Abstract
Monitoring for liver injury remains an important aspect of drug safety assessment, including for oncotherapeutics. When present, drug-induced liver injury may limit the use or result in the discontinuation of these agents. Drug-induced liver injury can exhibit with a wide spectrum of clinical and biochemical manifestations, ranging from transient asymptomatic elevations in aminotransferases (TAEAT) to acute liver failure. Numerous oncotherapeutics have been associated with TAEAT, with published reports indicating a phenomenon in which patients may be asymptomatic without overt liver injury despite the presence of grade ≥3 aminotransferase elevations. In this review, we discuss the occurrence of TAEAT in the context of oncology clinical trials and clinical practice, as well as the clinical relevance of this phenomenon as an adverse event in response to oncotherapeutics and the related cellular and molecular mechanisms that may underlie its occurrence. We also identify several gaps in knowledge relevant to the diagnosis and the management of TAEAT in patients receiving oncotherapeutics, and identify areas warranting further study to enable the future development of consensus guidelines to support clinical decision-making.
Key Words: adverse drug event, aminotransferases, drug-induced liver injury, oncology, cancer therapy
Hundreds of drugs are associated with drug-induced liver injury (DILI),1,2 which has a wide spectrum of clinical and biochemical manifestations, ranging from transient asymptomatic elevations in aminotransferases (TAEAT) to acute liver failure.1,3 Most cases (>90%) of DILI resolve fully within several weeks after drug discontinuation, although some cases can persist as chronic low-level enzyme elevations for 6 to 12 months despite the drug cessation.4,5 Drug adaptation may also occur, defined as the phenomenon whereby an agent fails to cause progressive worsening of DILI beyond what are generally transient, low-level, asymptomatic reversible alanine aminotransferase (ALT) elevations despite the drug continuation.6
Although international working groups have defined threshold levels of ALT or aspartate aminotransferase (AST) to distinguish acute DILI from mild elevations,7,8 the clinical significance of higher level (ie, grade 3 [G3] and grade 4 [G4]) asymptomatic transient elevations has not been extensively studied,9 especially in oncology.10 In addition, there are gaps in the understanding of mechanisms leading to significantly elevated aminotransferase levels, specifically in cases of asymptomatic and transient presentations without clinical signs of hepatocyte damage.
Herein, TAEAT is used to indicate the occurrence of transient asymptomatic G3 and G4 elevations in aminotransferases (ALT and/or AST) without associated elevations in bilirubin or alkaline phosphatase or corresponding histologic liver abnormalities. A review of TAEAT in oncology clinical trials and clinical practice is provided. In addition, the gaps in our current understanding of the phenomenon are identified, including those areas where consensus guidelines and practical suggestions for clinicians involved in managing oncology patients with elevated aminotransferases may offer value.
TAEAT DEFINITION
The Common Terminology Criteria for Adverse Events version 5 grades AST and ALT levels11 based on reference to upper limits of normal (ULN) or baseline values (if baseline is abnormal). G1 elevations are >1.0–≤3.0×ULN (1.5–3.0×baseline), and G2 elevations are >3.0–5.0×ULN/baseline, both of which are below the threshold defining acute DILI.7 Only G3 (>5.0–20.0×ULN/>5.0–20.0×baseline) and G4 (>20×ULN/(>20×baseline) elevations meet currently accepted criteria for acute DILI.7,8 Although all AST and ALT elevations require close monitoring to establish whether they are isolated and transient events and to evaluate the risk of continuing drug therapy, the heightened significance of TAEAT requires further investigation.
Just as a third clinical phenotype has been proposed for DILI (ie, indirect hepatotoxicity),12 TAEAT might be best classified as a fourth DILI phenotype (ie, part of an extended spectrum of drug adaptation). This distinguishes TAEAT from clinically overt hepatotoxicity, given that after initial aminotransferase elevations develop in patients with TAEAT, levels gradually decline over days to weeks. Thus, TAEAT are akin to the adaptive response seen among several other drug classes; notably the statins6 and tacrine, which has reached G4 ALT elevations.13
TAEAT does not always preclude continuation of the causative oncotherapeutic agent and may resolve quickly without treatment interruption, as shown for blinatumomab, tyrosine kinase inhibitors, interleukin (IL)–2, interferon-α, and fluorouracil.14,15 However, regulatory restrictions and treatment guidelines that recommend (or mandate) treatment interruption/discontinuation for asymptomatic ≥G3 AST and ALT elevations can limit data generation for further identification and characterization of TAEAT.16 Additional research is needed to establish criteria for isolated ≥G3 AST and ALT elevations, so clinicians can differentiate between those that spontaneously resolve without consequences (ie, TAEAT) and those that may progress further to liver-related symptoms such as jaundice or acute or subacute hepatic failure, that is, coagulopathy (international normalized ratio [INR] ≥1.5) and new onset encephalopathy.17,18
Elevated Aminotransferases and Risk of Liver Injury
Zimmerman1 observed that drug-induced hepatocellular jaundice potentially predicted serious and even fatal outcomes. Subsequently, “Hy law” was coined and defined as serum ALT levels >3×ULN combined with total bilirubin levels >2×ULN, after the exclusion of other underlying causes to help identify patients most likely to progress to serious liver injury.19 Elevated ALT levels are sensitive for liver injury and, although not entirely specific, are viewed as being more predictive than AST levels. Healthy liver tissue has excess bilirubin-excreting capacity; therefore, hepatic injury sufficient to cause hyperbilirubinemia (ie, 2×ULN) represents a degree of hepatocyte loss that may become irreversible.18,20,21
Hy law criteria have been historically useful for predicting serious drug-induced hepatocellular liver injury, with ~1 in 10 Hy law cases leading to death from liver-related causes or the need for liver transplant.1,4,6,18,22 Failure to detect a Hy law case in clinical trials does not imply an acceptable hepatocellular safety profile because large clinical trials (>3000 patients) are needed for a high probability of detection, trial sizes that are rare in oncology. However, the detection of ≥2 Hy law cases in clinical trials is a strong predictor of significant risk and may prevent further development.18
Even when Hy law cases are detected during clinical evaluation, the risk:benefit to the population must be considered before determining whether clinical trials should continue.10,23 In oncology, some degree of hepatotoxicity may be acceptable given the potential benefit provided.10 The US Food and Drug Administration’s (FDA) general recommendations for evaluating and monitoring symptomatic DILI in clinical trials suggest that modification for special patient populations (eg, oncology) may be needed, particularly for those with underlying hepatic involvement.10,18 Recent approvals of oncotherapeutics demonstrate that a certain degree of hepatotoxicity, with careful monitoring of hepatic function, is acceptable to bring novel potentially life-prolonging drugs to market (Table 1). For instance, the risk of hepatotoxicity among all new drug classes was highest for oncotherapeutic agents approved by the FDA in recent years.37
TABLE 1.
Drug (Ref) | Drug Class/Therapeutic Use | Warning | Liver Chemistry Elevations | Dose Modifications |
---|---|---|---|---|
Tumorocentric drugs | ||||
Selpercatinib24 | Kinase inhibitor/various solid tumors | Hepatotoxicity: monitor ALT and AST before starting the therapy and Q2W for first 3 mo, then monthly | Serious hepatic AE in 2.6% ALT increased: G3–4, 9% AST increased: G3–4, 8% Bilirubin increased: G3–4, 2% | G3–G4 AST or ALT: withhold doses until G1 or baseline Reduce dose by 2 dose levels and monitor ALT/AST weekly Increase by 1 dose level after a minimum of 2 wk |
Capmatinib25 | Kinase inhibitor/metastatic NSCLC | Hepatotoxicity: monitor liver chemistry before starting therapy and Q2W for 3 mo, then monthly | ALT increased: G3–4, 8% AST increased: G3–4, 4.9% | G3 AST or ALT without increase in bilirubin: withhold doses until recovery to baseline ALT/AST G4: permanently discontinue Hy law criteria: permanently discontinue |
Tucatinib26 | Kinase inhibitor/HER2+ breast cancer | Severe hepatotoxicity (G3–4, 9.2%); monitor ALT, AST, bilirubin before starting therapy and Q3W | ALT increased: ≥G3, 8% AST increased: ≥G3, 6% Bilirubin increased: ≥G3, 1.5% | G3 AST/ALT or G3 bilirubin: withhold until recovery to G1 or baseline levels; resume at next lower dose level G4 AST/ALT or G4 bilirubin: permanently discontinue Hy law criteria: permanently discontinue |
Entrectinib27 | Kinase inhibitor/NSCLC, solid tumors | Hepatotoxicity: monitor ALT, AST Q2W during first month and then monthly | ALT increased: G3–4, 2.9% AST increased: G3–4, 2.7% | G3–4 AST/ALT: withhold until recovery to G1 or baseline, resume at same dose if G3 event resolved within 4 wk, or a reduced dose for recurrent G3 events or G4 event Recurrent G4 AST/ALT: permanently discontinue Hy law criteria: permanently discontinue |
Pexidartinib28 | Kinase inhibitor/TGCT | Boxed warning: can cause serious and potentially fatal liver injury, available only through a restricted program | ALT increased: ≥G3, 20% AST increased: ≥G3, 12% ALP increased: ≥G3, 4.9% Bilirubin increased: ≥G3, 3.3% | ALT/AST ≥3–5×ULN: withhold and monitor weekly, if ≤3×ULN within 4 wk, resume at reduced dose; otherwise, permanently discontinue ALT/AST >5–10×ULN: withhold and monitor twice weekly, if ≤3×ULN within 4 wk, resume at reduced dose; otherwise, permanently discontinue ALT/AST >10×ULN, permanently discontinue (continue to monitor) |
Polatuzumab vedotin-piiq29 | CD79b-directed antibody-drug conjugate/relapsed or refractory diffuse large B-cell lymphoma | Hepatotoxicity; monitor liver enzymes and bilirubin | G3 and G4 transaminase elevations developed in 1.9% and 1.9%, respectively; laboratory values suggestive of DILI occurred in 2.3% of patients | Bilirubin >ULN to ≤1.5×ULN or AST >ULN; no starting dose adjustments required when administering polatuzumab vedotin to patients with mild hepatic impairment (bilirubin >ULN to ≤1.5×ULN or AST >ULN). |
Tagraxofusp-erzs30 | CD123-directed cytotoxin/BPDCN | Hepatotoxicity: monitor liver enzymes and bilirubin | ALT increased: ≥G3, 30% AST increased: ≥G3, 37% ALP increased: ≥G3, 1% Bilirubin increased: ≥G3, 0% | ALT or AST increase >5×ULN; withhold treatment until transaminase elevations are ≤2.5×ULN |
Calaspargase pegol –mknl31 | Asparagine-specific enzyme | Hepatotoxicity: monitor for toxicity through recovery from cycle | Transaminases increased, ≥G3, 52% Bilirubin increased, ≥3G, 20% | Total bilirubin >3×ULN to no more than 10×ULN; withhold treatment until total bilirubin levels go down to ≤1.5×ULN Total bilirubin >10×ULN; discontinue and do not make up for missed doses |
Larotrectinib32 | Kinase inhibitor/solid tumors with an NTRK gene fusion without a resistance mutation, that are metastatic without the option of surgical resection, with no satisfactory alternative treatments | Hepatotoxicity: monitor liver test results, including ALT and AST Q2W during the first month of treatment, then monthly and as clinically indicated | ALT increased: G3–4, 3% AST increased: G3–4, 3% ALP increased: G3–4, 3% | Withhold and modify dosage, or permanently discontinue based on severity Reduce the starting dose by 50% in patients with moderate (Child-Pugh B) to severe (Child-Pugh C) hepatic impairment |
Duvelisib33 | Kinase inhibitor/relapsed or refractory CLL or SLL, relapsed or refractory follicular lymphoma | Hepatotoxicity: monitor hepatic function | ALT or AST increase >3×ULN and total bilirubin >2×ULN, 2% Patients with B-cell malignancies ALT increased: ≥G3, 8% AST increased: ≥G3, 6% ALP increased: ≥G3, 2% Patients with CLL/SLL ALT increased: ≥G3, 7% AST increased: ≥G3, 3% ALP increased: ≥G3, 0% | G2 ALT/AST elevation (3–5×ULN): maintain dose and monitor at least weekly until return to <3×ULN G3 ALT/AST elevation (>20×ULN): withhold and monitor at least weekly until return to <3×ULN; resume treatment at same dose (first occurrence) or at reduced dose for subsequent occurrence G4 ALT/AST elevation (>20×ULN): discontinue treatment |
Binimetinib34 | Kinase inhibitor in combination with encorafenib/unresectable or metastatic melanoma with BRAF V600E or V600K mutations | Hepatotoxicity: monitor liver chemistry before and during treatment and as clinically indicated | In combination with encorafenib ALT increased: G3–4, 6% AST increased: G3–4, 2.6% ALP increased: G3–4, 0.5% | G2 AST or ALT increased: maintain dose; if no improvement within 2 wk, withhold treatment until improved to G0–1 or to pretreatment/baseline levels and then resume at the same dose G3 AST or ALT increased: for first occurrence of G3 (or recurrent G2), withhold treatment for ≤4 wk; if levels improve to G0–1 or pretreatment/baseline levels, resume at the same dose; if no improvement, discontinue. For recurrent events, consider permanent discontinuation G4 AST or ALT increased: for first occurrence, permanently discontinue or withhold treatment for ≤4 wk; if levels improve to G0–1 or pretreatment/baseline levels, resume at the same dose; if no improvement, discontinue; for recurrent events, permanent discontinuation For patients with moderate or severe hepatic impairment, the recommended dosage is 30 mg orally taken BID |
Lutetium Lu 177 dotatate35 | Radiolabeled somatostatin analog/GEP-NET | Hepatotoxicity: monitor transaminases, bilirubin and albumin | ALT increased: G3–4, 4% AST increased: G3–4, 5% ALP increased: G3–4, 5% Bilirubin increased: G3–4, 2% | Bilirubinemia >3×ULN, or hypoalbuminemia <30 g/L, with a prothrombin ratio <70%: withhold until complete resolution, resume at reduced dose; for hepatotoxicity requiring treatment delay of ≥16 wk, permanent discontinuation |
Immuno-Oncology Drugs | ||||
Cemiplimab-rwlc36 | PD-1–blocking antibody/metastatic CSCC or locally advanced CSCC not qualified surgery or curative radiation | Evaluate clinical chemistries, including hepatic and thyroid function, at baseline and periodically during treatment | Immune-mediated hepatitis: any grade, 2.1%; G4, 0.2%; G5, 0.2% AST increased: G3–4, 3% | Hepatitis: withhold if AST/ALT increases to >3×ULN/baseline to ≤10×ULN/baseline or if total bilirubin increases ≤3×ULN Discontinue if AST/ALT increases to >10×ULN/baseline or total bilirubin increases to >3×ULN |
ALP indicates alkaline phosphatase; ALT, alanine aminotransferase; AST, aspartate aminotransferase; BID, twice daily; BPDCN, blastic plasmacytoid dendric cell neoplasm; CLL, chronic lymphocytic leukemia; CSCC, cutaneous squamous cell carcinoma; DILI, drug-induced liver injury; FDA, US Food and Drug Administration; G, Grade; GEP-NET, gastroenteropancreatic neuroendocrine tumor; NSCLC, non–small cell lung cancer; NTRK, neurotrophic receptor tyrosine kinase; PD-1, programmed death receptor-1; Q2W, every 2 weeks; SCLC, small-cell lung cancer; SLL, small lymphocytic lymphoma; TGCT, tenosynovial giant cell tumor; ULN, upper limit of normal.
TAEAT IN ONCOLOGY
Monitoring for liver injury during discovery, clinical development, and postapproval phases of the drug life cycle remains essential.6 However, one third of 500 oncotherapeutic trials failed to clearly define thresholds for abnormal liver injury.38 In addition, the 2009 FDA guidance for the evaluation and management of potential hepatotoxicity18 did not specifically address risk:benefit considerations, nor did it specifically consider the phenomenon of TAEAT. Considering the risk:benefit of drugs is particularly important for patients with potentially fatal malignancies who may be prescribed agents with known adverse side effects including hepatotoxicity,10,39,40 which may otherwise preclude their use in more benign conditions. Assuming therapeutic benefits are sufficient, further data and guidance would help support the clinical community and health authorities if some instances of DILI (including TAEAT) are to be accepted.
Incidence of TAEAT
The frequency of TAEAT for tumorocentric drugs approved since 2018 is summarized in Table 2. The percentages of patients with ≥G3 aminotransferase elevations ranged from 1% to 15% for ALT and 0% to 15% for AST.
TABLE 2.
Drug (Ref) | Drug Class/Therapeutic Use | Hepatic-Related Event | Notes |
---|---|---|---|
First FDA approved in 2020 | |||
Tumorocentric therapies | |||
Belantamab41 | B-cell maturation antigen/RRMM | AST increased: G3–4: 2% ALP increased: G3–4: 1% | Patients with mild to moderate renal impairment included in pivotal study |
Decitabine/cedazuridine42 | Nucleoside metabolic inhibitor and cytidine deaminase inhibitor/myelodysplastic syndrome | Transaminase increased: G3–4, 3% | |
Pertuzumab/trastuzumab/hyaluronidase zzxf43 | HER2/neu receptor antagonists + endoglycosidase/breast cancer | ALT increased: G3–4, 1.6% AST increased: G3–4, 0.8% | |
Tazemetostat44 | EZH2 inhibitor/RR-FL | ALT increased: ≥G3, 3.4% AST increased: ≥G3, 3.5% | |
Lurbinectedin45 | Alkylating drug/metastatic SCLC | ALT increased: ≥G3, 4% AST increased: ≥G3, 2% | |
Ripretinib46 | Kinase inhibitor/GI stromal tumors | ALT increased: ≥G3, 1.2% Increased blood bilirubin: G1 and G2, 22% | |
Sacituzumab govitecan-hziy47 | Trop-2–directed antibody, topoisomerase inhibitor conjugate /triple-negative breast cancer | ALT increased: ≥G3, 2% AST increased: ≥G3, 3% | Not evaluated in patients with moderate to severe hepatic impairment; some patients with brain metastases included in trials |
Pemigatinib48 | Kinase inhibitor/metastatic cholangiocarcinoma | ALT increased: ≥G3, 4.1% AST increased: ≥G3, 6% Bilirubin increased: ≥G3, 6% | |
Tucatinib26 | Kinase inhibitor/HER2+ breast cancer | Warning: severe hepatotoxicity (≥G3, 9.2%); monitor ALT, AST, bilirubin before starting therapy and Q3W ALT increased: ≥G3, 8% AST increased: ≥G3, 6% Bilirubin increased: ≥G3, 1.5% | Patients with brain metastases eligible for clinical trials |
Selumetinib49 | Kinase inhibitor/ NF1 or PN | ALT increased: ≥G3, 4% AST increased: ≥G3, 2% | |
Avapritinib50 | Kinase inhibitor/metastatic GI stromal tumor | ALT increased: ≥G3, 0.5% AST increased: ≥G3, 1.5% Bilirubin increased: ≥G3, 9% | Excluded patients with brain metastases |
Immuno-oncology therapies | |||
Tafasitamab (in combination with lenalidomide)51 | CD19-directed cytolytic antibody/ RR-DLBCL | AST increased: ≥G3: 0% Albumin decreased: ≥G3; 0% APPT increased: ≥G3: 4.1 | |
Brexucabtagene autoleucel52 | CD19-directed immunotherapy/RR-MCL | ALT increased: ≥G3, 15% AST increased: ≥G3, 15% | Patients with brain metastases excluded from pivotal study |
First FDA approved in 2019 | |||
Tumorocentric therapies | |||
Fam-trastuzumab deruxtecan-nxki53 | HER2-directed antibody drug conjugate/HER2+ breast cancer | ALT increased: ≥G3, 0.9% AST increased: ≥G3, 0.4% | Bone metastases in 31%/brain metastases in 13% |
Zanubrutinib54 | Kinase inhibitor/MCL | ALT increased: ≥G3, 0.9% Bilirubin increased: ≥G3, 0.9% | Hepatic enzymes ≤2.5×ULN |
Darolutamide55 | Androgen receptor inhibitor/CRPC | AST increased: ≥G3, 0.5% Bilirubin increased: ≥G3, 0.1% | |
Alpelisib56 | Kinase inhibitor/ advanced or metastatic breast cancer | ALT increased: G3–4, 3.5% | |
Ivosidenib57 | IDH1 inhibitor/AML | Newly diagnosed AML: ALT increased: ≥G3, 4% AST increased: ≥G3, 4% ALP increased: ≥G3, 0% Relapsed or refractory AML: ALT increased: ≥G3, 1% AST increased: ≥G3, 1% ALP increased: ≥G3, 1% Bilirubin increased: ≥G3, 1% | |
Erdafitinib58 | Kinase inhibitor/ locally advanced or metastatic urothelial carcinoma | ALT increased: G3–4, 1% AST increased: G3–4, 0% ALP increased: G3–4, 1% | |
Trastuzumab and hyaluronidase-oysk59 | Trastuzumab: HER2/neu receptor antagonist; hyaluronidase: endoglycosidase/breast cancer | ALT increased: G3–4, 1.7% | |
First FDA approved in 2018 | |||
Tumorocentric therapies | |||
Gliteritinib60 | Kinase inhibitor/ relapsed or refractory AML | ALT increased: ≥G3, 12% AST increased: ≥G3, 10% ALP increased: ≥G3, 1% | |
Glasdegib61 | Hedgehog pathway inhibitor/newly diagnosed AML | When used in combination with low-dose cytarabine ALT increased: G3–4, 0% AST increased: G3–4, 1% ALP increased: G3–4, 0% Bilirubin increased: G3–4, 4% | Limitation of use: glasdegib has not been studied in patients with comorbidities of severe renal impairment or moderate to severe hepatic impairment |
Lorlatinib62 | Kinase inhibitor/ALK-positive NSCLC | ALT increased: G3–4, 2.1% AST increased: G3–4, 2.1% ALP increased: G3–4, 1.0% | No dose adjustment for mild hepatic impairment; dose not established for moderate to severe hepatic impairment; potential for hepatotoxicity when used with rifampin |
Talazoparib63 | PARP inhibitor/germ line BRCA-mutated HER2-negative locally advanced or metastatic breast cancer | ALT increased: G3, 1%; G4, 0% AST increased: G3, 2%; G4, 0% ALP increased: G3, 2%; G4, 0% | Talazoparib has not been studied in patients with moderate or severe hepatic impairment Mild hepatic impairment had no effect on PK |
Dacomitinib64 | Kinase inhibitor/metastatic NSCLC with epidermal growth factor receptor mutations | ALT increased: G3–4, 1.4% AST increased: G3–4, 0.5% ALP increased: G3–4, 0.5% Hyperbilirubinemia: G3–4, 0.5% | Mild or moderate hepatic impairment had no effect on PK |
Iobenguane I 13165 | Radioactive therapeutic agent/ iobenguane scan positive, unresectable, locally advanced or metastatic pheochromocytoma or paraganglioma requiring systemic anticancer therapy | Patients with PPGL: ALT increased: G3–4, 2% AST increased: G3–4, 2% ALP increased: G3–4, 5% | |
Encorafenib66 | Kinase inhibitor in combination with binimetinib/ unresectable or metastatic melanoma with BRAF V600E or V600K mutations | In combination with binimetinib: ALT increased: G3–4, 6% AST increased: G3–4, 2.6% ALP increased: G3–4, 0.5% | |
Immuno-Oncology Therapies | |||
Moxetumomab Pasudotox-tdfk67 | CD22-directed cytotoxin indicated for relapsed or refractory hairy cell leukemia | ALT increased: G3, 3.8% AST increased: G3, 1.3% Bilirubin increased; G3, 1.3% | Mild hepatic impairment had no clinically relevant effect on PK PK in patients with moderate to severe hepatic impairment is unknown |
The following groups terms may be used: AST increased, ALT increased, ALP increased, γ-glutamyltransferase increased, hepatic enzyme increased, hepatic function abnormal, hepatoxicity, liver function test increased, and transaminases increased.
ALK indicates anaplastic lymphoma kinase; ALP, alkaline phosphatase; ALT, alanine aminotransferase; AML, acute myeloid leukemia; APPT, activated partial thromboplastin time; AST, aspartate aminotransferase; DLBCL, diffuse large B-cell lymphoma; FDA, US Food and Drug Administration; FL, follicular lymphoma; G, Grade; GI, gastrointestinal; IDH-1, Isocitrate dehydrogenase-1; MCL, mantle cell lymphoma; MM, multiple myeloma; NF1, neurofibromatosis type 1; NSCLC, non–small cell lung cancer; PARP, poly (ADP-ribose) polymerase; PK, pharmacokinetics; PN, plexiform neurofibromas; PPGL, pheochromocytoma and paraganglioma; RR, relapsed or refractory; SCLC, small cell lung cancer.
Cancer immunotherapy (eg, immune checkpoint inhibitors [ICI] and bispecific T-cell engager molecules) is now standard for various solid and hematological cancers.68–70 Immuno-oncology (IO) is expanding rapidly, with multiple ICIs, immune agonists, T-cell engagers, and cellular therapies under investigation. ICI-induced aminotransferase elevations have typically been G2–G371–74; however, deaths due to hepatic failure have been rarely reported.71,75 The frequency of TAEATs for IO drugs approved since 2018 is summarized in Table 2, and those without the mention of hepatic abnormalities are summarized in Table 3.
TABLE 3.
Drug (Ref) | Drug Class/Therapeutic Use |
---|---|
Tumorocentric drugs | |
Enfortumab vedotin-ejfv76 | Nectin-4–directed antibody-drug conjugate/urothelial cancer |
Apalutamide77 | Androgen receptor inhibitor/prostate cancer |
Selinexor78 | Nuclear export inhibitor/RRMM |
Immuno-oncology drugs | |
Daratumumab + hyaluronidase79 | CD38-directed cytolytic antibody + endoglycosidase/MM |
Isatuximab-irfc80 | CD38-directed cytolytic antibody/RRMM |
Mogamulizumab-kpkc81 | CCR4-directed monoclonal antibody/relapsed or refractory mycosis fungoides or Sézary syndrome after ≥1 prior systemic therapy |
MM indicates multiple myeloma; RR, relapsed or refractory.
Dose-Limiting Toxicities and TAEAT
The determination of dose-limiting toxicities (DLTs) is crucial in establishing the maximum tolerated dose and recommended dose for phases 2 and 3.82 Current FDA guidance recommends consideration be given to discontinuing an investigational drug in an asymptomatic patient if ALT or AST levels are >8×ULN or >5×ULN for >2 weeks.18 In patients with any symptoms of hepatitis or with total bilirubin levels >2×ULN or an INR >1.5, drug discontinuation is recommended when AST or ALT levels are >3×ULN.18 Consensus guidelines have been developed for assessing and managing suspected symptomatic DILI in clinical trials in patients with underlying liver disease.83,84 In otherwise asymptomatic patients with ALT elevations at baseline, ALT elevations ≥5×baseline (or absolute values ≥300 U/L) are the current threshold for interrupting treatment. However, current FDA guidance does not specifically address the occurrence of TAEAT, which takes on greater importance when evaluating oncotherapeutic agents for life-threatening malignant diseases.
TAEAT does not appear to preclude further clinical development, with several approved clinical trials defining G3 aminotransferase levels as DLTs only when levels remain elevated for ≥7 days (eg, ClinicalTrials.gov: NCT03439280) or are associated with symptomatic disease (eg, ClinicalTrials.gov: NCT03918278). Interestingly, matching a current DLT definition, a median TAEAT duration of 7 days has been demonstrated in a real-world study in cancer patients treated with ICI, although the maximal duration of TAEAT was 128 days.85 On the basis of this and given risk:benefit assessment in cancer patients, a question is raised as to whether the further extension of acceptable TAEAT duration during clinical studies in patients with aggressive types of cancer (eg, acute myeloid leukemia; AML) is feasible to ensure potentially life-preserving treatment is not being unnecessarily withheld. Illustratively, the results from a prospective, phase 1 study of clofarabine with 2 Gy total body irradiation indicated approximately one third of patients with AML or acute lymphoblastic leukemia experienced ≥G3 ALT and AST elevations without any manifestations of hepatotoxicity.86
COMPLICATING FACTORS IN TAEAT ASSESSMENT IN ONCOLOGY
Abnormal Baseline Liver Chemistries
In oncology trials, abnormal baseline liver chemistries can be affected by various factors, including prior anticancer therapies, potentially hepatotoxic concomitant medications, alcohol use, liver metastases, and preexisting chronic liver disease10,18; nonetheless, patients may remain asymptomatic.87 Because pre-existing chronic liver disease (eg, nonalcoholic steatohepatitis and viral hepatitis) might be responsible for baseline aminotransferase elevations, the pretreatment screening of liver chemistries is usually undertaken.84,88 Although patients with G1 aminotransferase elevations are often enrolled in clinical trials, careful assessment of further on-therapy elevations helps clinicians make informed decisions regarding the management of potential hepatotoxicity.74 Negative tests for hepatitis B virus and hepatitis C virus are usually a part of inclusion criteria in most clinical trials, and patients with affected liver functions are often included in additional postregistrational clinical studies. As outlined in consensus guidelines for clinical trials in patients with underlying liver disease, elevations in aminotransferases based on baseline values are likely to be more meaningful than ULN comparisons,74,83,84 as reflected in Common Terminology Criteria for Adverse Events version 5.11 During such studies and in clinical practice, it is advisable to refer patients with elevated aminotransferases for a hepatology consult for intensive follow-up monitoring to enable early initiation or resumption of potentially life-preserving cancer therapy.
In addition, various oncotherapeutics used as standard of care treatment can potentially lead to elevated aminotransferases, including 6-mercaptopurine treatment for solid tumors,89 doxorubicin for ALL,90 mitoxantrone for relapsed or primary refractory ALL,91 and cisplatin for ovarian cancer.92 Thus, control arms in phase 3 trials may be associated with reversible elevation of aminotransferases consistent with TAEAT (eg, tyrosine kinase inhibitors).18,93,94 Furthermore, the lack of a control group in most phase 1 oncology trials makes the assessment of elevated liver enzymes challenging in patients with underlying factors associated with aminotransferase elevations. Thus, the causality assessment by independent DILI experts is considered the current gold standard95 and essential to assess relatedness in TAEAT. Tools such as the objective scoring system (used in the Roussel Uclaf Causality Assessment Method) can be useful but require a certain degree of expertise and are often combined with expert opinion, as used by the US DILI Network.96,97
Concomitant Medications
The use of concomitant medications (eg, antibiotics/antimycotics) may complicate the interpretation of abnormal liver chemistries and the relationship between TAEAT and the oncotherapeutic agent.98–100 Polypharmacy is prevalent among patients with cancer.101 It is estimated that approximately one third of the US elderly population (2005 to 2006) was prescribed ≥5 concurrent medications.102 Medications such as statins, antiepileptics (phenytoin, carbamazepine, and valproic acid), antifungals (ketoconazole and itraconazole), antituberculosis drugs (rifampin and isoniazid), cotrimoxazole, and allopurinol may all potentially elevate aminotransferases.1,3 Azole antifungals, in particular, are frequently used for prolonged periods in patients with hematologic malignancies, and have been implicated in idiosyncratic DILI, with nearly all azoles associated with minor changes in liver chemistries.103 The LiverTox Bookshelf and other resources provide an up-to-date summary of drugs implicated in DILI,2,3 and can aid in the differential diagnosis of elevated liver enzymes in patients being treated for malignancies.
Metastases
The effect of hepatic and bone metastases on liver enzymes is variable and confounded by relatively limited and often contrasting findings in the literature. Although alkaline phosphatase may be elevated with space-occupying lesions (eg, liver metastases) or due to extrahepatic biliary obstruction from enlarged lymphadenopathy in the area of the porta hepatis (as in breast cancer),104 firm incidence data are lacking. Aminotransferase elevations may reflect the infiltration of liver diseases, such as leukemia or lymphoma,105,106 or liver tests may remain normal. For example, despite having no obvious liver involvement in AML, autopsy reports indicated hepatic infiltration in >75% of patients.107 In addition, higher rates of elevated aminotransferases (>5×ULN) were reported in patients treated with onapristone with bone (2.4% to 4.8%) or liver (4.0%to 12.0%) metastases compared with those without these metastases (0.0% to 4.3% and 0.0% to 1.6%, respectively).108 Similarly, aminotransferases were significantly elevated in patients with solid tumors and liver metastasis versus those without metastases.109–111 In contrast, a pooled analysis of 31 phase 2 and 3 oncology trials found that the incidence of ALT and AST elevations was generally similar in patients with or without liver metastases.112
REAL-WORLD EVALUATION AND MANAGEMENT OF TAEAT IN ONCOLOGY
Limited information is available on how practicing oncologists manage TAEAT or what criteria are used to predict whether the elevated aminotransferases will progress to more serious DILI. A recent real-world US evaluation of elevated aminotransferases associated with IO therapies found that isolated ALT and AST elevations of ≥G3 were relatively transient (up to 128 d with median duration ~7 d), with only 5.3% subsequently progressing to elevated bilirubin levels.85 In this study, oncologists discontinued ICI therapy in 8% of cases, with 92% of patients proceeding with their anticancer treatment. In 37% of cases, ≥G3 aminotransferase elevations were managed with corticosteroids without interruption of ICI therapy, illustrating decision-making based on risk:benefit assessment. Additional real-world studies are needed to assess current trends in TAEAT management in cancer patients treated with non-IO drugs including drug interruptions/discontinuations, use of corticosteroids, and frequency of liver function assessment. An analysis of 1670 patients in 85 phase 1 oncology studies found similar rates of DILI for patients in immune-based versus targeted therapy trials (5.0% vs. 4.9%); DILI resolved in 96% of patients, with no reports of drug-related liver failure,113 consistent with TAEAT representing drug adaptation. Nevertheless, additional real-world data and prospective clinical trials in specific oncology populations are needed to identify factors that predict which patients with elevated aminotransferases are likely to progress to more serious liver injury. Additional work is underway to develop best practice guidelines for the assessment of liver chemistries in oncology trials. In particular, consensus is needed regarding the continuation of an oncotherapeutic agent in patients experiencing TAEAT based on individual risk:benefit assessment.
POSSIBLE MECHANISMS OF TAEAT
It is commonly accepted that cell damage with plasma membrane disruption followed by release of cellular contents into the plasma is principally responsible for aminotransferase elevations present in symptomatic DILI.114 However, the lack of histologic findings (ie, hepatic necrosis) in liver biopsies from some patients with isolated ≥G3 aminotransferase elevations calls into question the potential alternative mechanisms involved in TAEAT.115 The release of hepatoprotective cytokines is postulated as one of the main reasons why mild ALT elevations fail to progress in patients in whom drug adaptation is seen, such as with statins.6,116 Dampening the innate immune response to liver injury or other cellular mechanisms that prevent liver injury from crossing the threshold to irreversibility are suggested as the main reasons that most drugs fail to cause serious liver injury.117 Several of these potential mechanisms are reviewed below and are likely to apply to all-grade aminotransferase elevations. Once established in preclinical settings, human studies might be warranted to assess not only the mechanisms driving TAEAT, but most importantly, optimal mitigation approaches in clinic.
Role of Liver Cells
Approximately 80% of liver cells are parenchymal (hepatocytes), with the nonparenchymal cells comprising endothelial cells (8%), stellate cells (4%), Kupffer cells (4%), and intrahepatic lymphocytes (4%).118,119 Infiltrating T cells, natural killer (NK)/NK T cells, Kupffer cells, and infiltrating tumor cells may contribute to aminotransferase elevations.120 Various studies have shown that activated CD8+ T cells might cause inflammation in the liver leading to elevations in aminotransferases.121–123 Hepatic NK cells can also respond to a local cytokine milieu and contribute to liver injury by a nonantigen-specific TNF-related apoptosis-inducing ligand-mediated pathway.124–126 Activated Kupffer cells are a major source of inflammatory mediators such as superoxide, nitric oxide, eicosanoids, cytokines, lysosomal, and other proteolytic enzymes that lead to altered hepatic homeostasis,127 potentially leading to elevated aminotransferases.
Membrane Blebbing
Large plasma membrane blebs, clear and round cytoplasmic protrusions, may form as a physiological response to activating stimuli and are not the sole indicators of extreme cell stress or initiation of death pathways.128,129 Their formation may be dependent on external calcium, indicating that signaling events initiating the formation may be downstream of the high-affinity IgE receptor/inositol trisphosphate/calcium release–activated channels (FcεRI/IP3/CRAC) pathway for store-operated calcium entry.129 Alterations in plasma membrane caused by the absence of oxygen have been reported.130 Under hypoxic conditions, blebs ruptured and released their contents, including aminotransferases, into the circulation resulting in elevated aminotransferases without overt hepatocellular damage.130
Hypoxic Hepatitis and Cytokine Release Syndrome
Hypoxic hepatitis, also known as “shock liver,” is characterized by massive, rapid, and transient elevations of aminotransferases (AST often >20–100×ULN) due to an imbalance in hepatic oxygen demand and supply.131 Hypotension is one of the principal causes of hypoxic hepatitis132 and is also associated with cytokine release syndrome (CRS).133 Along with oncotherapeutics such as rituximab,134 obinutuzumab,135 oxaliplatin,136 and lenalidomide,137 CRS has been reported with bispecific T-cell engager molecules, such as blinatumomab,138 and chimeric antigen receptor T-cell therapies.139 Cytokine levels have been shown to be modulated by hypoxic hepatitis140; thus, CRS-inducing drugs may be associated with elevations in aminotransferases with or without typical hepatic symptoms, in the context of liver chemistry abnormalities.
Increased Expression of ALT and AST
Induction of expression of the genes encoding ALT and AST is another possible mechanism leading to aminotransferase elevations without apparent injury. Fenofibrate has been shown to increase the expression of genes encoding ALT and AST in human hepatoma cell line HepG2 and by binding of peroxisome proliferator-activated receptor α (PPARα) to the peroxisome proliferator-activated receptor response element in the proximal ALT1 promoter resulting in elevated ALT levels.141,142 Dexamethasone, used for CRS prophylaxis and treatment,143 has been shown to elevate the levels of aminotransferases via increased transcription of human and rat ALT1 reporter genes and increased the transcription of GPT1/GOT1 genes.142,144,145 Increases in ALT and AST (usually G1–G2, but occasionally G3) induced by bardoxlone methyl are thought to be related to the pharmacologic induction of aminotransferases via nuclear factor erythroid 2–related factor 2 activation rather than any intrinsic form of hepatotoxicity.146
Indirect Effect Via Local Microenvironment
Agents that affect the liver through indirect means may also cause elevated aminotransferase levels. Various oncotherapeutics possess a half-life-extending crystallizable fragment (Fc) domain, which could enhance the immune responses, including antibody-dependent cytotoxicity, complement-dependent cytotoxicity, and antibody-dependent cell-mediated phagocytosis.147,148 Kupffer cells and NK cells express Fc receptors on their surfaces,149,150 and their stimulation may result in the production of proinflammatory cytokines such as TNF-α, IL-1β, and IL-6.149–151 Hence, Fc receptor–mediated cell activation by the Fc fragment of various molecules might lead to TAEAT as a consequence of local inflammation.
Macroenzymes
Type 1 macroenzymes include high molecular mass complexes of AST and ALT with immunoglobulins, which aid in protecting aminotransferases from degradation slowing their clearance and leading to increased serum levels.152,153 Although the role of immunoglobulins, mostly IgG and IgA, in the formation of type 1 complexes is well established, it remains unknown whether protein-based drugs can contribute to AST and ALT macroenzyme formation. Nevertheless, prolonged elevations of aminotransferases due to macroenzymes may be interpreted as TAEAT.153,154
CLINICAL SIGNIFICANCE OF TAEAT AND GAPS IN KNOWLEDGE
Although the clinical significance of symptomatic DILI cases or cases with multiple laboratory abnormalities is well established, there is no consensus on how to interpret and manage TAEAT. Several of the gaps in our current knowledge regarding various aspects of TAEAT, which may benefit from further research, are summarized below.
Should Risk:Benefit Assessment be Used to Determine TAEAT Management?
The determination of when to continue treatment and risk:benefit considerations for various types or stages of cancer are areas requiring further research. The interpretation and clinical significance of a single Hy law case or TAEAT in oncology trials can be especially challenging. Modification of Hy law criteria using fold elevations in liver chemistries in patients with baseline abnormalities155 have been proposed to improve the assessment of possible TAEAT in oncology trials. Although oncotherapeutic agents causing liver enzyme elevations may still gain FDA approval, specific guidelines are needed to differentiate between management approaches based on risk:benefit assessment for each patient, to identify when to monitor liver chemistries and continue treatment, or when to modify, withhold, or discontinue treatment.
Can Hepatic Histology Help in Understanding the Biology of TAEAT?
Aminotransferases are not only released into plasma after hepatocyte death but also because of extrahepatic causes, such as hemolysis and muscle injury.114,156 Across a range of indications and patient populations, there are reports of TAEAT without associated significant liver injury, for which extrahepatic causes may be responsible.86,115,157,158 Aminotransferases are also released into the circulation without cell death. A better understanding of the pathophysiology of TAEAT may help identify the optimal approach to clinical management. In many of the cases described, patients with TAEAT continued treatment and aminotransferase levels either plateaued or resolved; features consistent with drug adaptation. In patients in whom aminotransferases remain elevated and noninvasive investigations show no obvious alternative cause, liver biopsy may be recommended.159 Although hepatic histology cannot completely establish causality to a specific drug,115 it is useful in helping to differentiate TAEAT from other causes, such as autoimmune hepatitis.40,160 Defining the histologic pattern of TAEAT injury can provide an indication of severity, enabling the clinician to balance risk:benefit of continuing the suspected causative therapy.159
This is specifically important because minor nonspecific changes can be observed in the biopsy results of patients with TAEAT.161 From available biopsy data, ≥G3 elevations do not routinely imply liver necrosis,115 indicating that with some therapies, large increases in aminotransferases may occur without significant hepatocyte death. However, aminotransferase elevations, with or without hyperbilirubinemia, have also been associated with histologic liver injury (particularly with ICI therapy),73,75 and liver cell necrosis typically occurred with aminotransferase elevations associated with hyperbilirubinemia.73 Hepatic necrosis or apoptosis distinguishes this type of liver injury from TAEAT. Nevertheless, liver biopsy data are limited, and in some malignancies (eg, those associated with thrombocytopenia), histology may be difficult to obtain. Although ultrasound, magnetic resonance imaging (MRI), and computed tomography are noninvasive alternatives, they provide limited information compared with histologic evaluation. Although several exploratory biomarkers for DILI have been proposed,162 further validation is required in tandem with biopsies undertaken in the context of DILI to support such validation in different clinical scenarios. Therefore, a need for novel technologies and biomarkers is evident to assess liver injury at a cellular and molecular levels.
Should TAEAT Management Include Drug Interruption and Rechallenge?
For some oncotherapeutic agents, treatment interruption may potentially compromise efficacy outcomes. This raises the question of whether the treatment interruption can be avoided in patients with TAEAT with steroid treatment and careful liver chemistry monitoring to ensure that no subsequent hyperbilirubinemia or clinical symptoms of hepatic injury develop.85 Desensitization rechallenge is controversial, but may be considered when benefits of therapy outweigh risks.163 Indeed, for some oncotherapeutics (eg, ICI for solid cancers), rechallenge after TAEAT is increasingly being accepted.40,71,164–170 The decision to resume treatment after the detection of hepatitis/hepatotoxicity is based on individualized risk:benefit assessments,40,167–170 and these considerations should likely apply to rechallenge after the confirmation of elevated aminotransferase levels. Although current recommendations for rechallenge are outlined in the approved prescribing information (Table 1), further discussion is needed with a goal to generate patient-tailored decision-making algorithms to ensure optimal patient outcomes.
CONCLUSIONS
In both clinical trials and routine clinical practice, physicians generally focus on monitoring higher grades (≥G3) of aminotransferase elevations. Published reports indicate that some patients are asymptomatic without any overt liver injury despite having ≥G3 aminotransferase elevations, and these elevations do not progress and may resolve while the drug is continued, a phenomenon similar to drug adaptation associated with nononcology agents. As such, prospective studies of rechallenge in patients with no other signs of impaired hepatic function could be considered. The recognition and understanding of TAEAT within oncology is further complicated by the coexistence of chronic liver disease, liver/bone metastases, prior treatments, and concomitant medications. Despite these challenges, the occurrence of TAEAT with oncotherapeutics may be more common than previously appreciated.
A number of mechanisms is likely involved in these transient, asymptomatic elevations of aminotransferase levels. However, it is not clear whether these mechanisms are drug specific, indication specific, or patient specific. Additional studies aimed at elucidating these mechanisms at a cellular and molecular level are needed to better determine the cause of TAEAT, for both oncotherapeutics and other drugs, including in specific populations, such as those with liver metastases.
In conclusion, TAEAT with ≥G3 elevation of aminotransferases is associated with many oncotherapeutic agents. Although usually reported as an adverse event, its clinical implications remain incompletely understood, as progressive liver injury may not develop. Additional research and clinical studies, including real-world data, are needed to gain a better understanding of TAEAT pathophysiology and management. This increased understanding will enable the development of consensus guidelines for the use of drugs associated with TAEAT in patients with cancer, including in those who have exhausted all other treatment options.
ACKNOWLEDGMENTS
The authors thank Indira Venkatasubramanian, PhD (Amgen Inc.), Advait Joshi, PhD (Cactus Communications), Erin P. O’Keefe, Lee Hohaia, and Rick Davis (ICON plc, Blue Bell, PA) for the medical writing and editing support, which was funded by Amgen Inc.
Footnotes
J.H.L. and M.S. are co-corresponding authors.
This work was supported by Amgen Inc.
J.H.L.: roles/writing—original draft, writing—review and editing. S.K.K.: conceptualization, data curation, funding acquisition, supervision, roles/writing—original draft, writing—review and editing. C.D.B.: writing—review and editing. A.H.W.: writing—review and editing. M.S.: conceptualization, data curation, writing—review and editing.
J.H.L. has no known competing financial interests or personal relationships, that could have appeared to influence the work reported in this paper. S.K.K. and C.D.B. are employed by and own stock in Amgen Inc. A.H.W. has served on advisory boards for Novartis, Janssen, Amgen, Roche, Pfizer, AbbVie, Servier, Celgene-BMS, Macrogenics, Agios, and Gilead; received research funding to his institution from Novartis, AbbVie, Servier, Celgene-BMS, Astra Zeneca, and Amgen; served on the speakers’ bureau for AbbVie, Novartis, and Celgene. M.S. served in a consulting or advisory role for Amgen, Celgene, Gilead Sciences, Janssen, Novartis, Pfizer, and Seattle Genetics; served on the speakers’ bureau for Amgen, Celgene, Gilead Sciences, Janssen, Novartis, and Pfizer; received travel, accommodations, and expenses from Amgen, Celgene, and Gilead Sciences; received research funding from Amgen, Celgene, Gilead Sciences, Miltenyi Biotec, MorphoSys, Novartis, and Seattle Genetics. The other authors declare no conflicts of interest.
Contributor Information
James H. Lewis, Email: lewisjh@gunet.georgetown.edu.
Sophia K. Khaldoyanidi, Email: skhaldoy@amgen.com.
Carolyn D. Britten, Email: cbritten@amgen.com.
Andrew H. Wei, Email: andrew.wei@monash.edu.
Marion Subklewe, Email: marion.subklewe@med.uni-muenchen.de.
REFERENCES
- 1.Zimmerman HJ. Hepatotoxicity: The Adverse Effects of Drugs and Other Chemicals on the Liver. Philadelphia, PA: Lippincott Williams & Wilkins; 1999. [Google Scholar]
- 2.Björnsson ES. Drug-induced liver injury: an overview over the most critical compounds. Arch Toxicol. 2015;89:327–334. [DOI] [PubMed] [Google Scholar]
- 3.National Institute of Diabetes and Digestive and Kidney Diseases. LiverTox: clinical and research information on drug-induced liver injury. Available at: https://www.ncbi.nlm.nih.gov/books/NBK548776/. Accessed January 19, 2021. [PubMed]
- 4.Fontana RJ, Hayashi PH, Gu J, et al. Idiosyncratic drug-induced liver injury is associated with substantial morbidity and mortality within 6 months from onset. Gastroenterology. 2014;147:96–108.e4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Medina-Caliz I, Robles-Diaz M, Garcia-Muñoz B, et al. Definition and risk factors for chronicity following acute idiosyncratic drug-induced liver injury. J Hepatol. 2016;65:532–542. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Lewis JH. Drug-induced liver injury throughout the drug development life cycle: where we have been, where we are now, and where we are headed. Perspectives of a clinical hepatologist. Pharm Med. 2013;27:165–191. [Google Scholar]
- 7.Aithal GP, Watkins PB, Andrade RJ, et al. Case definition and phenotype standardization in drug-induced liver injury. Clin Pharmacol Ther. 2011;89:806–815. [DOI] [PubMed] [Google Scholar]
- 8.Kullak-Ublick GA, Andrade RJ, Merz M, et al. Drug-induced liver injury: recent advances in diagnosis and risk assessment. Gut. 2017;66:1154–1164. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Oh RC, Hustead TR, Ali SM, et al. Mildly elevated liver transaminase levels: causes and evaluation. Am Fam Physician. 2017;96:709–715. [PubMed] [Google Scholar]
- 10.Kullak-Ublick GA, Merz M, Griffel L, et al. Liver safety assessment in special populations (hepatitis B, C, and oncology trials). Drug Saf. 2014;37:S57–S62. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.US Department of Health and Human Services. Common Terminology Criteria for Adverse Events (CTCAE) Version 5.0; 2017.
- 12.Hoofnagle JH, Bjornsson ES. Drug-induced liver injury - types and phenotypes. N Engl J Med. 2019;381:264–273. [DOI] [PubMed] [Google Scholar]
- 13.Watkins PB, Zimmerman HJ, Knapp MJ, et al. Hepatotoxic effects of tacrine administration in patients with Alzheimer’s disease. JAMA. 1994;271:992–998. [PubMed] [Google Scholar]
- 14.Marx G, Taylor J, Goldstein D. Outpatient treatment with subcutaneous interleukin-2, interferon alpha and fluorouracil in patients with metastatic renal cancer: an Australian experience. Intern Med J. 2005;35:34–38. [DOI] [PubMed] [Google Scholar]
- 15.King AC, Pappacena JJ, Tallman MS, et al. Blinatumomab administered concurrently with oral tyrosine kinase inhibitor therapy is a well-tolerated consolidation strategy and eradicates measurable residual disease in adults with Philadelphia chromosome positive acute lymphoblastic leukemia. Leuk Res. 2019;79:27–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Brahmer JR, Lacchetti C, Schneider BJ, et al. Management of immune-related adverse events in patients treated with immune checkpoint inhibitor therapy: American Society of Clinical Oncology Clinical Practice Guideline. J Clin Oncol. 2018;36:1714–1768. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Lee WM. Acute liver failure. Semin Respir Crit Care Med. 2012;33:36–45. [DOI] [PubMed] [Google Scholar]
- 18.US Food and Drug Administration. Guidance for Industry Drug-induced Liver Injury: Premarketing Clinical Evaluation. Silver Spring, MD: US Department of Health and Human Services; 2009. [Google Scholar]
- 19.Temple R. Hy’s law: predicting serious hepatotoxicity. Pharmacoepidemiol Drug Saf. 2006;15:241–243. [DOI] [PubMed] [Google Scholar]
- 20.Kwo PY, Cohen SM, Lim JK. ACG clinical guideline: evaluation of abnormal liver chemistries. Am J Gastroenterol. 2017;112:18–35. [DOI] [PubMed] [Google Scholar]
- 21.Chung JY, Longo DM, Watkins PB. A rapid method to estimate hepatocyte loss due to drug-induced liver injury. Clin Pharmacol Ther. 2019;105:746–753. [DOI] [PubMed] [Google Scholar]
- 22.Stephens C, Robles-Diaz M, Medina-Caliz I, et al. Comprehensive analysis and insights gained from long-term experience of the Spanish DILI Registry. J Hepatol. 2021;75:86–97. [DOI] [PubMed] [Google Scholar]
- 23.Lewis JH. ‘Hy’s law,’ the ‘Rezulin Rule,’ and other predictors of severe drug-induced hepatotoxicity: puting risk-benefit into perspective. Pharmacoepidemiol Drug Saf. 2006;15:221–229. [DOI] [PubMed] [Google Scholar]
- 24.Retevmo (selpercatinib). Full Prescribing Information. Indianapolis. IN: Eli Lilly; 2020. [Google Scholar]
- 25.Tabrecta (capmatinib). Full Prescribing Information. East Hanover, NJ: Novartis; 2020. [Google Scholar]
- 26.Tukysa (tucatinib). Full Prescribing Information. Bothell, WA: Seattle Genetics; 2020. [Google Scholar]
- 27.Rozlytrek (entrectinib). Full Prescribing Information. South San Francisco, CA: Genentech; 2019. [Google Scholar]
- 28.Turalio (pexidartinib). Full Prescribing Information. Basking Ridge, NJ: Daiichi Sankyo; 2019. [Google Scholar]
- 29.Polivy (polatuzumab vedotin-piiq). Full Prescribing Information. South San Francisco, CA: Genentech; 2019. [Google Scholar]
- 30.Elzonris (tagraxofusp-erzs). Full Prescribing Information. New York, NY: Stemline Therapeutics; 2018. [Google Scholar]
- 31.Asparlas (calaspargase pegol-mknl). Full Prescribing Information. Boston, MA: Servier Pharmaceuticals; 2018. [Google Scholar]
- 32.Vitrakvi (larotrectinib). Full Prescribing Information. Stamford, CT: Loxo Oncology; 2018. [Google Scholar]
- 33.Copiktra (duvelisib). Full Prescribing Information. Needham, MA: Verastem; 2018. [Google Scholar]
- 34.Mektovi (binimetinib). Full Prescribing Information. Boulder, CO: Array BioPharma; 2018. [Google Scholar]
- 35.Sackstein PE, O’Neil DS, Neugut AI, et al. Epidemiologic trends in neuroendocrine tumors: an examination of incidence rates and survival of specific patient subgroups over the past 20 years. Semin Oncol. 2018;45:249–258. [DOI] [PubMed] [Google Scholar]
- 36.Libtayo (cemiplimab-rwlc). Full Prescribing Information. Tarrytown, NY: Regeneron Pharmaceuticals; 2018. [Google Scholar]
- 37.Clinton JW, Kiparizoska S, Aggarwal S, et al. Drug-induced liver injury: highlights and controversies in the recent literature. Drug Safety. 2021;44:1125–1149. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Wang E, Song F, Paulus JK, et al. Qualitative and quantitative variations in liver function thresholds among clinical trials in cancer: a need for harmonization. Cancer Chemother Pharmacol. 2019;84:213–216. [DOI] [PubMed] [Google Scholar]
- 39.Desjardin M, Bonhomme B, Le Bail B, et al. Hepatotoxicities induced by neoadjuvant chemotherapy in colorectal cancer liver metastases: distinguishing the true from the false. Clin Med Insights Oncol. 2019;13:1179554918825450. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.De Martin E, Michot JM, Rosmorduc O, et al. Liver toxicity as a limiting factor to the increasing use of immune checkpoint inhibitors. JHEP Rep. 2020;2:100170. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Blenrep (belantamab mafodotin-blmf). Full Prescribing Information. Research Triangle Park, NC: GlaxoSmithKline; 2020. [Google Scholar]
- 42.INQOVI (decitabine and cedazuridine). Full Prescribing Information. Japan: Otsuka Pharmaceutical Co., Ltd.; 2020. [Google Scholar]
- 43.Phesgo (pertuzumab, trastuzumab, and hyaluronidase-zzxf). Full Prescribing Information. South San Francisco, CA: Genentech; 2020. [Google Scholar]
- 44.Tazverik (tazemetostat). Full Prescribing Information. Cambridge, MA: Epizyme; 2020. [Google Scholar]
- 45.Zepzelca (lurbinectedin). Full Prescribing Information. Palo Alto, CA: Jazz Pharmaceuticals; 2020. [Google Scholar]
- 46. Qinlock (ripretinib). Full Prescribing Information. Waltham, MA: Deciphera Pharmaceuticals; 2020. [Google Scholar]
- 47.Trodelvy (sacituzumab govitecan-hziy). Full Prescribing Information. Morris Plains, NJ: Immunomedics; 2020. [Google Scholar]
- 48.Pemazyre (pemigatinib). Full Prescribing Information. Wilmington, DE: Incyte; 2020. [Google Scholar]
- 49.Koselugo (selumetinib). Full Prescribing Information. Wilmington, DE: AstraZeneca; 2020. [Google Scholar]
- 50.Ayvakit (avapritinib). Full Prescribing Information. Cambridge, MA: Blueprint Medicines; 2020. [Google Scholar]
- 51.Monjuvi (tafasitamab-cxix). Full Prescribing Information. Boston, MA: Morphosys US; 2020. [Google Scholar]
- 52.Tecartus (brexucabtagene autoleucel). Full Prescribing Information. Santa Monica, CA: Kite Pharma; 2020. [Google Scholar]
- 53.Enhertu (fam-trastuzumab deruxtecan-nxki). Full Prescribing Information. Basking Ridge, NJ: Daiichi Sankyo; 2019. [Google Scholar]
- 54.Brukinsa (zanubrutinib). Full Prescribing Information. San Mateo, CA: BeiGene USA; 2019. [Google Scholar]
- 55.Nubeqa (darolutamide). Full Prescribing Information. Whippany, NJ: Bayer HealthCare Pharmaceuticals; 2019. [Google Scholar]
- 56.Piqray (alpelisib). Full Prescribing Information. East Hanover, NJ: Novartis Pharmaceuticals; 2019. [Google Scholar]
- 57.Tibsovo (ivosidenib tablets). Full Prescribing Information. Cambridge, MA: Agios Pharmaceuticals; 2018. [Google Scholar]
- 58.Balversa (erdafitinib). Full Prescribing Information. Horsham, PA: Janssen Products; 2020. [Google Scholar]
- 59.Herceptin Hylecta (trastuzumab and hyaluronidase-oysk). Full Prescribing Information. South San Francisco, CA: Genentech; 2019. [Google Scholar]
- 60.Xospata (gilteritinib). Full Prescribing Information. Northbrook, IL: Astellas Pharma; 2018. [Google Scholar]
- 61.Daurismo (glasdegib). Full Prescribing Information. New York, NY: Pfizer Labs; 2018. [Google Scholar]
- 62.Lorbrena (lorlatinib). Full Prescribing Information. New York, NY: Pfizer Labs; 2018. [Google Scholar]
- 63.Talzenna (talazoparib). Full Prescribing Information. New York, NY: Pfizer Labs; 2018. [Google Scholar]
- 64.Vizimpro (dacomitinib). Full Prescribing Information. New York, NY: Pfizer Labs; 2018. [Google Scholar]
- 65.Azedra (iobenguane I 131). Full Prescribing Information. New York, NY: Progenics Pharmaceuticals; 2018. [Google Scholar]
- 66.Braftovi (encorafenib). Full Prescribing Information. Boulder, CO: Array BioPharma; 2018. [Google Scholar]
- 67.Lumoxiti (moxetumomab pasudotox-tdfk). Full Prescribing Information. Wilmington, DE: AstraZeneca; 2018. [Google Scholar]
- 68.Marshall HT, Djamgoz MBA. Immuno-oncology: emerging targets and combination therapies. Front Oncol. 2018;8:315. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Einsele H, Borghaei H, Orlowski RZ, et al. The BiTE (bispecific T-cell engager) platform: development and future potential of a targeted immuno-oncology therapy across tumor types. Cancer. 2020;126:3192–3201. [DOI] [PubMed] [Google Scholar]
- 70.Esfahani K, Roudaia L, Buhlaiga N, et al. A review of cancer immunotherapy: from the past, to the present, to the future. Curr Oncol. 2020;27:S87–S97. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Jennings JJ, Mandaliya R, Nakshabandi A, et al. Hepatotoxicity induced by immune checkpoint inhibitors: a comprehensive review including current and alternative management strategies. Expert Opin Drug Metab Toxicol. 2019;15:231–244. [DOI] [PubMed] [Google Scholar]
- 72.Johncilla M, Misdraji J, Pratt DS, et al. Ipilimumab-associated hepatitis: clinicopathologic characterization in a series of 11 cases. Am J Surg Pathol. 2015;39:1075–1084. [DOI] [PubMed] [Google Scholar]
- 73.Kim KW, Ramaiya NH, Krajewski KM, et al. Ipilimumab associated hepatitis: imaging and clinicopathologic findings. Invest New Drugs. 2013;31:1071–1077. [DOI] [PubMed] [Google Scholar]
- 74.Regev A, Avigan MI, 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. J Autoimmun. 2020;114:102514. [DOI] [PubMed] [Google Scholar]
- 75.O’Day SJ, Maio M, Chiarion-Sileni V, et al. Efficacy and safety of ipilimumab monotherapy in patients with pretreated advanced melanoma: a multicenter single-arm phase II study. Ann Oncol. 2010;21:1712–1717. [DOI] [PubMed] [Google Scholar]
- 76.Padcev (enfortumab vedotin-ejfv). Full Prescribing Information. Bothell, WA: Seattle Genetics; 2019. [Google Scholar]
- 77.Erleada (apalutamide). Full Prescribing Information. Horsham, PA: Janssen Products; 2018. [Google Scholar]
- 78.Xpovio (selinexor). Full Prescribing Information. Newton, MA: Karyopharm Therapeutics; 2019. [Google Scholar]
- 79.Darzalex (daratumumab). Full Prescribing Information. Horsham, PA: Janssen Biotech; 2016. [Google Scholar]
- 80.Sarclisa (isatuximab-irfc). Full Prescribing Information. Bridgewater, NJ: Sanofi-Aventis US; 2020. [Google Scholar]
- 81.Poteligeo (mogamulizumab-kpkc). Full Prescribing Information. Bedminster, NJ: Kyowa Kirin; 2018. [Google Scholar]
- 82.LoRusso PM, Boerner SA, Seymour L. An overview of the optimal planning, design, and conduct of phase I studies of new therapeutics. Clin Cancer Res. 2010;16:1710–1718. [DOI] [PubMed] [Google Scholar]
- 83.Regev A, Palmer M, Avigan MI, et al. Consensus guidelines: best practices for detection, assessment and management of suspected acute drug-induced liver injury during clinical trials in patients with nonalcoholic steatohepatitis. Aliment Pharmacol Ther. 2019;49:702–713. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Palmer M, Regev A, Lindor K, et al. Consensus guidelines: best practices for detection, assessment and management of suspected acute drug-induced liver injury occurring during clinical trials in adults with chronic cholestatic liver disease. Aliment Pharmacol Ther. 2020;51:90–109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Kim C, Zhu S, Kouros-Mehr H, et al. Incidence of elevated aminotransferase with or without bilirubin elevation during treatment with immune checkpoint inhibitors: a retrospective study of patients from community oncology clinics in the United States. Cureus. 2022;14:e24053. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Soni S, Abdel-Azim H, McManus M, et al. Phase I study of clofarabine and 2-Gy total body irradiation as a nonmyeloablative preparative regimen for hematopoietic stem cell transplantation in pediatric patients with hematologic malignancies: a therapeutic advances in childhood leukemia consortium study. Biol Blood Marrow Transplant. 2017;23:1134–1141. [DOI] [PubMed] [Google Scholar]
- 87.Mathiesen UL, Franzen LE, Fryden A, et al. The clinical significance of slightly to moderately increased liver transaminase values in asymptomatic patients. Scand J Gastroenterol. 1999;34:85–91. [DOI] [PubMed] [Google Scholar]
- 88.Chalasani N, Regev A. Drug-induced liver injury in patients with preexisting chronic liver disease in drug development: how to identify and manage? Gastroenterology. 2016;151:1046–1051. [DOI] [PubMed] [Google Scholar]
- 89.Adamson PC, Zimm S, Ragab AH, et al. A phase II trial of continuous-infusion 6-mercaptopurine for childhood solid tumors. Cancer Chemother Pharmacol. 1990;26:343–344. [DOI] [PubMed] [Google Scholar]
- 90.Aviles A, Herrera J, Ramos E, et al. Hepatic injury during doxorubicin therapy. Arch Pathol Lab Med. 1984;108:912–913. [PubMed] [Google Scholar]
- 91.Paciucci PA, Sklarin NT. Mitoxantrone and hepatic toxicity. Ann Intern Med. 1986;105:805–806. [DOI] [PubMed] [Google Scholar]
- 92.Pollera CF, Ameglio F, Nardi M, et al. Cisplatin-induced hepatic toxicity. J Clin Oncol. 1987;5:318–319. [DOI] [PubMed] [Google Scholar]
- 93.Le Tourneau C, Lee JJ, Siu LL. Dose escalation methods in phase I cancer clinical trials. J Natl Cancer Inst. 2009;101:708–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Shah RR, Morganroth J, Shah DR. Hepatotoxicity of tyrosine kinase inhibitors: clinical and regulatory perspectives. Drug Saf. 2013;36:491–503. [DOI] [PubMed] [Google Scholar]
- 95.Regev A, Seeff LB, Merz M, et al. Causality assessment for suspected DILI during clinical phases of drug development. Drug Saf. 2014;37:S47–S56. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Danan G, Teschke R. Roussel Uclaf Causality Assessment Method for drug-induced liver injury: present and future. Front Pharmacol. 2019;10:853. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Chalasani N, Bonkovsky HL, Fontana R, et al. Features and outcomes of 899 patients with drug-induced liver injury: the DILIN prospective study. Gastroenterology. 2015;148:1340–1352.e7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Schreve RH, Terpstra OT, Ausema L, et al. Detection of liver metastases. A prospective study comparing liver enzymes, scintigraphy, ultrasonography and computed tomography. Br J Surg. 1984;71:947–949. [DOI] [PubMed] [Google Scholar]
- 99.Bonfanti G, Bombelli L, Bozzetti F, et al. The role of CEA and liver function tests in the detection of hepatic metastases from colo-rectal cancer. HPB Surg. 1990;3:29–36. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Kamath PS. Clinical approach to the patient with abnormal liver test results. Mayo Clin Proc. 1996;71:1089–1094. [DOI] [PubMed] [Google Scholar]
- 101.Barlow A, Prusak ES, Barlow B, et al. Interventions to reduce polypharmacy and optimize medication use in older adults with cancer. J Geriatr Oncol. 2021;12:863–871. [DOI] [PubMed] [Google Scholar]
- 102.Qato DM, Alexander GC, Conti RM, et al. Use of prescription and over-the-counter medications and dietary supplements among older adults in the United States. JAMA. 2008;300:2867–2878. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Benitez LL, Carver PL. Adverse effects associated with long-term administration of azole antifungal agents. Drugs. 2019;79:833–853. [DOI] [PubMed] [Google Scholar]
- 104.Van Laethem JL, De Broux S, Eisendrath P, et al. Clinical impact of biliary drainage and jaundice resolution in patients with obstructive metastases at the hilum. Am J Gastroenterol. 2003;98:1271–1277. [DOI] [PubMed] [Google Scholar]
- 105.Segal I, Rassekh SR, Bond MC, et al. Abnormal liver transaminases and conjugated hyperbilirubinemia at presentation of acute lymphoblastic leukemia. Pediatr Blood Cancer. 2010;55:434–439. [DOI] [PubMed] [Google Scholar]
- 106.Lu TX, Wu S, Cai DY, et al. Prognostic significance of serum aspartic transaminase in diffuse large B-cell lymphoma. BMC Cancer. 2019;19:553. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Singh MM, Pockros PJ. Hematologic and oncologic diseases and the liver. Clin Liver Dis. 2011;15:69–87. [DOI] [PubMed] [Google Scholar]
- 108.Lewis JH, Cottu PH, Lehr M, et al. Onapristone extended release: safety evaluation from phase I-II studies with an emphasis on hepatotoxicity. Drug Saf. 2020;43:1045–1055. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Cao R, Wang LP. Serological diagnosis of liver metastasis in patients with breast cancer. Cancer Biol Med. 2012;9:57–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Wu XZ, Ma F, Wang XL. Serological diagnostic factors for liver metastasis in patients with colorectal cancer. World J Gastroenterol. 2010;16:4084–4088. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Cotogno PM, Ranasinghe LK, Ledet EM, et al. Laboratory-based biomarkers and liver metastases in metastatic castration-resistant prostate cancer. Oncologist. 2018;23:791–797. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Shantakumar S, Landis S, Lawton A, et al. Prevalence and incidence of liver enzyme elevations in a pooled oncology clinical trial cohort. Regul Toxicol Pharmacol. 2016;77:257–262. [DOI] [PubMed] [Google Scholar]
- 113.Mondaca SP, Liu D, Flynn JR, et al. Clinical implications of drug-induced liver injury in early-phase oncology clinical trials. Cancer. 2020;126:4967–4974. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.McGill MR. The past and present of serum aminotransferases and the future of liver injury biomarkers. EXCLI Journal. 2016;15:817–828. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Kebenko M, Goebeler ME, Wolf M, et al. A multicenter phase 1 study of solitomab (MT110, AMG 110), a bispecific EpCAM/CD3 T-cell engager (BiTE(R)) antibody construct, in patients with refractory solid tumors. Oncoimmunology. 2018;7:e1450710. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Dara L, Liu ZX, Kaplowitz N. Mechanisms of adaptation and progression in idiosyncratic drug induced liver injury, clinical implications. Liver Int. 2016;36:158–165. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Jee A, Sernoskie SC, Uetrecht J. Idiosyncratic drug-induced liver injury: mechanistic and clinical challenges. Int J Mol Sci. 2021;22:2954. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Gerber MA, Thung SN. Histology of the liver. Am J Surg Pathol. 1987;11:709–722. [DOI] [PubMed] [Google Scholar]
- 119.Schiff ER, Maddrey WC, Reddy KR. Schiff’s Diseases of the Liver. Chichester, West Sussex, UK: John Wiley & Sons; 2017. [Google Scholar]
- 120.Racanelli V, Rehermann B. The liver as an immunological organ. Hepatology. 2006;43:S54–S62. [DOI] [PubMed] [Google Scholar]
- 121.Park S, Murray D, John B, et al. Biology and significance of T-cell apoptosis in the liver. Immunol Cell Biol. 2002;80:74–83. [DOI] [PubMed] [Google Scholar]
- 122.Russell JQ, Morrissette GJ, Weidner M, et al. Liver damage preferentially results from CD8(+) T cells triggered by high affinity peptide antigens. J Exp Med. 1998;188:1147–1157. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Bowen DG, Warren A, Davis T, et al. Cytokine-dependent bystander hepatitis due to intrahepatic murine CD8 T-cell activation by bone marrow-derived cells. Gastroenterology. 2002;123:1252–1264. [DOI] [PubMed] [Google Scholar]
- 124.Dunn C, Brunetto M, Reynolds G, et al. Cytokines induced during chronic hepatitis B virus infection promote a pathway for NK cell-mediated liver damage. J Exp Med. 2007;204:667–680. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Zhang Z, Zhang S, Zou Z, et al. Hypercytolytic activity of hepatic natural killer cells correlates with liver injury in chronic hepatitis B patients. Hepatology. 2011;53:73–85. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Oliviero B, Varchetta S, Paudice E, et al. Natural killer cell functional dichotomy in chronic hepatitis B and chronic hepatitis C virus infections. Gastroenterology. 2009;137:1151–1160. [DOI] [PubMed] [Google Scholar]
- 127.Winwood PJ, Arthur MJ. Kupffer cells: their activation and role in animal models of liver injury and human liver disease. Semin Liver Dis. 1993;13:50–59. [DOI] [PubMed] [Google Scholar]
- 128.Hogue MJ. The effect of hypotonic and hypertonic solutions on fibroblasts of the embryonic chick heart in vitro. J Exp Med. 1919;30:617–648. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Jansen C, Tobita C, Umemoto EU, et al. Calcium-dependent, non-apoptotic, large plasma membrane bleb formation in physiologically stimulated mast cells and basophils. J Extracell Vesicles. 2019;8:1578589. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Gores GJ, Herman B, Lemasters JJ. Plasma membrane bleb formation and rupture: a common feature of hepatocellular injury. Hepatology. 1990;11:690–698. [DOI] [PubMed] [Google Scholar]
- 131.Aboelsoud MM, Javaid AI, Al-Qadi MO, et al. Hypoxic hepatitis—its biochemical profile, causes and risk factors of mortality in critically-ill patients: a cohort study of 565 patients. J Crit Care. 2017;41:9–15. [DOI] [PubMed] [Google Scholar]
- 132.Birrer R, Takuda Y, Takara T. Hypoxic hepatopathy: pathophysiology and prognosis. Intern Med. 2007;46:1063–1070. [DOI] [PubMed] [Google Scholar]
- 133.Shimabukuro-Vornhagen A, Godel P, Subklewe M, et al. Cytokine release syndrome. J Immunother Cancer. 2018;6:56. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Winkler U, Jensen M, Manzke O, et al. Cytokine-release syndrome in patients with B-cell chronic lymphocytic leukemia and high lymphocyte counts after treatment with an anti-CD20 monoclonal antibody (rituximab, IDEC-C2B8). Blood. 1999;94:2217–2224. [PubMed] [Google Scholar]
- 135.Freeman CL, Morschhauser F, Sehn L, et al. Cytokine release in patients with CLL treated with obinutuzumab and possible relationship with infusion-related reactions. Blood. 2015;126:2646–2649. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Silver J, Garcia-Neuer M, Lynch DM, et al. Endophenotyping oxaliplatin hypersensitivity: personalizing desensitization to the atypical platin. J Allergy Clin Immunol Pract. 2020;8:1668–1680.e2. [DOI] [PubMed] [Google Scholar]
- 137.Nakamura N, Kanemura N, Shibata Y, et al. Lenalidomide-induced cytokine release syndrome in a patient with multiple myeloma. Leuk Lymphoma. 2014;55:1691–1693. [DOI] [PubMed] [Google Scholar]
- 138.Badar T, Szabo A, Advani A, et al. Real-world outcomes of adult B-cell acute lymphocytic leukemia patients treated with blinatumomab. Blood Adv. 2020;4:2308–2316. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Brudno JN, Kochenderfer JN. Toxicities of chimeric antigen receptor T cells: recognition and management. Blood. 2016;127:3321–3330. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Weemhoff JL, Woolbright BL, Jenkins RE, et al. Plasma biomarkers to study mechanisms of liver injury in patients with hypoxic hepatitis. Liver Int. 2017;37:377–384. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Edgar AD, Tomkiewicz C, Costet P, et al. Fenofibrate modifies transaminase gene expression via a peroxisome proliferator activated receptor alpha-dependent pathway. Toxicol Lett. 1998;98:13–23. [DOI] [PubMed] [Google Scholar]
- 142.Thulin P, Rafter I, Stockling K, et al. PPARalpha regulates the hepatotoxic biomarker alanine aminotransferase (ALT1) gene expression in human hepatocytes. Toxicol Appl Pharmacol. 2008;231:1–9. [DOI] [PubMed] [Google Scholar]
- 143.Kantarjian H, Stein A, Gokbuget N, et al. Blinatumomab versus chemotherapy for advanced acute lymphoblastic leukemia. N Engl J Med. 2017;376:836–847. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Josekutty J, Iqbal J, Iwawaki T, et al. Microsomal triglyceride transfer protein inhibition induces endoplasmic reticulum stress and increases gene transcription via Ire1alpha/cJun to enhance plasma ALT/AST. J Biol Chem. 2013;288:14372–14383. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Thulin P, Bamberg K, Buler M, et al. The peroxisome proliferator-activated receptor alpha agonist, AZD4619, induces alanine aminotransferase-1 gene and protein expression in human, but not in rat hepatocytes: correlation with serum ALT levels. Int J Mol Med. 2016;38:961–968. [DOI] [PubMed] [Google Scholar]
- 146.Lewis JH, Jadoul M, Block GA, et al. Effects of bardoxolone methyl on hepatic enzymes in patients with type 2 diabetes mellitus and stage 4 CKD. Clin Transl Sci. 2021;14:299–309. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Ecker DM, Jones SD, Levine HL. The therapeutic monoclonal antibody market. MAbs. 2015;7:9–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Bakema JE, van Egmond M. Fc receptor-dependent mechanisms of monoclonal antibody therapy of cancer. Curr Top Microbiol Immunol. 2014;382:373–392. [DOI] [PubMed] [Google Scholar]
- 149.Muro H, Shirasawa H, Maeda M, et al. Fc receptors of liver sinusoidal endothelium in normal rats and humans. A histologic study with soluble immune complexes. Gastroenterology. 1987;93:1078–1085. [DOI] [PubMed] [Google Scholar]
- 150.Perussia B. Fc receptors on natural killer cells. Curr Top Microbiol Immunol. 1998;230:63–88. [DOI] [PubMed] [Google Scholar]
- 151.Vogelpoel LT, Baeten DL, de Jong EC, et al. Control of cytokine production by human fc gamma receptors: implications for pathogen defense and autoimmunity. Front Immunol. 2015;6:79. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.Galasso PJ, Litin SC, O’Brien JF. The macroenzymes: a clinical review. Mayo Clin Proc. 1993;68:349–354. [DOI] [PubMed] [Google Scholar]
- 153.Lee M, Vajro P, Keeffe EB. Isolated aspartate aminotransferase elevation: think macro-AST. Dig Dis Sci. 2011;56:311–13. [DOI] [PubMed] [Google Scholar]
- 154.Ono S, Kurata C, Nishimura N, et al. Importance of laboratory detection of macro-aspartate aminotransferase. Int J Gen Med. 2019;12:433–436. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155.Parks D, Lin X, Painter JL, et al. A proposed modification to Hy’s law and Edish criteria in oncology clinical trials using aggregated historical data. Pharmacoepidemiol Drug Saf. 2013;22:571–578. [DOI] [PubMed] [Google Scholar]
- 156.Dufour DR, Lott JA, Nolte FS, et al. Diagnosis and monitoring of hepatic injury. I. Performance characteristics of laboratory tests. Clin Chem. 2000;46:2027–2049. [PubMed] [Google Scholar]
- 157.van der Lely AJ, Biller BM, Brue T, et al. Long-term safety of pegvisomant in patients with acromegaly: comprehensive review of 1288 subjects in ACROSTUDY. J Clin Endocrinol Metab. 2012;97:1589–1597. [DOI] [PubMed] [Google Scholar]
- 158.Freda PU, Gordon MB, Kelepouris N, et al. Long-term treatment with pegvisomant as monotherapy in patients with acromegaly: experience from ACROSTUDY. Endocr Pract. 2015;21:264–274. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159.Costa-Moreira P, Gaspar R, Pereira P, et al. Role of liver biopsy in the era of clinical prediction scores for “drug-induced liver injury” (DILI): experience of a tertiary referral hospital. Virchows Arch. 2020;477:517–525. [DOI] [PubMed] [Google Scholar]
- 160.European Association for the Study of the Liver. EASL Clinical Practice Guidelines: Drug-induced liver injury. J Hepatol. 2019;70:1222–1261. [DOI] [PubMed] [Google Scholar]
- 161.Ettel MG, Appelman HD. Approach to the liver biopsy in the patient with chronic low-level aminotransferase elevations. Arch Pathol Lab Med. 2018;142:1186–1190. [DOI] [PubMed] [Google Scholar]
- 162.Roth SE, Avigan MI, Bourdet D, et al. Next-generation DILI biomarkers: prioritization of biomarkers for qualification and best practices for biospecimen collection in drug development. Clin Pharmacol Ther. 2020;107:333–346. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163.Hunt CM, Papay JI, Stanulovic V, et al. Drug rechallenge following drug-induced liver injury. Hepatology. 2017;66:646–654. [DOI] [PubMed] [Google Scholar]
- 164.Watanabe H, Kubo T, Ninomiya K, et al. The effect and safety of immune checkpoint inhibitor rechallenge in non-small cell lung cancer. Jpn J Clin Oncol. 2019;49:762–765. [DOI] [PubMed] [Google Scholar]
- 165.Kitagawa S, Hakozaki T, Kitadai R, et al. Switching administration of anti-PD-1 and anti-PD-L1 antibodies as immune checkpoint inhibitor rechallenge in individuals with advanced non-small cell lung cancer: case series and literature review. Thorac Cancer. 2020;11:1927–1933. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166.Gobbini E, Charles J, Toffart AC, et al. Current opinions in immune checkpoint inhibitors rechallenge in solid cancers. Crit Rev Oncol Hematol. 2019;144:102816. [DOI] [PubMed] [Google Scholar]
- 167.Dolladille C, Ederhy S, Sassier M, et al. Immune checkpoint inhibitor rechallenge after immune-related adverse events in patients with cancer. JAMA Oncol. 2020;6:865–871. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168.Miller ED, Abu-Sbeih H, Styskel B, et al. Clinical characteristics and adverse impact of hepatotoxicity due to immune checkpoint inhibitors. Am J Gastroenterol. 2020;115:251–261. [DOI] [PubMed] [Google Scholar]
- 169.Patrinely JR, Jr, McGuigan B, Chandra S, et al. A multicenter characterization of hepatitis associated with immune checkpoint inhibitors. Oncoimmunology. 2021;10:1875639. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170.Li M, Sack JS, Rahma OE, et al. Outcomes after resumption of immune checkpoint inhibitor therapy after high-grade immune-mediated hepatitis. Cancer. 2020;126:5088–5097. [DOI] [PMC free article] [PubMed] [Google Scholar]