child‐Pugh Class B Advanced Hepatocellular Carcinoma: A Systematic Review and Meta‐Analysis,” JAMA Oncology 9, no. 10 (2023): 1423–1431. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35. Robert C., Thomas L., Bondarenko I., et al., “Ipilimumab plus dacarbazine for previously untreated metastatic melanoma,” New England Journal of Medicine 364, no. 26 (2011): 2517–2526. [DOI] [PubMed] [Google Scholar]
  • 36. Chour A., Basse C., Lebosse F., Bonte P. E., Girard N., and Duruisseaux M., “Management of Sotorasib‐Related Adverse Events and Hepatotoxicities Following Anti‐PD‐(L)1 Therapy: Experience With Sotorasib in Two French Anti‐Cancer Centers and Practical Guidance Proposal,” Lung Cancer 191 (2024): 107789. [DOI] [PubMed] [Google Scholar]
  • 37. Ernst S. M., Hofman M. M., van der Horst T. E., et al., “Hepatotoxicity in Patients With Non‐small Cell Lung Cancer Treated With Sotorasib After Prior Immunotherapy: A Comprehensive Clinical and Pharmacokinetic Analysis,” eBioMedicine 102 (2024): 105074. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38. Desai A. and Dimou A., “Toxicity From Sotorasib After Immune Checkpoint Inhibitors: A Note of Caution and Reflections of Future Advancements in the Field,” Journal of Thoracic Oncology 18, no. 10 (2023): 1265–1267. [DOI] [PubMed] [Google Scholar]
  • 39. Begum P., Goldin R. D., Possamai L. A., and Popat S., “Severe Immune Checkpoint Inhibitor Hepatitis in KRAS G12C‐Mutant NSCLC Potentially Triggered by Sotorasib: Case Report,” JTO Clin Res Rep 2, no. 9 (2021): 100213. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40. Carretero‐Gonzalez A., Salamanca Santamaria J., Castellano D., and de Velasco G., “Three Case Reports: Temporal Association Between Tyrosine‐Kinase Inhibitor‐Induced Hepatitis and Immune Checkpoint Inhibitors in Renal Cell Carcinoma,” Medicine (Baltimore) 98, no. 47 (2019): e18098. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41. Fontana R. J., Li Y. J., Chen V., et al., “Genetic Variants Associated With Immune‐Mediated Liver Injury From Checkpoint Inhibitors,” Hepatol Commun 8, no. 9 (2024): 1–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42. Owen C. N., Bai X., Quah T., et al., “Delayed Immune‐Related Adverse Events With Anti‐PD‐1‐Based Immunotherapy in Melanoma,” Annals of Oncology 32, no. 7 (2021): 917–925. [DOI] [PubMed] [Google Scholar]
  • 43. Tanaka T., Sakai A., Tsujimae M., et al., “Delayed Immune‐Related Sclerosing Cholangitis After Discontinuation of Pembrolizumab: A Case Report,” World Journal of Gastroenterology 28, no. 28 (2022): 3732–3738. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44. Vozy A., De Martin E., Johnson D. B., Lebrun‐Vignes B., Moslehi J. J., and Salem J. E., “Increased Reporting of Fatal Hepatitis Associated With Immune Checkpoint Inhibitors,” European Journal of Cancer 123 (2019): 112–115. [DOI] [PubMed] [Google Scholar]
  • 45. Gauci M. L., Baroudjian B., Zeboulon C., et al., “Immune‐Related Hepatitis With Immunotherapy: Are Corticosteroids Always Needed?,” Journal of Hepatology 69, no. 2 (2018): 548–550. [DOI] [PubMed] [Google Scholar]
  • 46. De Martin E., Michot J. M., Rosmorduc O., Guettier C., and Samuel D., “Liver Toxicity as a Limiting Factor to the Increasing Use of Immune Checkpoint Inhibitors,” JHEP Rep 2, no. 6 (2020): 100170. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47. De Martin E., Michot J. M., Papouin B., et al., “Characterization of Liver Injury Induced by Cancer Immunotherapy Using Immune Checkpoint Inhibitors,” Journal of Hepatology 68, no. 6 (2018): 1181–1190. [DOI] [PubMed] [Google Scholar]
  • 48. Brahmer J. R., Drake C. G., Wollner I., et al., “Phase I Study of Single‐Agent Anti‐Programmed Death‐1 (MDX‐1106) in Refractory Solid Tumors: Safety, Clinical Activity, Pharmacodynamics, and Immunologic Correlates,” Journal of Clinical Oncology: Official Journal of the American Society of Clinical Oncology 28, no. 19 (2010): 3167–3175. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49. J. R. Patrinely, Jr. , McGuigan B., Chandra S., et al., “A Multicenter Characterization of Hepatitis Associated With Immune Checkpoint Inhibitors,” Oncoimmunology 10, no. 1 (2021): 1875639. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50. LoPiccolo J., Brener M. I., Oshima K., Lipson E. J., and Hamilton J. P., “Nodular Regenerative Hyperplasia Associated With Immune Checkpoint Blockade,” Hepatology 68, no. 6 (2018): 2431–2433. [DOI] [PubMed] [Google Scholar]
  • 51. Charvet E., Lheure C., Isnard C., et al., “Hepatic Sinusoidal Obstruction Syndrome Induced by Nivolumab in Advanced Melanoma: A Case Report,” Annals of Oncology 31, no. 5 (2020): 661–662. [DOI] [PubMed] [Google Scholar]
  • 52. Velev M., Baroudjian B., Pruvost R., et al., “Immune‐Related Generalised Oedema ‐ A New Category of Adverse Events With Immune Checkpoint Inhibitors,” European Journal of Cancer 179 (2023): 28–47. [DOI] [PubMed] [Google Scholar]
  • 53. Hountondji L., Ferreira De Matos C., Lebosse F., et al., “Clinical Pattern of Checkpoint Inhibitor‐Induced Liver Injury in a Multicentre Cohort,” JHEP Rep 5, no. 6 (2023): 100719. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54. Berry P., Kotha S., Zen Y., et al., “Immune Checkpoint Inhibitor‐Related Cholangiopathy: Novel Clinicopathological Description of a Multi‐Centre Cohort,” Liver International: Official Journal of the International Association for the Study of the Liver 43, no. 1 (2023): 147–154. [DOI] [PubMed] [Google Scholar]
  • 55. Yoshikawa Y., Imamura M., Yamaoka K., et al., “A Case With Life‐Threatening Secondary Sclerosing Cholangitis Caused by Nivolumab,” Clinical Journal of Gastroenterology 14, no. 1 (2021): 283–287. [DOI] [PubMed] [Google Scholar]
  • 56. Onoyama T., Takeda Y., Yamashita T., et al., “Programmed Cell Death‐1 Inhibitor‐Related Sclerosing Cholangitis: A Systematic Review,” World Journal of Gastroenterology 26, no. 3 (2020): 353–365. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57. Hamoir C., de Vos M., Clinckart F., Nicaise G., Komuta M., and Lanthier N., “Hepatobiliary and Pancreatic: Nivolumab‐Related Cholangiopathy,” Journal of Gastroenterology and Hepatology 33, no. 10 (2018): 1695. [DOI] [PubMed] [Google Scholar]
  • 58. Nabeshima S., Yamasaki M., Matsumoto N., et al., “Atezolizumab‐Induced Sclerosing Cholangitis in a Patient With Lung Cancer: A Case Report,” Cancer Treat Res Commun 26 (2021): 100270. [DOI] [PubMed] [Google Scholar]
  • 59. Gelsomino F., Vitale G., and Ardizzoni A., “A Case of Nivolumab‐Related Cholangitis and Literature Review: How to Look for the Right Tools for a Correct Diagnosis of This Rare Immune‐Related Adverse Event,” Investigational New Drugs 36, no. 1 (2018): 144–146. [DOI] [PubMed] [Google Scholar]
  • 60. Kawakami H., Tanizaki J., Tanaka K., et al., “Imaging and Clinicopathological Features of Nivolumab‐Related Cholangitis in Patients With Non‐small Cell Lung Cancer,” Investigational New Drugs 35, no. 4 (2017): 529–536. [DOI] [PubMed] [Google Scholar]
  • 61. Pi B., Wang J., Tong Y., Yang Q., Lv F., and Yu Y., “Immune‐Related Cholangitis Induced by Immune Checkpoint Inhibitors: A Systematic Review of Clinical Features and Management,” European Journal of Gastroenterology & Hepatology 33, no. 1S Suppl 1 (2021): e858–e867. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62. Likhitsup A. and Fontana R. J., “Diagnosis and Management of Immune Mediated Liver Injury From Checkpoint Inhibitors,” Current Opinion in Gastroenterology 40, no. 3 (2024): 164–171. [DOI] [PubMed] [Google Scholar]
  • 63. Meunier L., Hountondji L., Jantzem H., et al., “Cholangitis Induced by Immune Checkpoint Inhibitors: Analysis of Pharmacovigilance Data,” Clinical Gastroenterology and Hepatology: The Official Clinical Practice Journal of the American Gastroenterological Association 22, no. 7 (2024): 1542–1545 e1544. [DOI] [PubMed] [Google Scholar]
  • 64. Bhave P., Buckle A., Sandhu S., and Sood S., “Mortality due to Immunotherapy Related Hepatitis,” Journal of Hepatology 69, no. 4 (2018): 976–978. [DOI] [PubMed] [Google Scholar]
  • 65. Ascierto P. A., Simeone E., Sileni V. C., et al., “Clinical Experience With Ipilimumab 3 Mg/Kg: Real‐World Efficacy and Safety Data From an Expanded Access Programme Cohort,” Journal of Translational Medicine 12 (2014): 116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66. Kuraoka N., Hara K., Terai S., Yatabe Y., and Horio Y., “Peroral Cholangioscopy of Nivolumab‐Related (Induced) Ulcerative Cholangitis in a Patient With Non‐small Cell Lung Cancer,” Endoscopy 50, no. 9 (2018): E259–E261. [DOI] [PubMed] [Google Scholar]
  • 67. Zheng C., Huang S., Lin M., et al., “Hepatotoxicity of Immune Checkpoint Inhibitors: What Is Currently Known,” Hepatol Commun 7, no. 3 (2023): e0063. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68. Wang D. Y., Salem J. E., Cohen J. V., et al., “Fatal Toxic Effects Associated With Immune Checkpoint Inhibitors: A Systematic Review and Meta‐Analysis,” JAMA Oncology 4, no. 12 (2018): 1721–1728. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69. Fontana R. J., Bjornsson E. S., Reddy R., and Andrade R. J., “The Evolving Profile of Idiosyncratic Drug‐Induced Liver Injury,” Clinical Gastroenterology and Hepatology: The Official Clinical Practice Journal of the American Gastroenterological Association 21, no. 8 (2023): 2088–2099. [DOI] [PubMed] [Google Scholar]
  • 70. Rockey D. C., Seeff L. B., Rochon J., et al., “Causality Assessment in Drug‐Induced Liver Injury Using a Structured Expert Opinion Process: Comparison to the Roussel‐Uclaf Causality Assessment Method,” Hepatology 51, no. 6 (2010): 2117–2126. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71. Fontana R. J., Liou I., Reuben A., et al., “AASLD Practice Guidance on Drug, Herbal, and Dietary Supplement‐Induced Liver Injury,” Hepatology 77, no. 3 (2023): 1036–1065. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72. Chalasani N. P., Maddur H., Russo M. W., Wong R. J., and Reddy K. R., “Practice Parameters Committee of the American College of G. ACG Clinical Guideline: Diagnosis and Management of Idiosyncratic Drug‐Induced Liver Injury,” American Journal of Gastroenterology 116, no. 5 (2021): 878–898. [DOI] [PubMed] [Google Scholar]
  • 73. Schneider B. J., Naidoo J., Santomasso B. D., et al., “Management of Immune‐Related Adverse Events in Patients Treated With Immune Checkpoint Inhibitor Therapy: ASCO Guideline Update,” Journal of Clinical Oncology: Official Journal of the American Society of Clinical Oncology 39, no. 36 (2021): 4073–4126. [DOI] [PubMed] [Google Scholar]
  • 74. Hountondji L., Faure S., Palassin P., et al., “Time to Use the Right Classification to Predict the Severity of Checkpoint Inhibitor‐Induced Liver Injury, as Assessed for Causality Using the Updated RUCAM,” Alimentary Pharmacology & Therapeutics 60, no. 11‐12 (2024): 1561–1572. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75. Gauci M. L., Baroudjian B., Bederede U., et al., “Severe Immune‐Related Hepatitis Induced by Immune Checkpoint Inhibitors: Clinical Features and Management Proposal,” Clinics and Research in Hepatology and Gastroenterology 45, no. 2 (2021): 101491. [DOI] [PubMed] [Google Scholar]
  • 76. Li M., Wong D., Vogel A. S., et al., “Effect of Corticosteroid Dosing on Outcomes in High‐Grade Immune Checkpoint Inhibitor Hepatitis,” Hepatology 75, no. 3 (2022): 531–540. [DOI] [PubMed] [Google Scholar]
  • 77. Del Castillo M., Romero F. A., Arguello E., Kyi C., Postow M. A., and Redelman‐Sidi G., “The Spectrum of Serious Infections Among Patients Receiving Immune Checkpoint Blockade for the Treatment of Melanoma,” Clinical Infectious Diseases: An Official Publication of the Infectious Diseases Society of America 63, no. 11 (2016): 1490–1493. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78. Van Buren I., Madison C., Kohn A., Berry E., Kulkarni R. P., and Thompson R. F., “Survival Among Veterans Receiving Steroids for Immune‐Related Adverse Events After Immune Checkpoint Inhibitor Therapy,” JAMA Network Open 6, no. 10 (2023): e2340695. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79. Umehara K., Yama K., Goto K., et al., “Effect of Systemic Corticosteroid Therapy on the Efficacy and Safety of Nivolumab in the Treatment of Non‐Small‐Cell Lung Cancer,” Cancer Control 28 (2021): 1073274820985790. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80. Eleftheriotis G. and Skopelitis E., “Immune‐Checkpoint Inhibitor‐Associated Grade 3 Hepatotoxicity Managed With Enteric‐Coated Budesonide Monotherapy: A Case Report,” Medicine (Baltimore) 101, no. 31 (2022): e29473. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81. Diaz‐Gonzalez A., Hernandez‐Guerra M., Perez‐Medrano I., et al., “Budesonide as First‐Line Treatment in Patients With Autoimmune Hepatitis Seems Inferior to Standard Predniso(Lo)ne Administration,” Hepatology 77, no. 4 (2023): 1095–1105. [DOI] [PubMed] [Google Scholar]
  • 82. Haanen J., Obeid M., Spain L., et al., “Management of Toxicities From Immunotherapy: ESMO Clinical Practice Guideline for Diagnosis, Treatment and Follow‐Up,” Annals of Oncology 33, no. 12 (2022): 1217–1238. [DOI] [PubMed] [Google Scholar]
  • 83. Dougan M., Wang Y., Rubio‐Tapia A., and Lim J. K., “AGA Clinical Practice Update on Diagnosis and Management of Immune Checkpoint Inhibitor Colitis and Hepatitis: Expert Review,” Gastroenterology 160, no. 4 (2021): 1384–1393. [DOI] [PubMed] [Google Scholar]
  • 84. European Association for the Study of the Liver , “Electronic Address Eee, Clinical Practice Guideline Panel C, Panel m, Representative EGB. EASL Clinical Practice Guidelines: Drug‐Induced Liver Injury,” Journal of Hepatology 70, no. 6 (2019): 1222–1261. [DOI] [PubMed] [Google Scholar]
  • 85. Alouani E., Laparra A., Perret A., et al., “Immunosuppressant Mycophenolate Mofetil for Patients With Steroid‐Refractory Immune‐Related Hepatitis Induced by Checkpoint Inhibitors in Oncology,” European Journal of Cancer 193 (2023): 113313. [DOI] [PubMed] [Google Scholar]
  • 86. Luo J., Beattie J. A., Fuentes P., et al., “Beyond Steroids: Immunosuppressants in Steroid‐Refractory or Resistant Immune‐Related Adverse Events,” Journal of Thoracic Oncology 16, no. 10 (2021): 1759–1764. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87. Chen K., He J., Xu J., and Chen J., “Effectiveness of Immunosuppressant Use for the Treatment of Immune Checkpoint Inhibitor‐Induced Liver Injury: A Systematic Review and Meta‐Analysis,” Frontiers in Oncology 13 (2023): 1088741. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88. Iwamoto K., Ishitsuka Y., Tanaka R., Sekine I., and Fujimoto M., “Azathioprine Combination Therapy for Steroid‐Refractory Hepatic Immune System‐Related Adverse Events,” European Journal of Dermatology 27, no. 3 (2017): 301–303. [DOI] [PubMed] [Google Scholar]
  • 89. Ruf T., Kramer R., Forschner A., et al., “Second‐Line Therapies for Steroid‐Refractory Immune‐Related Adverse Events in Patients Treated With Immune Checkpoint Inhibitors,” European Journal of Cancer 203 (2024): 114028. [DOI] [PubMed] [Google Scholar]
  • 90. Sato K., Inoue J., Ninomiya M., et al., “Effectiveness of Tacrolimus in a Case of Immune Checkpoint Inhibitor‐Induced Hepatotoxicity That Was Refractory to Steroids and Mycophenolate Mofetil,” Clinical Journal of Gastroenterology 16, no. 5 (2023): 720–725. [DOI] [PubMed] [Google Scholar]
  • 91. Ea V., Ngu N. L. Y., Kua H. W., and Mishra G., “Tacrolimus for the Management of Delayed Onset and Treatment‐Refractory Immune‐Related Hepatitis,” ACG Case Rep J 10, no. 4 (2023): e01007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92. McIlwaine S., Cullen A., Stratton L., et al., “The Use of Tacrolimus in the Management of Checkpoint Inhibitor Immunotherapy‐Induced Hepatitis,” Journal of the Royal College of Physicians of Edinburgh 52, no. 1 (2022): 20–23. [DOI] [PubMed] [Google Scholar]
  • 93. Ziogas D. C., Gkoufa A., Cholongitas E., et al., “When Steroids Are Not Enough in Immune‐Related Hepatitis: Current Clinical Challenges Discussed on the Basis of a Case Report,” Journal for Immunotherapy of Cancer 8, no. 2 (2020): e001322. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94. Beardslee T., Draper A., and Kudchadkar R., “Tacrolimus for the Treatment of Immune‐Related Adverse Effects Refractory to Systemic Steroids and Anti‐Tumor Necrosis Factor Alpha Therapy,” Journal of Oncology Pharmacy Practice 25, no. 5 (2019): 1275–1281. [DOI] [PubMed] [Google Scholar]
  • 95. Gerard A., Romani S., Van‐Obberghen E., et al., “Case Report: Successful Treatment of Steroid‐Refractory Immune Checkpoint Inhibitor‐Related Pure Red Cell Aplasia With Cyclosporin,” Frontiers in Oncology 10 (2020): 1760. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96. Fixsen E., Patel J., Selim M. A., and Kheterpal M., “Resolution of Pembrolizumab‐Associated Steroid‐Refractory Lichenoid Dermatitis With Cyclosporine,” Oncologist 24, no. 3 (2019): e103–e105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97. Huffman B. M., Kottschade L. A., Kamath P. S., and Markovic S. N., “Hepatotoxicity After Immune Checkpoint Inhibitor Therapy in Melanoma: Natural Progression and Management,” American Journal of Clinical Oncology 41, no. 8 (2018): 760–765. [DOI] [PubMed] [Google Scholar]
  • 98. Corrigan M., Haydon G., Thompson F., et al., “Infliximab for the Treatment of Refractory Immune‐Related Hepatitis Secondary to Checkpoint Inhibitors: A Case Report,” JHEP Rep 1, no. 1 (2019): 66–69. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99. Cheung V., Gupta T., Payne M., et al., “Immunotherapy‐Related Hepatitis: Real‐World Experience From a Tertiary Centre,” Frontline Gastroenterol 10, no. 4 (2019): 364–371. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100. Wang M., Reynolds K. L., Montazeri K., Schaefer E. A., Sullivan R. J., and Dougan M., “Tofacitinib Is Effective in Treating Refractory Immune Checkpoint Inhibitor Hepatitis,” Clinical Gastroenterology and Hepatology: The Official Clinical Practice Journal of the American Gastroenterological Association 22, no. 7 (2024): 1539–1541 e1532. [DOI] [PubMed] [Google Scholar]
  • 101. Motomura D., Baetz T., Grin A., and Flemming J. A., “Severe Refractory Checkpoint Inhibitor‐Related Hepatitis Reversed With Anti‐Thymocyte Globulin and n‐Acetylcysteine,” Hepatology 72, no. 6 (2020): 2235–2238. [DOI] [PubMed] [Google Scholar]
  • 102. McGuire H. M., Shklovskaya E., Edwards J., et al., “Anti‐PD‐1‐Induced High‐Grade Hepatitis Associated With Corticosteroid‐Resistant T Cells: A Case Report,” Cancer Immunology, Immunotherapy 67, no. 4 (2018): 563–573. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103. Chmiel K. D., Suan D., Liddle C., et al., “Resolution of Severe Ipilimumab‐Induced Hepatitis After Antithymocyte Globulin Therapy,” Journal of Clinical Oncology: Official Journal of the American Society of Clinical Oncology 29, no. 9 (2011): e237–e240. [DOI] [PubMed] [Google Scholar]
  • 104. Ahmed T., Pandey R., Shah B., and Black J., “Resolution of Ipilimumab Induced Severe Hepatotoxicity With Triple Immunosuppressants Therapy,” BMJ Case Reports (2015): 1–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105. Ozluk A. A., Gunenc D., Yildirim S. S., and Karaca B., “Tocilizumab in the Treatment of Steroid Refractory Immune‐Related Hepatotoxicity: A Case Series and Review of the Literature,” Melanoma Research 34, no. 4 (2024): 335–342. [DOI] [PubMed] [Google Scholar]
  • 106. Ali S. B., Vembar P., Sukumaran S., Gunawardane D., Hughes T., and Smith A., “Tocilizumab in Grade 4 Hepatitis Secondary to Immune Checkpoint Inhibitor: A Case Report and Review of the Literature,” Immunotherapy 15, no. 14 (2023): 1125–1132. [DOI] [PubMed] [Google Scholar]
  • 107. Moi L., Bouchaab H., Mederos N., et al., “Personalized Cytokine‐Directed Therapy With Tocilizumab for Refractory Immune Checkpoint Inhibitor‐Related Cholangiohepatitis,” Journal of Thoracic Oncology 16, no. 2 (2021): 318–326. [DOI] [PubMed] [Google Scholar]
  • 108. Stroud C. R., Hegde A., Cherry C., et al., “Tocilizumab for the Management of Immune Mediated Adverse Events Secondary to PD‐1 Blockade,” Journal of Oncology Pharmacy Practice 25, no. 3 (2019): 551–557. [DOI] [PubMed] [Google Scholar]
  • 109. Dimitriou F., Hogan S., Menzies A. M., Dummer R., and Long G. V., “Interleukin‐6 Blockade for Prophylaxis and Management of Immune‐Related Adverse Events in Cancer Immunotherapy,” European Journal of Cancer 157 (2021): 214–224. [DOI] [PubMed] [Google Scholar]
  • 110. Katsumoto T. R., Wilson K. L., Giri V. K., et al., “Plasma Exchange for Severe Immune‐Related Adverse Events From Checkpoint Inhibitors: An Early Window of Opportunity?,” Immunother Adv 2(1):ltac012 (2022): 1–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111. Renault T., Meunier L., and Monet C., “Acute Liver Failure Following Immune Checkpoint Inhibitors,” Clinics and Research in Hepatology and Gastroenterology 47, no. 8 (2023): 102203. [DOI] [PubMed] [Google Scholar]
  • 112. Tan Y. W., Chen L., and Zhou X. B., “Efficacy of Artificial Liver Support System in Severe Immune‐Associated Hepatitis Caused by Camrelizumab: A Case Report and Review of the Literature,” World Journal of Clinical Cases 9, no. 17 (2021): 4415–4422. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113. Riveiro‐Barciela M., Munoz‐Couselo E., Fernandez‐Sojo J., Diaz‐Mejia N., Parra‐Lopez R., and Buti M., “Acute Liver Failure due to Immune‐Mediated Hepatitis Successfully Managed With Plasma Exchange: New Settings Call for New Treatment Strategies?,” Journal of Hepatology 70, no. 3 (2019): 564–566. [DOI] [PubMed] [Google Scholar]
  • 114. Verheijden R. J., van Eijs M. J. M., May A. M., van Wijk F., and Suijkerbuijk K. P. M., “Immunosuppression for Immune‐Related Adverse Events During Checkpoint Inhibition: An Intricate Balance,” Npj Precision Oncology 7, no. 1 (2023): 41. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115. Riveiro‐Barciela M., Barreira‐Diaz A., Callejo‐Perez A., et al., “Retreatment With Immune Checkpoint Inhibitors After a Severe Immune‐Related Hepatitis: Results From a Prospective Multicenter Study,” Clinical Gastroenterology and Hepatology: The Official Clinical Practice Journal of the American Gastroenterological Association 21, no. 3 (2023): 732–740. [DOI] [PubMed] [Google Scholar]
  • 116. Miller E. D., Abu‐Sbeih H., Styskel B., et al., “Clinical Characteristics and Adverse Impact of Hepatotoxicity due to Immune Checkpoint Inhibitors,” American Journal of Gastroenterology 115, no. 2 (2020): 251–261. [DOI] [PubMed] [Google Scholar]
  • 117. Zhao Q., Zhang J., Xu L., et al., “Safety and Efficacy of the Rechallenge of Immune Checkpoint Inhibitors After Immune‐Related Adverse Events in Patients With Cancer: A Systemic Review and Meta‐Analysis,” Frontiers in Immunology 12 (2021): 730320. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118. Weber J. S., Gibney G., Sullivan R. J., et al., “Sequential Administration of Nivolumab and Ipilimumab With a Planned Switch in Patients With Advanced Melanoma (CheckMate 064): An Open‐Label, Randomised, Phase 2 Trial,” Lancet Oncology 17, no. 7 (2016): 943–955. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119. Dolladille C., Ederhy S., Sassier M., et al., “Immune Checkpoint Inhibitor Rechallenge After Immune‐Related Adverse Events in Patients With Cancer,” JAMA Oncology 6, no. 6 (2020): 865–871. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120. Ziemer M., Koukoulioti E., Beyer S., Simon J. C., and Berg T., “Managing Immune Checkpoint‐Inhibitor‐Induced Severe Autoimmune‐Like Hepatitis by Liver‐Directed Topical Steroids,” Journal of Hepatology 66, no. 3 (2017): 657–659. [DOI] [PubMed] [Google Scholar]

Articles from Liver International are provided here courtesy of Wiley

RESOURCES