BPG is committed to discovery and dissemination of knowledge
Minireviews Open Access
Copyright ©The Author(s) 2025. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Hepatol. Dec 27, 2025; 17(12): 114084
Published online Dec 27, 2025. doi: 10.4254/wjh.v17.i12.114084
Drug-induced liver injury in inflammatory bowel disease: Challenges in diagnosis and monitoring
Arshdeep Singh, Arshia Bhardwaj, Ajit Sood, Department of Gastroenterology, Dayanand Medical College and Hospital, Ludhiana 141001, Punjab, India
Harmeet Kaur, Department of Obstetrics and Gynaecology, Dayanand Medical College and Hospital, Ludhiana 141001, Punjab, India
Ashutosh Bawa, Department of Surgery, Government Medical College, Srinagar 190001, Jammu and Kashmīr, India
Vandana Midha, Department of Internal Medicine, Dayanand Medical College and Hospital, Ludhiana 141001, Punjab, India
ORCID number: Arshdeep Singh (0000-0001-7163-0454); Arshia Bhardwaj (0000-0001-5820-7758); Vandana Midha (0000-0003-0192-3969); Ajit Sood (0000-0001-6961-6389).
Author contributions: Singh A was responsible for conceptualization, data curation, methodology and resources; Singh A and Bhardwaj A drafted the original manuscript; Singh A, Bhardwaj A, Kaur H, Bawa A, Midha V, and Sood A contributed to reviewing, editing, and visualization; Sood A provided supervision.
Conflict-of-interest statement: Sood A received honorarium for speaker events from Pfizer India and Takeda India. The remaining authors disclose no conflicts.
Open Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: https://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: Arshdeep Singh, Associate Professor, Department of Gastroenterology, Dayanand Medical College and Hospital, Tagore Nagar Civil Lines, Ludhiana 141001, Punjab, India. drarshdeepsingh@gmail.com
Received: September 15, 2025
Revised: October 1, 2025
Accepted: November 24, 2025
Published online: December 27, 2025
Processing time: 104 Days and 22.8 Hours

Abstract

Drug-induced liver injury (DILI) is an important but often underrecognized complication in the management of inflammatory bowel disease (IBD), particularly in patients receiving long-term immunomodulatory or biologic therapies. Agents such as thiopurines, methotrexate, anti-tumor necrosis factor agents, and newer small molecules including tofacitinib and upadacitinib have all been implicated in hepatotoxicity, with clinical manifestations ranging from asymptomatic elevations in liver enzymes to severe hepatic injury. Differentiating DILI from hepatobiliary disorders commonly associated with IBD, such as primary sclerosing cholangitis, metabolic dysfunction-associated steatotic liver disease, and autoimmune hepatitis, remains a significant diagnostic challenge. The absence of standardized monitoring protocols, coupled with the variable latency and heterogeneous presentation of DILI, further complicates early recognition and management. In this narrative review, we synthesize current evidence on the epidemiology, pathophysiological mechanisms, and clinical spectrum of DILI in IBD. We also outline diagnostic strategies, including the role and limitations of causality assessment tools, and propose practical considerations for baseline evaluation, longitudinal monitoring, and therapeutic decision-making.

Key Words: Drug-induced liver injury; Hepatotoxicity; Immunosuppressive agents; Inflammatory bowel diseases; Monitoring

Core Tip: Drug-induced liver injury (DILI) in inflammatory bowel disease (IBD) is often underrecognized due to overlapping hepatobiliary disorders and nonspecific presentations. Distinguishing true DILI from IBD-related liver disease requires systematic evaluation, multidisciplinary input, and structured monitoring strategies. This review synthesizes current evidence on the epidemiology, mechanisms, and diagnostic challenges of DILI in IBD, highlighting practical approaches to optimize early detection and safe therapeutic decision-making.



INTRODUCTION

Inflammatory bowel disease (IBD), including ulcerative colitis (UC) and Crohn’s disease (CD), is a chronic relapsing-remitting disorder that requires sustained pharmacological interventions to induce and maintain remission and to prevent disease-related complications. In the past two decades, treatment options for IBD have expanded significantly, with immunomodulators, biologics, and small molecules targeting key inflammatory pathways now forming the cornerstone of standard care. Although these therapies have transformed disease outcomes, they are not without risk.

Drug-induced liver injury (DILI) represents an important yet often underrecognized complication in the management of IBD. The liver, as the principal organ of drug metabolism, is vulnerable to diverse hepatotoxic insults. In patients with IBD, this risk is amplified by several factors, including the frequent requirement for polypharmacy, the coexistence of hepatobiliary manifestations, and increased susceptibility to concomitant infections. Clinically, DILI exhibits a heterogeneous spectrum, manifesting as hepatocellular, cholestatic, or mixed patterns of liver injury. DILI can be broadly categorized into two types based on underlying mechanisms. Intrinsic DILI is dose-dependent, predictable, and usually develops gradually over weeks to months of therapy. In contrast, idiosyncratic DILI is dose-independent, unpredictable, and often manifests abruptly shortly after drug exposure[1].

Importantly, the clinical and biochemical features of DILI may overlap with other hepatobiliary disorders commonly associated with IBD, such as primary sclerosing cholangitis (PSC), autoimmune hepatitis, viral hepatitis, and metabolic dysfunction associated steatotic liver disease (MASLD). This overlap introduces significant diagnostic uncertainty and complicates the attribution of liver dysfunction to drug-related causes. Therefore, timely recognition and accurate differentiation of DILI from alternative pathologies remain a major challenge in clinical practice.

DILI also carries significant clinical consequences in the context of IBD. Hepatotoxicity may necessitate interruption or permanent discontinuation of essential therapies, potentially precipitating disease relapse or prompting a switch to alternative agents with different efficacy and safety profiles. In more severe cases, DILI can result in morbidity, hospitalization, and, albeit rarely, progression to acute liver failure or chronic liver disease. Thus, optimizing therapeutic efficacy while ensuring hepatic safety constitutes a critical aspect of IBD management.

The objective of this review is to summarize drug-specific risks of hepatotoxicity in IBD, delineate the diagnostic challenges in distinguishing DILI from competing hepatobiliary pathologies and propose strategies for monitoring for DILI in IBD.

LITERATURE REVIEW

A comprehensive electronic literature search was conducted in PubMed/MEDLINE, EMBASE, Scopus, and Cochrane Library/CENTRAL to identify studies published up to July 2025. The search strategy combined keywords and Medical Subject Headings (MeSH) terms, including: ‘Drug-induced liver injury’, ‘hepatotoxicity’, ‘5-aminosalicylates’, ‘mesalazine’, ‘thiopurine’, ‘anti-TNF’, ‘vedolizumab’, ‘ustekinumab’, ‘tofacitinib’, ‘upadacitinib’, ‘ozanimod’, ‘etrasimod’, ‘inflammatory bowel disease’, ‘ulcerative colitis’, and ‘Crohn’s disease’.

Studies were included if they reported on hepatotoxicity associated with IBD therapies in adult or pediatric populations. Exclusion criteria encompassed non-English articles, conference abstracts without full text, studies not involving IBD, and reports without clear liver-related outcomes.

EPIDEMIOLOGY AND RISK DETERMINANTS OF DILI IN IBD

The true incidence of DILI in patients with IBD remains difficult to determine[2]. This challenge largely arises from the frequent coexistence of hepatobiliary disorders in IBD which complicate the attribution of liver injury solely to pharmacological agents. Nevertheless, accumulating evidence indicates that individuals with IBD carry a higher risk of DILI compared with the general population. Although the overall risk appears broadly comparable between UC and CD, subtle differences are observed, reflecting variations in therapeutic practices, disease phenotype, and associated comorbidities.

Patient-specific risk factors significantly modulate the susceptibility to DILI[3]. Advanced age is associated with nearly a threefold increase in incidence, secondary to age-related changes in pharmacokinetics, diminished hepatic regenerative capacity, and a higher prevalence of comorbidities[4]. Polypharmacy is common in elderly patients with IBD, who often require treatment for cardiovascular, metabolic, and infectious comorbidities. This substantially increases the risk of hepatotoxicity by raising the likelihood of drug-drug interactions and adding to the cumulative hepatic burden. With improved survival and the rising prevalence of elderly-onset IBD, this subgroup is expected to contribute disproportionately to the overall burden of DILI, highlighting an important clinical concern.

Sex also appears to influence the risk of DILI. Although data specific to IBD remain scarce, studies in the general population demonstrate that females have a 1.5-1.7-fold higher risk of hepatotoxicity. This increased vulnerability is multifactorial, driven by sex-related differences in drug pharmacokinetics and pharmacodynamics, hormonal influences, and a greater propensity for aberrant immune responses following drug exposure[5,6].

The gut microbiota has recently emerged as a key determinant of pathogenesis of DILI[7,8]. Dysbiosis, a hallmark of IBD, alters microbial metabolism and affects drug biotransformation and toxicity. The interaction between host, microbiota, and drugs is bidirectional: Impaired gut barrier and dysbiosis increase translocation of bacterial products like lipopolysaccharides, triggering hepatic inflammation, while many IBD therapies-including antibiotics, immunomodulators, and biologics-can further disrupt the microbiome, exacerbating dysbiosis and hepatotoxicity.

Beyond microbiota-related mechanisms, genetic predisposition plays an important role. Polymorphisms in genes encoding drug-metabolizing enzymes and drug transporters have been implicated in interindividual variability in the risk of DILI[9]. Additional host factors such as MASLD, driven by systemic inflammation, metabolic comorbidities, and gut dysbiosis, further predispose IBD patients to hepatotoxicity while complicating diagnosis due to overlapping clinical and biochemical features[10]. Similarly, malnutrition, a frequent complication of active IBD, has been shown to impair hepatic resilience, thereby increasing susceptibility to DILI[11,12].

The interaction between host-related determinants, including age, sex, genetic susceptibility, comorbidities, and gut microbiota composition, and treatment-related exposures highlights the multifactorial nature of DILI. Nevertheless, the intrinsic hepatotoxicity of specific drugs remains a pivotal determinant of clinical outcomes. A detailed appraisal of these drug-specific risks is therefore essential to guide monitoring, facilitate timely recognition of hepatotoxicity, and inform therapeutic decision-making in clinical practice.

DRUGS IMPLICATED IN DILI IN IBD
Aminosalicylates (5-aminosalicylates)

5-aminosalicylates (5-ASA), including mesalazine and sulfasalazine, are first-line therapies in the management of mild-to-moderate UC and, to a lesser extent, in low-risk colonic CD[13]. Although hepatotoxicity with these agents is rare (0%-4%), it remains a clinically relevant concern. In a pharmacoepidemiologic analysis of 4.7 million sulfasalazine or mesalazine prescriptions, only seventeen cases of DILI were reported among patients with IBD[14].

The hepatotoxic potential of sulfasalazine is largely attributed to its sulfapyridine moiety, which can precipitate immune-mediated hypersensitivity reactions. These may manifest as cholestatic disease, granulomatous hepatitis, or, in rare instances, acute liver failure[15]. In contrast, mesalazine and its newer formulations, which avoid sulfapyridine exposure, exhibit a more favourable hepatic safety profile. Importantly, no significant differences in hepatotoxic risk have been observed among the various mesalazine formulations[16,17]. Reported patterns of mesalazine-associated DILI include mild, asymptomatic elevation of transaminases and idiosyncratic hepatocellular or cholestatic injury. Liver biopsy may reveal noncaseating granulomas with infiltration of epithelioid cells[18]. Unlike sulfasalazine associated DILI, mesalazine induced hepatotoxicity is infrequently associated with manifestations of systemic hypersensitivity such as fever, rash, or eosinophilia.

The majority of 5-ASA related hepatotoxic events resolve promptly upon discontinuation of the drug, with rapid normalization of liver biochemistry. In cases of persistent and significantly elevated transaminases or when features of systemic hypersensitivity are present, corticosteroids may be considered if liver injury does not regress after drug withdrawal.

Thiopurines

Thiopurines, including azathioprine (AZA), 6-mercaptopurine (6-MP), and 6-thioguanine (6-TG), are widely used immunomodulators in the management of IBD. AZA is a prodrug that undergoes conversion to 6-MP and subsequently to 6-TG nucleotides (6-TGN), the active metabolite that inhibits purine metabolism in activated T lymphocytes[19].

Hepatotoxicity has been reported in approximately 4%-14% of patients treated with thiopurines[20,21]. The development of hepatotoxicity is closely linked to thiopurine metabolism. 6-MP, either directly or as a metabolite of AZA, is methylated by thiopurine-S-methyltransferase (TPMT) to form 6-methylmercaptopurine (6-MMP), which is subsequently converted to 6-MMP ribonucleotides (6-MMPR). These metabolites are implicated in thiopurine-induced hepatotoxicity. The pattern of thiopurine toxicity depends on TPMT activity. Patients with low TPMT activity accumulate excessive levels of 6-TGN, predisposing to myelotoxicity. In contrast, patients with normal or high TPMT activity produce elevated levels of methylated metabolites, resulting in high 6-MMP/6-MMPR concentrations and an increased risk of hepatotoxicity[19,22]. The presence of pre-existing liver disease, including MASLD, further increases susceptibility to thiopurine-related DILI[23].

Thiopurine-related DILI encompasses a broad clinical spectrum, ranging from asymptomatic biochemical abnormalities to severe vascular complications. The most frequent presentation is transient, asymptomatic elevation of transaminases, which often resolves spontaneously and permits continuation of therapy under close monitoring. Persistent elevations may require dose reduction or drug discontinuation, and in selected cases, strategies such as split dosing or low-dose thiopurine-allopurinol co-therapy may mitigate hepatotoxicity[24-27]. Hypersensitivity reactions, typically occurring within the first three months of therapy, present as acute hepatitis and necessitate immediate drug withdrawal. Cholestatic liver injury, another early idiosyncratic manifestation, usually presents with fatigue and jaundice; pruritus is uncommon. Cholestatic liver injury generally resolves upon discontinuation, rare cases may persist or progress to vanishing bile duct syndrome[28].

Endothelial injury represents a dose-dependent pattern of hepatotoxicity and includes sinusoidal obstruction syndrome (SOS), peliosis hepatis, and nodular regenerative hyperplasia (NRH), typically arising between three months and three years of therapy[29,30]. Both AZA and 6-TG have been implicated in SOS and NRH. Clinical manifestations range from asymptomatic presentation to fatigue, hepatic dysfunction, and portal hypertension. SOS may present acutely with abdominal pain, ascites, hyperbilirubinemia, and elevation of transaminases. NRH is associated with older age, male sex, prior intestinal resection (≥ 50 cm), stricturing disease phenotype, cytopenias, elevated gamma-glutamyl transferase (GGT), elevated alkaline phosphatase (ALP), and macrocytosis[31]. In patient with endothelial injury, continuation of thiopurines is contraindicated, as ongoing therapy may lead to progression to end-stage liver disease.

Methotrexate

Methotrexate (MTX) is used for maintaining remission in corticosteroid dependent CD[32]. The mechanisms underlying MTX-induced hepatotoxicity are not fully elucidated. Proposed pathways include hepatocellular accumulation of MTX metabolites, impaired folate-dependent nucleotide synthesis, and activation of hepatic stellate cells leading to fibrosis. Genetic polymorphisms in folate-metabolizing enzymes, particularly MTHFR C677T and A1298C, have been investigated for their association with MTX toxicity. While some studies suggest an increased risk with the C677T variant, others report no consistent association[33,34].

The incidence of MTX-associated hepatotoxicity, as reflected by elevations in transaminases and treatment discontinuation due to such abnormalities, remains relatively low[35-37]. The majority of patients who develop elevated transaminases while receiving MTX have coexisting risk factors for liver injury, including obesity, dyslipidemia, type 2 diabetes, metabolic syndrome, or significant alcohol consumption[38,39]. Concerns have been raised regarding the potential risk of hepatic fibrosis with long-term MTX therapy, leading to the use of non-invasive modalities such as elastography and FibroScan for assessment of fibrosis in patients receiving MTX[40-42]. However, a meta-analysis demonstrated no significant association between cumulative MTX exposure and the development of liver injury[43]. Overall, the incidence of hepatic fibrosis or cirrhosis in IBD patients treated with MTX is low, and the occurrence of advanced fibrosis remains exceptional[44].

Despite low rates of MTX-related DILI, all patients receiving MTX should receive folic acid supplementation, either 5 mg weekly (ideally 1-2 days after the MTX dose) or 1 mg daily, as this has consistently been shown to reduce the risk of hepatotoxicity[45,46].

Anti-tumour necrosis factor agents

Anti-tumor necrosis factor (anti-TNF) agents, including infliximab (IFX), adalimumab, golimumab, and certolizumab pegol, constitute a cornerstone in the treatment of moderate-to-severe IBD. These biologics have significantly improved patient outcomes by inducing and maintaining remission, promoting mucosal healing, and reducing the need for hospitalization and surgery in both UC and CD. Despite their efficacy, anti-TNF agents have been associated with idiosyncratic and indirect hepatotoxicity.

The pathophysiology of anti-TNF-induced hepatotoxicity remains incompletely defined and is likely multifactorial[47]. Immune-mediated mechanisms are thought to be central, with genetic susceptibility influencing the risk. For example, carriage of the HLA-B39:01 allele has been linked to IFX-related hepatotoxicity[48]. A subset of patients develops an autoimmune hepatitis-like phenotype characterized by antinuclear antibody positivity, hypergammaglobulinemia, interface hepatitis, and lymphoplasmacytic infiltrates, suggesting that tumor necrosis factor-α (TNF-α) blockade may disrupt immune tolerance to hepatic autoantigens[47,49]. In addition, IFX can contribute to indirect hepatotoxicity by reactivating latent hepatitis B virus (HBV) infection due to impaired cytotoxic T-cell-mediated immunity[50]. Mechanistically, TNF-α plays a role in antiviral defence through pathways such as APOBEC-mediated degradation of HBV cccDNA; and blockade of this pathway by anti-TNFs facilitates viral replication and reactivation[51]. Pre-existing liver disease, metabolic syndrome, obesity, hepatic steatosis, and concomitant hepatotoxic medications may further enhance susceptibility to anti-TNF mediated DILI.

From an epidemiological perspective, clinically significant hepatotoxicity is uncommon, although mild biochemical abnormalities are frequently observed. Transient elevations of transaminases or ALP are the most common findings and often resolve spontaneously or after discontinuation of therapy. The usual latency period is between two weeks and six months after initiating treatment[49]. Some patients develop autoimmune hepatitis-like syndrome, which may require corticosteroid therapy if abnormalities persist after IFX withdrawal[52]. Importantly, recurrence is rare when patients are switched to other biologics such as adalimumab. Cholestatic hepatitis is less frequent but can manifest with jaundice and raised ALP, that usually resolve after drug discontinuation. Advanced fibrosis and cirrhosis remain rare[53].

A particularly important manifestation of DILI with anti TNFs is reactivation of HBV, which can potentially lead to acute liver failure, especially in hepatitis B surface antigen (HBsAg)-positive patients. The reactivation rates range from 5%-39%, and is more pronounced with IFX compared with other TNF-α inhibitors. Given the risk, universal HBV screening (HBsAg, anti-HBs, and anti-HBc) is recommended prior to initiating anti-TNF therapy. For both HBsAg-positive and HBsAg negative, anti-HBc positive patients, antiviral prophylaxis with entecavir or tenofovir is recommended throughout the duration of the therapy and continued for at least six months after discontinuation[54,55].

Monitoring during anti-TNF therapy should include liver function tests every 3-4 months, with more frequent assessments in patients with metabolic comorbidities or pre-existing liver disease. Persistent elevation of transaminases (> 3 × upper limited normal), after excluding alternative causes of liver injury, necessitates drug discontinuation.

Anti integrins

Anti-integrin agents, including vedolizumab (VDZ) and natalizumab, are biologic therapies that inhibit leukocyte trafficking to the gut, thereby attenuating intestinal inflammation while preserving systemic immune function. Natalizumab is associated with the risk of developing progressive multifocal leukoencephalopathy, which significantly limits its clinical application. VDZ, in contrast, has demonstrated a favourable safety profile and is widely used in both UC and CD.

With respect to hepatotoxicity, VDZ is generally considered safe, with rare reports of liver injury[56]. In the pivotal GEMINI phase 3 and subsequent open-label extension trials, approximately 4% of patients developed liver enzyme abnormalities, none of which required drug discontinuation[57]. Reported liver injuries with VDZ include mild elevations in transaminases, autoimmune-like hepatitis, and, less frequently, cholestatic injury[58-61]. Importantly, VDZ does not appear to worsen hepatic steatosis[62]. While the risk of HBV reactivation is considered minimal, screening for HBsAg and anti-HBc should be done prior to initiating VDZ[63].

Anti interleukins

Anti-interleukin (anti-IL) therapies represent an important advancement in the treatment of IBD. Ustekinumab, directed against the p40 subunit shared by IL-12 and IL-23, effectively inhibits both Th1- and Th17-mediated immune responses. More recently, selective anti-IL-23 agents, including risankizumab, guselkumab, and mirikizumab, have been approved for IBD, offering greater specificity. Ustekinumab has been associated with rare and typically mild elevations in liver enzymes or, occasionally, an autoimmune hepatitis-like presentation, while clinically significant liver injury remains uncommon[64,65]. Data on the newer anti-IL-23 agents remain limited due to their recent approval; however, preliminary evidence indicates a minimal and generally reversible risk of hepatotoxicity[66,67]. Nonetheless, regular monitoring of liver functions is recommended during therapy.

JAK inhibitors

JAK inhibitors, such as tofacitinib, upadacitinib, and filgotinib, represent an important class of small molecules used in the management of moderate-to-severe IBD. By targeting intracellular signalling pathways downstream of multiple cytokine receptors, these have broad immunomodulatory effects[68,69]. With regard to hepatotoxicity, mild elevations in liver enzymes have been observed, most often in patients with coexistent risk factors for liver disease. Neither registration trials nor real-world studies have demonstrated a significant risk of clinically meaningful or irreversible liver injury with JAK inhibitors[70-75]. It is also important to note that CYP3A4 is the primary enzyme responsible for the metabolism of tofacitinib. Careful monitoring for potential drug-drug interactions is recommended when tofacitinib is co-administered with agents that modulate CYP3A4 activity. Also, routine monitoring of liver function tests is advised during therapy.

S1P receptor modulators

S1P receptor modulators, including ozanimod and etrasimod, are emerging therapeutic options for UC, acting through modulation of lymphocyte trafficking to reduce intestinal inflammation. Data on their hepatic safety remain limited. Elevations in GGT have been reported in clinical trials, though without clear evidence of serious hepatotoxicity[76]. Given the paucity of long-term data and the potential for subclinical liver injury, serial monitoring of liver function tests is recommended for patients receiving S1P modulators.

Corticosteroids

Corticosteroids are frequently used for induction of remission in moderate-to-severe IBD, including acute severe UC. Although short term use of corticosteroids is generally considered hepatically safe, they can, in rare instances, precipitate hepatotoxicity. Reported manifestations include hepatic steatosis, steatohepatitis, and, less commonly, idiosyncratic liver injury[77]. These effects are often reversible upon dose reduction or discontinuation. However long-term corticosteroid exposure may contribute indirectly to metabolic risk factors such as obesity, insulin resistance, and dyslipidemia, thereby exacerbating the risk of MASLD.

Antibiotic-associated hepatotoxicity

Antibiotics are prescribed in IBD for perianal fistulizing disease, pouchitis, or infectious complications. Ciprofloxacin has been implicated in hepatotoxicity, with reported presentations ranging from cholestatic jaundice to fulminant hepatic failure, typically attributed to idiosyncratic hypersensitivity. A delayed and protracted cholestatic hepatitis with ductopenia has also been described in a patient with CD after prolonged therapy[78]. Levofloxacin has also been associated with ductopenia and vanishing bile duct syndrome[79,80]. Given their frequent use in IBD, monitoring for new-onset elevation of transaminases is suggested in patients receiving fluoroquinolones.

Hepatotoxicity of anti-tubercular therapy

The management of latent or active tuberculosis is a critical consideration in patients with IBD receiving biologic therapy, particularly anti-TNF agents, given the risk of reactivation of TB. Standard first-line anti-tubercular therapy (ATT) comprising isoniazid, rifampicin, pyrazinamide, and ethambutol is well recognized for its hepatotoxic potential, with isoniazid and pyrazinamide most frequently implicated. Hepatotoxicity may range from asymptomatic elevations in transaminases to acute hepatitis, and, in rare instances, fulminant hepatic failure[81]. The risk is further amplified in patients with underlying liver disease or concomitant use of other hepatotoxic agents. Systematic monitoring of liver function tests during ATT is essential to ensure timely detection and management of hepatotoxicity.

Table 1 summarizes the hepatotoxic profiles of drugs used in the management of IBD and provides a risk gradient for each category of drugs.

Table 1 Hepatotoxic profile of inflammatory bowel disease therapies.
Drug class
Agents
Reported hepatotoxicity
Risk profile/mechanism
Risk gradient
5-aminosalicylatesMesalazine, SulfasalazineRare hepatitis, cholestatic disease, granulomatous hepatitis, mild hepatocellular/cholestatic injury (mesalazine)Sulfapyridine moiety associated with immune hypersensitivityLow
ThiopurinesAzathioprine, 6-MP, 6-TGTransient rise in transaminases, hypersensitivity hepatitis, cholestatic liver injury, vanishing bile duct syndrome; endothelial injury (sinusoidal obstruction syndrome, nodular regenerative hyperplasia, peliosis hepatis)TPMT activity, 6-MMP accumulation; risk increases with concomitant MASLDHigh
MethotrexateRise in transaminases, steatosis, steatohepatitisRisk increases with concomitant obesity, diabetes mellitus, alcohol, metabolic syndromeModerate
Anti-TNF agentsInfliximab, adalimumab, golimumab, CertolizumabRise in transaminases, alkaline phosphatase, autoimmune hepatitis-like syndrome, cholestatic hepatitis, HBV reactivationImmune-mediated, genetic (HLA-B39:01)Moderate
Anti-integrinsVedolizumab, NatalizumabRare; mild rise in transaminases, autoimmune-like hepatitis, cholestatic injuryMinimal riskVery low
Anti-interleukinsUstekinumab (anti-IL12/23), Risankizumab, Guselkumab, Mirikizumab (anti-IL23)Rare; mild rise in transaminases, autoimmune-like hepatitisFavourable hepatic safety profile, long term data neededVerly low
JAK inhibitorsTofacitinib, Upadacitinib, FilgotinibMild transient increase in transaminases, usually with other risk factors, autoimmune-like hepatitis, HBV reactivation, no serious DILI reportedCYP3A4 metabolism; caution with CYP3A4 modulators, long term data neededLow
S1P modulatorsOzanimod, EtrasimodGamma-glutamyl transferase elevationsUnclear mechanism; clinical significance uncertain, long term data neededLow
CorticosteroidsPrednisone, Budesonide, MethylprednisoloneRare idiosyncratic reactions, HBV reactivation, long-term use associated with steatosis, MASLDIndirect metabolic injuryLow
AntibioticsCiprofloxacin (fluoroquinolones)Rare cholestatic jaundice, fulminant failure, delayed ductopenic cholestasisIdiosyncratic hypersensitivityModerate
Anti-tubercular therapyIsoniazid, Rifampicin, Pyrazinamide, EthambutolRise in transaminases, acute hepatitis, fulminant failure (isoniazid and pyrazinamide)High risk with pre-existing liver disease, polypharmacyHigh
CHALLENGES IN DIAGNOSIS OF DILI IN IBD

The diagnosis of DILI in IBD is challenging owing to overlapping clinical manifestations with IBD-associated hepatobiliary disorders, the frequent absence of specific symptoms, and the influence of multiple potential confounders (Figure 1). The key diagnostic challenges are outlined below.

Figure 1
Figure 1 Challenges in the diagnosis of drug induced liver injury in patients with inflammatory bowel disease. AIH: Autoimmune hepatitis; LFT: Liver function test; MASLD: Metabolic dysfunction associated steatotic liver disease; PSC: Primary sclerosing cholangitis; RUCAM: Roussel Uclaf Causality Assessment Method; DILI: Drug induced liver injury; IBD: Inflammatory bowel disease.
Clinical overlap

The spectrum of hepatobiliary disorders associated with IBD, including PSC, autoimmune hepatitis, and MASLD, frequently mimics the clinical and biochemical presentation of DILI[82]. Autoimmune-like hepatitis triggered by biologics, such as IFX, may be indistinguishable from de novo autoimmune hepatitis. Similarly, cholestatic injury patterns can overlap with early PSC. Differentiating DILI from these intrinsic hepatobiliary conditions requires careful longitudinal evaluation and systematic exclusion of alternative etiologies.

Nonspecific symptoms

DILI in IBD is often asymptomatic and is most commonly detected incidentally during routine laboratory monitoring. When present, clinical manifestations such as fatigue, anorexia, or mild jaundice are nonspecific and can easily be misattributed to systemic disease activity, treatment-related adverse effects, or nutritional deficiencies.

Limitations of causality assessment tools

The Roussel Uclaf Causality Assessment Method (RUCAM) remains the most widely used instrument for the attribution of DILI, offering a structured approach that incorporates latency, dechallenge, alternative causes, and response to rechallenge[83]. However, its utility in IBD is constrained by several factors. Patients with IBD frequently present with concurrent hepatobiliary disorders, making exclusion of alternative causes challenging, particularly given the fluctuating and often subclinical course of these conditions. Furthermore, IBD management frequently involves polypharmacy, with simultaneous use of thiopurines/methotrexate and biologics, all with hepatotoxic potential. As RUCAM was designed to assess a single suspect drug, its reliability diminishes when multiple agents are plausible contributors. Additionally, baseline liver enzyme abnormalities due to PSC, cholestasis, or prior drug exposure complicate the timing and pattern recognition required by RUCAM, potentially obscuring atypical or delayed presentations. Importantly, RUCAM places significant emphasis on drug withdrawal and re-exposure, both of which may be clinically impractical or unsafe in patients reliant on continuous therapy for disease control.

Role of imaging and biopsy

Imaging modalities, particularly ultrasound and magnetic resonance cholangiopancreatography (MRCP), are essential for excluding structural or biliary pathology such as PSC but have limited specificity in establishing a diagnosis of DILI. Liver biopsy remains the reference standard in ambiguous cases, providing histological characterization such as autoimmune-like hepatitis, steatosis, or cholestatic injury[84]. However, biopsy findings are not pathognomonic, and their interpretation requires integration with clinical history and pharmacological data.

Confounding factors

Multiple comorbidities may further complicate the diagnosis of DILI in IBD. Chronic viral hepatitis (HBV, HCV), alcohol consumption, metabolic syndrome, and obesity-related MASLD can independently produce biochemical and histological abnormalities. In addition, polypharmacy remains a significant confounder, as patients are often treated with several concurrent medications with overlapping hepatotoxic potential.

MONITORING OF DILI IN IBD: PRACTICAL IMPLICATIONS AND SUGGESTED APPROACHES

Effective monitoring of DILI in IBD requires a systematic and individualized strategy, reflecting the heterogeneity of hepatotoxic risks across therapeutic classes and the frequent coexistence of alternative causes of liver dysfunction (Figure 2). The following practical considerations and structured approaches can optimize monitoring in clinical practice.

Figure 2
Figure 2 Approach to drug induced liver injury in a patient with inflammatory bowel disease. 1Patients with symptomatic disease have poor outcomes. AST: Aspartate transaminase; ALT: Alanine transaminase; ALP: Alkaline phosphatase; ANA: Antinuclear antibody; ASMA: Anti-smooth muscle antibody; AMA: Anti-mitochondrial antibody; IgG: Immunoglobulin G; HBsAg: Hepatitis B surface antigen; Anti HBs: Antibody to hepatitis B surface antigen; HAV: Hepatitis A virus; HCV: Hepatitis C virus; HEV: Hepatitis E virus; LKM-1: Liver kidney microsome antibody; ULN: Upper limited normal; SOS: Sinusoidal obstruction syndrome; NRH: Nodular regenerative hyperplasia; DILI: Drug induced liver injury; IBD: Inflammatory bowel disease.
Role of causality assessment tools

The RUCAM remains a widely used tool for assessing suspected DILI, but its application in IBD is limited by polypharmacy and the presence of concomitant hepatobiliary disease. In this context, RUCAM should be regarded as an organized aid rather than a definitive arbiter. Its utility is maximized when integrated with expert clinical judgment and multidisciplinary input. Emerging digital or revised RUCAM tools may enhance reproducibility, although they require validation in IBD-specific cohorts.

Broadening the differential diagnosis

Given the clinical overlap between DILI and IBD-related hepatobiliary disorders, an expanded differential is essential. Conditions such as PSC, autoimmune hepatitis, MASLD, and cholestatic injury secondary to sepsis or ischemia should be systematically excluded. Imaging modalities, including MRCP, and liver biopsy may be necessary when uncertainty persists, particularly to differentiate DILI from chronic cholangiopathies or overlap syndromes.

Baseline assessment and serial monitoring

Accurate interpretation of potential hepatotoxicity requires robust baseline liver function tests prior to initiation of therapy initiation. Serial monitoring should follow drug-specific schedules, with closer intervals during the initial 3-6 months and longer intervals once stability is achieved. Evaluation of serial trends of liver enzymes and other biochemical markers is generally more informative than isolated elevations.

Polypharmacy and confounding agents

Many IBD patients are exposed to multiple potentially hepatotoxic agents simultaneously. In cases of suspected DILI, temporary discontinuation of non-essential drugs and therapeutic drug monitoring, when available, can help refine causality. Expert adjudication, akin to approaches used in the DILI network, is valuable when multiple plausible causative agents exist.

Non-invasive tools for assessment of fibrosis, such as transient elastography or shear-wave elastography, may be helpful in monitoring for chronic liver injury. Pharmacogenetic testing (e.g., TPMT and NUDT15 polymorphisms for thiopurines) may provide additional risk stratification, though its role in DILI is adjunctive.

Multidisciplinary management

Optimal monitoring necessitates collaboration between gastroenterologists, hepatologists, and clinical pharmacologists. Joint review of complex cases ensures accurate diagnosis, prevents unnecessary drug discontinuation, and facilitates timely intervention when true DILI occurs.

UNMET NEEDS AND FUTURE DIRECTIONS

Despite increasing recognition of DILI in IBD, substantial gaps remain in its diagnosis, monitoring, and management. No IBD-specific guidelines currently exist, and practice is often extrapolated from general hepatotoxicity recommendations, which may not account for polypharmacy, immunosuppressive regimens, or underlying hepatobiliary comorbidities. Future strategies should focus on IBD-specific algorithms integrating clinical, biochemical, and histological parameters. Artificial intelligence and machine learning models have potential to predict hepatotoxic risk using large datasets incorporating laboratory trends, pharmacogenomics, and comorbidities[85-87]. Novel biomarkers may enable earlier detection and improved risk stratification[88-90]. Large-scale registries and robust pharmacovigilance initiatives are essential for capturing real-world data across diverse therapeutic classes. Ultimately, personalized monitoring strategies, tailored to individual patient risk, are critical to optimize safety while maintaining effective IBD therapy.

CONCLUSION

DILI represents a clinically significant yet frequently under-recognized complication in patients with IBD. Its diagnosis is often challenging due to overlapping etiologies, including IBD-related hepatobiliary disorders, metabolic liver disease, viral hepatitis, and polypharmacy with potentially hepatotoxic agents. Consequently, distinguishing true DILI from alternative causes requires careful clinical assessment, integration of laboratory trends, imaging, and, when appropriate, histological evaluation. Vigilant monitoring is therefore essential, beginning with baseline liver function assessment and risk stratification, followed by serial evaluations tailored to the therapeutic agents used and individual patient risk profiles. Multidisciplinary collaboration between gastroenterologists, hepatologists, and clinical pharmacologists enhances diagnostic accuracy, guides safe continuation or discontinuation of therapy, and optimizes patient outcomes. Looking ahead, development of IBD-specific diagnostic algorithms, incorporation of novel biomarkers, and application of artificial intelligence and large-scale pharmacovigilance initiatives hold promise for advancing precision in risk prediction, early detection, and personalized monitoring strategies.

Footnotes

Provenance and peer review: Invited article; Externally peer reviewed.

Peer-review model: Single blind

Specialty type: Gastroenterology and hepatology

Country of origin: India

Peer-review report’s classification

Scientific Quality: Grade B

Novelty: Grade B

Creativity or Innovation: Grade C

Scientific Significance: Grade B

P-Reviewer: Vargas-Beltran AM, MD, Mexico S-Editor: Qu XL L-Editor: A P-Editor: Zhang YL

References
1.  Broekman MMTJ, Coenen MJH, van Marrewijk CJ, Wanten GJA, Wong DR, Verbeek ALM, Klungel OH, Hooymans PM, Guchelaar HJ, Scheffer H, Derijks LJJ, de Jong DJ; TOPIC Recruitment Team. More Dose-dependent Side Effects with Mercaptopurine over Azathioprine in IBD Treatment Due to Relatively Higher Dosing. Inflamm Bowel Dis. 2017;23:1873-1881.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 18]  [Cited by in RCA: 27]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
2.  Haque T, Sasatomi E, Hayashi PH. Drug-Induced Liver Injury: Pattern Recognition and Future Directions. Gut Liver. 2016;10:27-36.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 31]  [Cited by in RCA: 40]  [Article Influence: 4.4]  [Reference Citation Analysis (0)]
3.  Suzuki A, MinjunChen. Epidemiology and Risk Determinants of Drug-Induced Liver Injury: Current Knowledge and Future Research Needs. Liver Int. 2025;45:e16146.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 1]  [Cited by in RCA: 7]  [Article Influence: 7.0]  [Reference Citation Analysis (0)]
4.  Kobayashi T, Iwaki M, Nogami A, Yoneda M. Epidemiology and Management of Drug-induced Liver Injury: Importance of the Updated RUCAM. J Clin Transl Hepatol. 2023;11:1239-1245.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 7]  [Reference Citation Analysis (0)]
5.  Sun X, Zhang X, Liu M. Increased attention to women with drug-induced liver injury: Risk factors and early intervention. ILIVER. 2024;3:100125.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 1]  [Reference Citation Analysis (0)]
6.  Floreani A, Bizzaro D, Shalaby S, Taliani G, Burra P; Special Interest Group Gender in Hepatology of the Italian Association for the Study of the Liver (AISF). Sex disparity and drug-induced liver injury. Dig Liver Dis. 2023;55:21-28.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 17]  [Reference Citation Analysis (0)]
7.  Li G, Hou Y, Zhang C, Zhou X, Bao F, Yang Y, Chen L, Yu D. Interplay Between Drug-Induced Liver Injury and Gut Microbiota: A Comprehensive Overview. Cell Mol Gastroenterol Hepatol. 2024;18:101355.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 5]  [Reference Citation Analysis (0)]
8.  Niu MW, Chen P. Gut microbiota and drug-induced liver injury: an update. Chin Med J (Engl). 2020;133:494-495.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 2]  [Cited by in RCA: 8]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
9.  Umamaheswaran G, Kumar DK, Adithan C. Distribution of genetic polymorphisms of genes encoding drug metabolizing enzymes & drug transporters - a review with Indian perspective. Indian J Med Res. 2014;139:27-65.  [PubMed]  [DOI]
10.  Navarro P, Gutiérrez-Ramírez L, Tejera-Muñoz A, Arias Á, Lucendo AJ. Systematic Review and Meta-Analysis: Prevalence of Non-Alcoholic Fatty Liver Disease and Liver Fibrosis in Patients with Inflammatory Bowel Disease. Nutrients. 2023;15:4507.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 11]  [Cited by in RCA: 11]  [Article Influence: 5.5]  [Reference Citation Analysis (0)]
11.  Sobhonslidsuk A, Poovorawan K, Soonthornworasiri N, Pan-Ngum W, Phaosawasdi K. The incidence, presentation, outcomes, risk of mortality and economic data of drug-induced liver injury from a national database in Thailand: a population-base study. BMC Gastroenterol. 2016;16:135.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 26]  [Cited by in RCA: 33]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
12.  Ali N, Gupta N, Saravu K. Malnutrition as an important risk factor for drug-induced liver injury in patients on anti-tubercular therapy: an experience from a tertiary care center in South India. Drug Discov Ther. 2020;14:135-138.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 4]  [Cited by in RCA: 11]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
13.  Raine T, Bonovas S, Burisch J, Kucharzik T, Adamina M, Annese V, Bachmann O, Bettenworth D, Chaparro M, Czuber-Dochan W, Eder P, Ellul P, Fidalgo C, Fiorino G, Gionchetti P, Gisbert JP, Gordon H, Hedin C, Holubar S, Iacucci M, Karmiris K, Katsanos K, Kopylov U, Lakatos PL, Lytras T, Lyutakov I, Noor N, Pellino G, Piovani D, Savarino E, Selvaggi F, Verstockt B, Spinelli A, Panis Y, Doherty G. ECCO Guidelines on Therapeutics in Ulcerative Colitis: Medical Treatment. J Crohns Colitis. 2022;16:2-17.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 584]  [Cited by in RCA: 635]  [Article Influence: 211.7]  [Reference Citation Analysis (0)]
14.  Ransford RA, Langman MJ. Sulphasalazine and mesalazine: serious adverse reactions re-evaluated on the basis of suspected adverse reaction reports to the Committee on Safety of Medicines. Gut. 2002;51:536-539.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 222]  [Cited by in RCA: 224]  [Article Influence: 9.7]  [Reference Citation Analysis (0)]
15.  Khokhar OS, Lewis JH. Hepatotoxicity of agents used in the management of inflammatory bowel disease. Dig Dis. 2010;28:508-518.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 50]  [Cited by in RCA: 49]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
16.  Feagan BG, Chande N, MacDonald JK. Are there any differences in the efficacy and safety of different formulations of Oral 5-ASA used for induction and maintenance of remission in ulcerative colitis? evidence from cochrane reviews. Inflamm Bowel Dis. 2013;19:2031-2040.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 13]  [Cited by in RCA: 28]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
17.  Barnhill MS, Steinberg JM, Jennings JJ, Lewis JH. Hepatotoxicty of Agents Used in the Management of Inflammatory Bowel Disease: a 2020 Update. Curr Gastroenterol Rep. 2020;22:47.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 3]  [Cited by in RCA: 16]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
18.  Braun M, Fraser GM, Kunin M, Salamon F, Tur-Kaspa R. Mesalamine-induced granulomatous hepatitis. Am J Gastroenterol. 1999;94:1973-1974.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 42]  [Cited by in RCA: 40]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
19.  Singh A, Mahajan R, Kedia S, Dutta AK, Anand A, Bernstein CN, Desai D, Pai CG, Makharia G, Tevethia HV, Mak JW, Kaur K, Peddi K, Ranjan MK, Arkkila P, Kochhar R, Banerjee R, Sinha SK, Ng SC, Hanauer S, Verma S, Dutta U, Midha V, Mehta V, Ahuja V, Sood A. Use of thiopurines in inflammatory bowel disease: an update. Intest Res. 2022;20:11-30.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 63]  [Cited by in RCA: 74]  [Article Influence: 24.7]  [Reference Citation Analysis (0)]
20.  Chaparro M, Ordás I, Cabré E, Garcia-Sanchez V, Bastida G, Peñalva M, Gomollón F, García-Planella E, Merino O, Gutiérrez A, Esteve M, Márquez L, Garcia-Sepulcre M, Hinojosa J, Vera I, Muñoz F, Mendoza JL, Cabriada JL, Montoro MA, Barreiro-de Acosta M, Ceña G, Saro C, Aldeguer X, Barrio J, Maté J, Gisbert JP. Safety of thiopurine therapy in inflammatory bowel disease: long-term follow-up study of 3931 patients. Inflamm Bowel Dis. 2013;19:1404-1410.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 201]  [Cited by in RCA: 235]  [Article Influence: 19.6]  [Reference Citation Analysis (0)]
21.  Gisbert JP, González-Lama Y, Maté J. Thiopurine-induced liver injury in patients with inflammatory bowel disease: a systematic review. Am J Gastroenterol. 2007;102:1518-1527.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 138]  [Cited by in RCA: 137]  [Article Influence: 7.6]  [Reference Citation Analysis (0)]
22.  Appell ML, Wagner A, Hindorf U. A skewed thiopurine metabolism is a common clinical phenomenon that can be successfully managed with a combination of low-dose azathioprine and allopurinol. J Crohns Colitis. 2013;7:510-513.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 18]  [Cited by in RCA: 18]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
23.  Schröder T, Schmidt KJ, Olsen V, Möller S, Mackenroth T, Sina C, Lehnert H, Fellermann K, Büning J. Liver steatosis is a risk factor for hepatotoxicity in patients with inflammatory bowel disease under immunosuppressive treatment. Eur J Gastroenterol Hepatol. 2015;27:698-704.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 22]  [Cited by in RCA: 28]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
24.  Vasudevan A, Beswick L, Friedman AB, Moltzen A, Haridy J, Raghunath A, Sparrow M, van Langenberg D. Low-dose thiopurine with allopurinol co-therapy overcomes thiopurine intolerance and allows thiopurine continuation in inflammatory bowel disease. Dig Liver Dis. 2018;50:682-688.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 21]  [Cited by in RCA: 22]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
25.  Gargallo-Puyuelo CJ, Laredo V, Gomollón F. Thiopurines in Inflammatory Bowel Disease. How to Optimize Thiopurines in the Biologic Era? Front Med (Lausanne). 2021;8:681907.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 4]  [Cited by in RCA: 15]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
26.  Vasudevan A, Con D, De Cruz P, Sparrow MP, Friedman AB, Garg M, Kashkooli S, Gibson PR, van Langenberg DR. Clinical trial: Combination allopurinol-thiopurine versus standard thiopurine in patients with IBD escalating to immunomodulators (the DECIDER study). Aliment Pharmacol Ther. 2024;59:504-514.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 7]  [Cited by in RCA: 12]  [Article Influence: 12.0]  [Reference Citation Analysis (0)]
27.  Turbayne AK, Sparrow MP. Low-Dose Azathioprine in Combination with Allopurinol: The Past, Present and Future of This Useful Duo. Dig Dis Sci. 2022;67:5382-5391.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 15]  [Reference Citation Analysis (0)]
28.  Chouchana L, Terris B, Sogni P, Treluyer JM, Costedoat-Chalumeau N, Loriot MA. Azathioprine-induced vanishing bile duct syndrome: The value of early thiopurine metabolism assessment. Br J Clin Pharmacol. 2023;89:2625-2630.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 1]  [Reference Citation Analysis (0)]
29.  Musumba CO. Review article: the association between nodular regenerative hyperplasia, inflammatory bowel disease and thiopurine therapy. Aliment Pharmacol Ther. 2013;38:1025-1037.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 47]  [Cited by in RCA: 50]  [Article Influence: 4.2]  [Reference Citation Analysis (0)]
30.  Vernier-Massouille G, Cosnes J, Lemann M, Marteau P, Reinisch W, Laharie D, Cadiot G, Bouhnik Y, De Vos M, Boureille A, Duclos B, Seksik P, Mary JY, Colombel JF. Nodular regenerative hyperplasia in patients with inflammatory bowel disease treated with azathioprine. Gut. 2007;56:1404-1409.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 132]  [Cited by in RCA: 125]  [Article Influence: 6.9]  [Reference Citation Analysis (0)]
31.  Seksik P, Mary JY, Beaugerie L, Lémann M, Colombel JF, Vernier-Massouille G, Cosnes J. Incidence of nodular regenerative hyperplasia in inflammatory bowel disease patients treated with azathioprine. Inflamm Bowel Dis. 2011;17:565-572.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 49]  [Cited by in RCA: 44]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
32.  Sequier L, Caron B, Loeuille D, Honap S, Jairath V, Netter P, Danese S, Sibilia J, Peyrin-Biroulet L. Systematic review: Methotrexate-A poorly understood and underused medication in inflammatory bowel disease. Aliment Pharmacol Ther. 2024;60:686-700.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 7]  [Reference Citation Analysis (0)]
33.  Fisher MC, Cronstein BN. Metaanalysis of methylenetetrahydrofolate reductase (MTHFR) polymorphisms affecting methotrexate toxicity. J Rheumatol. 2009;36:539-545.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 91]  [Cited by in RCA: 82]  [Article Influence: 5.1]  [Reference Citation Analysis (0)]
34.  Cáliz R, del Amo J, Balsa A, Blanco F, Silva L, Sanmarti R, Martínez FG, Collado MD, Ramirez Mdel C, Tejedor D, Artieda M, Pascual-Salcedo D, Oreiro N, Andreu JL, Graell E, Simon L, Martínez A, Mulero J. The C677T polymorphism in the MTHFR gene is associated with the toxicity of methotrexate in a Spanish rheumatoid arthritis population. Scand J Rheumatol. 2012;41:10-14.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 49]  [Cited by in RCA: 47]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
35.  Saibeni S, Bollani S, Losco A, Michielan A, Sostegni R, Devani M, Lupinacci G, Pirola L, Cucino C, Meucci G, Basilisco G, D'Incà R, Bruno S. The use of methotrexate for treatment of inflammatory bowel disease in clinical practice. Dig Liver Dis. 2012;44:123-127.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 43]  [Cited by in RCA: 50]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
36.  Conway R, Low C, Coughlan RJ, O'Donnell MJ, Carey JJ. Risk of liver injury among methotrexate users: A meta-analysis of randomised controlled trials. Semin Arthritis Rheum. 2015;45:156-162.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 52]  [Cited by in RCA: 62]  [Article Influence: 6.2]  [Reference Citation Analysis (0)]
37.  Khan N, Abbas AM, Whang N, Balart LA, Bazzano LA, Kelly TN. Incidence of liver toxicity in inflammatory bowel disease patients treated with methotrexate: a meta-analysis of clinical trials. Inflamm Bowel Dis. 2012;18:359-367.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 50]  [Cited by in RCA: 57]  [Article Influence: 4.4]  [Reference Citation Analysis (0)]
38.  Fournier MR, Klein J, Minuk GY, Bernstein CN. Changes in liver biochemistry during methotrexate use for inflammatory bowel disease. Am J Gastroenterol. 2010;105:1620-1626.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 45]  [Cited by in RCA: 46]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
39.  Devarbhavi H, Devamsh GN, Chiranth AB, Patil M, Singh R, Parikh E, Shafiq S. Risk of Liver Fibrosis in Patients With Psoriasis on Long-term Methotrexate: Role of Cumulative Dose and Comorbidities in 483 Patients. J Clin Exp Hepatol. 2025;15:102606.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 2]  [Cited by in RCA: 1]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
40.  Cervoni JP, Alby-Lepresle B, Weil D, Zhong P, Aubin F, Wendling D, Toussirot E, Vuitton L, Carbonnel F, Blondet R, Thévenot T, Calès P, Monnet E, Di Martino V. A pragmatic non-invasive assessment of liver fibrosis in patients with psoriasis, rheumatoid arthritis or Crohn's disease receiving methotrexate therapy. Clin Res Hepatol Gastroenterol. 2020;44S:100003.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 14]  [Cited by in RCA: 20]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
41.  Barbero-Villares A, Mendoza Jiménez-Ridruejo J, Taxonera C, López-Sanromán A, Pajares R, Bermejo F, Pérez-Calle JL, Mendoza JL, Algaba A, Moreno-Otero R, Maté J, Gisbert JP; Madrid Group for the Study of Inflammatory Bowel Disease ENICMAD. Evaluation of liver fibrosis by transient elastography (Fibroscan®) in patients with inflammatory bowel disease treated with methotrexate: a multicentric trial. Scand J Gastroenterol. 2012;47:575-579.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 38]  [Cited by in RCA: 48]  [Article Influence: 3.7]  [Reference Citation Analysis (2)]
42.  Wang Y, Li Y, Liu Y, Zhang Y, Ke Z, Zhang Y, Liu Y. Patients With IBD Receiving Methotrexate Are at Higher Risk of Liver Injury Compared With Patients With Non-IBD Diseases: A Meta-Analysis and Systematic Review. Front Med (Lausanne). 2021;8:774824.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 8]  [Reference Citation Analysis (0)]
43.  Azzam A, Jiyad Z, O'Beirne J. Is methotrexate hepatotoxicity associated with cumulative dose? A systematic review and meta-analysis. Australas J Dermatol. 2021;62:130-140.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 3]  [Cited by in RCA: 17]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
44.  Di Martino V. Methotrexate-induced liver fibrosis: The end of a long-held belief. J Hepatol. 2023;78:896-897.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 10]  [Cited by in RCA: 17]  [Article Influence: 8.5]  [Reference Citation Analysis (0)]
45.  Shea B, Swinden MV, Tanjong Ghogomu E, Ortiz Z, Katchamart W, Rader T, Bombardier C, Wells GA, Tugwell P. Folic acid and folinic acid for reducing side effects in patients receiving methotrexate for rheumatoid arthritis. Cochrane Database Syst Rev. 2013;2013:CD000951.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 77]  [Cited by in RCA: 100]  [Article Influence: 8.3]  [Reference Citation Analysis (0)]
46.  Sasaki K, Tsuji T, Kimoto Y, Yanagihara Y, Masuguchi K, Chikamori A, Watanabe H, Murakami T, Oryoji D, Hashimoto M, Horiuchi T, Egashira N. Usefulness of daily folic acid supplementation during methotrexate treatment of Japanese patients with rheumatoid arthritis. Mod Rheumatol. 2021;31:108-113.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 4]  [Cited by in RCA: 5]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
47.  French JB, Bonacini M, Ghabril M, Foureau D, Bonkovsky HL. Hepatotoxicity Associated with the Use of Anti-TNF-α Agents. Drug Saf. 2016;39:199-208.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 54]  [Cited by in RCA: 48]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
48.  Bruno CD, Fremd B, Church RJ, Daly AK, Aithal GP, Björnsson ES, Larrey D, Watkins PB, Chow CR. HLA associations with infliximab-induced liver injury. Pharmacogenomics J. 2020;20:681-686.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 11]  [Cited by in RCA: 20]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
49.  Worland T, Chin KL, van Langenberg D, Garg M, Nicoll A. Retrospective study of idiosyncratic drug-induced liver injury from infliximab in an inflammatory bowel disease cohort: the IDLE study. Ann Gastroenterol. 2020;33:162-169.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 6]  [Cited by in RCA: 6]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
50.  Loomba R, Liang TJ. Hepatitis B Reactivation Associated With Immune Suppressive and Biological Modifier Therapies: Current Concepts, Management Strategies, and Future Directions. Gastroenterology. 2017;152:1297-1309.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 426]  [Cited by in RCA: 468]  [Article Influence: 58.5]  [Reference Citation Analysis (0)]
51.  Lucifora J, Xia Y, Reisinger F, Zhang K, Stadler D, Cheng X, Sprinzl MF, Koppensteiner H, Makowska Z, Volz T, Remouchamps C, Chou WM, Thasler WE, Hüser N, Durantel D, Liang TJ, Münk C, Heim MH, Browning JL, Dejardin E, Dandri M, Schindler M, Heikenwalder M, Protzer U. Specific and nonhepatotoxic degradation of nuclear hepatitis B virus cccDNA. Science. 2014;343:1221-1228.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 640]  [Cited by in RCA: 757]  [Article Influence: 68.8]  [Reference Citation Analysis (0)]
52.  Björnsson HK, Gudbjornsson B, Björnsson ES. Infliximab-induced liver injury: Clinical phenotypes, autoimmunity and the role of corticosteroid treatment. J Hepatol. 2022;76:86-92.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 13]  [Cited by in RCA: 49]  [Article Influence: 16.3]  [Reference Citation Analysis (0)]
53.  Ghabril M, Bonkovsky HL, Kum C, Davern T, Hayashi PH, Kleiner DE, Serrano J, Rochon J, Fontana RJ, Bonacini M; US Drug-Induced Liver Injury Network. Liver injury from tumor necrosis factor-α antagonists: analysis of thirty-four cases. Clin Gastroenterol Hepatol. 2013;11:558-564.e3.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 173]  [Cited by in RCA: 159]  [Article Influence: 13.3]  [Reference Citation Analysis (0)]
54.  Ali FS, Nguyen MH, Hernaez R, Huang DQ, Wilder J, Piscoya A, Simon TG, Falck-Ytter Y. AGA Clinical Practice Guideline on the Prevention and Treatment of Hepatitis B Virus Reactivation in At-Risk Individuals. Gastroenterology. 2025;168:267-284.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 3]  [Cited by in RCA: 31]  [Article Influence: 31.0]  [Reference Citation Analysis (0)]
55.  European Association for the Study of the Liver. EASL Clinical Practice Guidelines on the management of hepatitis B virus infection. J Hepatol. 2025;83:502-583.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 15]  [Cited by in RCA: 81]  [Article Influence: 81.0]  [Reference Citation Analysis (1)]
56.  Milašinović B, Vezmar Kovačević S, Marković S, Jovanović M, Knežević Ivanovski T, Kralj Đ, Svorcan P, Miljković B, Vučićević K. Real-World Safety of Vedolizumab in Inflammatory Bowel Disease: A Retrospective Cohort Study Supported by FAERS Signal Analysis. Pharmaceuticals (Basel). 2025;18:1127.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 1]  [Reference Citation Analysis (0)]
57.  Colombel JF, Sands BE, Rutgeerts P, Sandborn W, Danese S, D'Haens G, Panaccione R, Loftus EV Jr, Sankoh S, Fox I, Parikh A, Milch C, Abhyankar B, Feagan BG. The safety of vedolizumab for ulcerative colitis and Crohn's disease. Gut. 2017;66:839-851.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 509]  [Cited by in RCA: 630]  [Article Influence: 78.8]  [Reference Citation Analysis (0)]
58.  Stine JG, Wang J, Behm BW. Chronic Cholestatic Liver Injury Attributable to Vedolizumab. J Clin Transl Hepatol. 2016;4:277-280.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 1]  [Cited by in RCA: 5]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
59.  Crosewski NI, Pereira AHM, Kotze LR, Pedroso MLA, Kampa KC, Rosevics L, Furlan TK, Junior OR. Hepatotoxicity associated with vedolizumab: case report in a patient with ulcerative colitis. Eur J Gastroenterol Hepatol. 2025;37:982-984.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 1]  [Reference Citation Analysis (0)]
60.  Mascarenhas Saraiva M, Ribeiro T, Dias E, Lopes JL, Cardoso H, Macedo G. Vedolizumab-Induced Liver Injury. GE Port J Gastroenterol. 2021;28:410-415.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 5]  [Reference Citation Analysis (0)]
61.  Honap S, Sticova E, Theocharidou E, Berry P, Irving PM, Samaan MA, Kotha S. Vedolizumab-Associated Drug-Induced Liver Injury: A Case Series. Inflamm Bowel Dis. 2021;27:e32-e34.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 8]  [Cited by in RCA: 10]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
62.  Mitrovic M, Marković S, Kalaba A, Zarić D, Kralj Đ, Milić A, Svorcan P. P704 The effect of antiTNFα and anti-integrin agents on liver steatosis in inlamatory bowel disease patients with non-alcoholic fatty liver disease. J Crohns Colitis. 2023;17:i837-i837.  [PubMed]  [DOI]  [Full Text]
63.  Ng SC, Hilmi IN, Blake A, Bhayat F, Adsul S, Khan QR, Wu DC. Low Frequency of Opportunistic Infections in Patients Receiving Vedolizumab in Clinical Trials and Post-Marketing Setting. Inflamm Bowel Dis. 2018;24:2431-2441.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 74]  [Cited by in RCA: 68]  [Article Influence: 9.7]  [Reference Citation Analysis (0)]
64.  Amini S, Ansari U, Divatia M, Basra T, Mathur N. S3977 A Rare Case of Drug Induced Liver Injury Secondary to Ustekinumab and Risankizumab in a Crohn’s Disease Patient. Am J Gastroenterol. 2024;119:S2582-S2582.  [PubMed]  [DOI]  [Full Text]
65.  Samah AS, Ruth T, Roua K, Louis VR. Ustekinumab-induced autoimmune hepatitis: a case report. Pan Afr Med J. 2023;44:44.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 7]  [Reference Citation Analysis (0)]
66.  Borahmah F, Salh B, Chahal D. A228 Risankizumab - drug induced liver injury; a case report. J Can Assoc Gastroenterol. 2024;7:182-182.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 2]  [Cited by in RCA: 1]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
67.  Jasti VV, Michaels A, Govani S. S4261 Risankizumab-Associated Idiosyncratic Drug-Induced Liver Injury. Am J Gastroenterol. 2024;119:S2743-S2743.  [PubMed]  [DOI]  [Full Text]
68.  Danese S, Grisham M, Hodge J, Telliez JB. JAK inhibition using tofacitinib for inflammatory bowel disease treatment: a hub for multiple inflammatory cytokines. Am J Physiol Gastrointest Liver Physiol. 2016;310:G155-G162.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 106]  [Cited by in RCA: 136]  [Article Influence: 15.1]  [Reference Citation Analysis (0)]
69.  Pippis EJ, Yacyshyn BR. Clinical and Mechanistic Characteristics of Current JAK Inhibitors in IBD. Inflamm Bowel Dis. 2021;27:1674-1683.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 4]  [Cited by in RCA: 14]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
70.  Shimizu H, Aonuma Y, Hibiya S, Kawamoto A, Takenaka K, Fujii T, Saito E, Nagahori M, Ohtsuka K, Okamoto R. Long-term efficacy and safety of tofacitinib in patients with ulcerative colitis: 3-year results from a real-world study. Intest Res. 2024;22:369-377.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 8]  [Reference Citation Analysis (0)]
71.  Sandborn WJ, Su C, Sands BE, D'Haens GR, Vermeire S, Schreiber S, Danese S, Feagan BG, Reinisch W, Niezychowski W, Friedman G, Lawendy N, Yu D, Woodworth D, Mukherjee A, Zhang H, Healey P, Panés J; OCTAVE Induction 1, OCTAVE Induction 2, and OCTAVE Sustain Investigators. Tofacitinib as Induction and Maintenance Therapy for Ulcerative Colitis. N Engl J Med. 2017;376:1723-1736.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 896]  [Cited by in RCA: 1247]  [Article Influence: 155.9]  [Reference Citation Analysis (0)]
72.  Singh A, Mahajan R, Midha V, Kaur K, Singh D, Kaur R, Garg S, Arora K, Bansal N, Sood A. Effectiveness of Tofacitinib in Ulcerative Proctitis Compared to Left Sided Colitis and Pancolitis. Dig Dis Sci. 2024;69:1389-1402.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 5]  [Reference Citation Analysis (0)]
73.  Panés J, D'Haens GR, Sands BE, Ng SC, Lawendy N, Kulisek N, Guo X, Wu J, Vranic I, Panaccione R, Vermeire S. Analysis of tofacitinib safety in ulcerative colitis from the completed global clinical developmental program up to 9.2 years of drug exposure. United European Gastroenterol J. 2024;12:793-801.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 4]  [Cited by in RCA: 20]  [Article Influence: 20.0]  [Reference Citation Analysis (0)]
74.  Khan N, Sundararajan R, Mahmud N. Long-Term Effectiveness and Safety of Tofacitinib in a Nationwide Veterans Affairs Cohort of Ulcerative Colitis Patients. Crohns Colitis 360. 2025;7:otaf037.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 1]  [Reference Citation Analysis (0)]
75.  Klein JA, St-Pierre J, Choi NK, Garcia N, Picker EA, Rubin DT. S1495 Elevation of Liver Enzymes In Patients With Inflammatory Bowel Disease Treated With Upadacitinib Is Associated With Metabolic Risk Factors. Am J Gastroenterol. 2024;119:S1081-S1082.  [PubMed]  [DOI]  [Full Text]
76.  Argollo M, Furfaro F, Gilardi D, Roda G, Allocca M, Peyrin-Biroulet L, Danese S. Modulation of sphingosine-1-phosphate in ulcerative colitis. Expert Opin Biol Ther. 2020;20:413-420.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 14]  [Cited by in RCA: 38]  [Article Influence: 7.6]  [Reference Citation Analysis (0)]
77.  Coelho J, Ozenne V, Dray X, Chaput U, Marteau P. Case of prednisolone-induced hepatitis in a patient with ulcerative colitis. Inflamm Bowel Dis. 2013;19:E34-E35.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 1]  [Cited by in RCA: 3]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
78.  Bataille L, Rahier J, Geubel A. Delayed and prolonged cholestatic hepatitis with ductopenia after long-term ciprofloxacin therapy for Crohn's disease. J Hepatol. 2002;37:696-699.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 44]  [Cited by in RCA: 34]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
79.  Shaffer K, Subramanian R. Levofloxacin as a Cause of Vanishing Bile Duct Syndrome. Am J Gastroenterol. 2014;109:S388.  [PubMed]  [DOI]  [Full Text]
80.  Levine C, Trivedi A, Thung SN, Perumalswami PV. Severe ductopenia and cholestasis from levofloxacin drug-induced liver injury: a case report and review. Semin Liver Dis. 2014;34:246-251.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 15]  [Cited by in RCA: 21]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
81.  Ramappa V, Aithal GP. Hepatotoxicity Related to Anti-tuberculosis Drugs: Mechanisms and Management. J Clin Exp Hepatol. 2013;3:37-49.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 233]  [Cited by in RCA: 304]  [Article Influence: 25.3]  [Reference Citation Analysis (0)]
82.  Núñez F P, Castro F, Mezzano G, Quera R, Diaz D, Castro L. Hepatobiliary manifestations in inflammatory bowel disease: A practical approach. World J Hepatol. 2022;14:319-337.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 13]  [Reference Citation Analysis (4)]
83.  Danan G, Benichou C. Causality assessment of adverse reactions to drugs--I. A novel method based on the conclusions of international consensus meetings: application to drug-induced liver injuries. J Clin Epidemiol. 1993;46:1323-1330.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 1015]  [Cited by in RCA: 1083]  [Article Influence: 33.8]  [Reference Citation Analysis (0)]
84.  Ahmad J, Barnhart HX, Bonacini M, Ghabril M, Hayashi PH, Odin JA, Rockey DC, Rossi S, Serrano J, Tillmann HL, Kleiner DE; Drug-Induced Liver Injury Network. Value of liver biopsy in the diagnosis of drug-induced liver injury. J Hepatol. 2022;76:1070-1078.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 36]  [Cited by in RCA: 42]  [Article Influence: 14.0]  [Reference Citation Analysis (0)]
85.  Kantagba YMK, Barro SG, Nikiema SLW, Staccini P. Artificial Intelligence for Drug-Induced Liver Injury Prediction: A Systematic Review. Stud Health Technol Inform. 2025;328:183-187.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 1]  [Reference Citation Analysis (0)]
86.  Niu H, Alvarez-Alvarez I, Chen M. Artificial Intelligence: An Emerging Tool for Studying Drug-Induced Liver Injury. Liver Int. 2025;45:e70038.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 5]  [Reference Citation Analysis (0)]
87.  Xiao T, Liu Y, Hu K, Guo K, Zhang M, Wang T, Lei W, Wang W, Zhou S, Hu Y, Su R. Drug-induced liver injury prediction based on graph convolutional networks and toxicogenomics. PLoS Comput Biol. 2025;21:e1013423.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 2]  [Cited by in RCA: 2]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
88.  McGill MR, Jaeschke H. Biomarkers of drug-induced liver injury. Adv Pharmacol. 2019;85:221-239.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 19]  [Cited by in RCA: 43]  [Article Influence: 7.2]  [Reference Citation Analysis (0)]
89.  Thakur S, Kumar V, Das R, Sharma V, Mehta DK. Biomarkers of Hepatic Toxicity: An Overview. Curr Ther Res Clin Exp. 2024;100:100737.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 45]  [Reference Citation Analysis (0)]
90.  Fu S, Wu D, Jiang W, Li J, Long J, Jia C, Zhou T. Molecular Biomarkers in Drug-Induced Liver Injury: Challenges and Future Perspectives. Front Pharmacol. 2019;10:1667.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 47]  [Cited by in RCA: 81]  [Article Influence: 16.2]  [Reference Citation Analysis (0)]