Opinion Review Open Access
Copyright ©The Author(s) 2021. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Meta-Anal. Dec 28, 2021; 9(6): 488-495
Published online Dec 28, 2021. doi: 10.13105/wjma.v9.i6.488
Is dose modification or discontinuation of nilotinib necessary in nilotinib-induced hyperbilirubinemia?
You-Wen Tan, Department of Hepatology, The Third Hospital of Zhenjiang Affiliated Jiangsu University, Zhenjiang 212003, Jiangsu Province, China
ORCID number: You-Wen Tan (0000-0002-5464-1407).
Author contributions: Tan YW completed the paper independently.
Supported by the Social Development Project of Jiangsu Province, No. BE2020775; and Medical Project of Health Department Jiangsu Province, No. H2018021.
Conflict-of-interest statement: The author has no conflict of interest to disclose.
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: http://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: You-Wen Tan, MD, Chief Doctor, Professor, Department of Hepatology, The Third Hospital of Zhenjiang Affiliated Jiangsu University, No. 300 Danjiamen, Runzhouqu, Zhenjiang 212003, Jiangsu Province, China. tyw915@sina.com
Received: December 29, 2020
Peer-review started: December 29, 2020
First decision: October 11, 2021
Revised: October 14, 2021
Accepted: December 24, 2021
Article in press: December 24, 2021
Published online: December 28, 2021

Abstract

Nilotinib is a specific breakpoint cluster region-Abelson leukemia virus-tyrosine kinase inhibitor that is used as an effective first- or second-line treatment in imatinib-resistant chronic myelogenous leukemia (CML) patients. Hepatotoxicity due to nilotinib is a commonly reported side effect; however, abnormal liver function test (LFT) results have been reported in asymptomatic cases. When alanine aminotransferase (ALT) and aspartate aminotransferase (AST) levels are more than five-fold the upper limit of the normal (ULN) or when the serum total bilirubin level is more than three-fold the ULN, dose modification or discontinuation of nilotinib is recommended, resulting in decreased levels of hematological indicators in certain patients with CML. Nilotinib-induced hyperbilirubinemia typically manifests as indirect bilirubinemia without elevated ALT or AST levels. Such abnormal liver functioning is thus not attributed to the presence of a true histologic lesion of the liver. The underlying mechanism may be related to the inhibition of uridine diphosphate glucuronosyltransferase activity. Therefore, nilotinib dose adjustment is not recommended for this type of hyperbilirubinemia, and in the absence of elevated liver enzyme levels or presence of abnormal LFT findings, physicians should consider maintaining nilotinib dose intensity without modifications.

Key Words: Tyrosine kinase inhibitors, Nilotinib, Chronic myelogenous leukemia, Hyperbilirubinemia, Drug induced liver injure, Liver injury

Core Tip: Hepatotoxicity due to nilotinib is a commonly reported side effect; however, abnormal liver function test (LFT) results have been reported in asymptomatic cases. Nilotinib-induced hyperbilirubinemia manifests usually as indirect bilirubinemia without observation of elevated alanine aminotransferase or aspartate aminotransferase levels. The underlying mechanism may be related to the inhibition of uridine diphosphate glucuronosyltransferase activity. Therefore, in the absence of elevated levels of liver enzymes or presence of abnormal LFT findings, physicians should consider maintaining nilotinib dose intensity without modifications.



INTRODUCTION

Chronic myelogenous leukemia (CML) originates in hematopoietic stem cells of malignant hyperplastic diseases and accounts for 15%–20% of all leukemia cases, with an incidence of 1–2/100000[1]. CML is one of the most common types of leukemia and is marked by the presence of a breakpoint cluster region (BCR)-Abelson leukemia virus (ABL) on the (9, 22)(C34, C11) chromosome translocation fusion gene, i.e., the gene encoding P210-type fusion protein which exerts a type of tyrosine kinase activity. The subsequent abnormal clonal proliferation of myeloid hematopoietic cells is the primary cause of CML.

BCR-ABL tyrosine kinase plays an important role in cell differentiation, division, adhesion, and stress response, and its constitutive activation can lead to the development of leukemia. BCR-ABL is expressed in tumor cells of 95% of patients with CML, whereas it is not expressed in normal cells. Therefore, specific BCR-ABL tyrosine kinase inhibitors (TKIs) can be used to effectively treat CML[2].

TKIs are known to induce hepatotoxicity[3]. A systematic study conducted on the basis of 12 previous studies demonstrated that TKIs are associated with a higher risk of hepatotoxicity, compared to placebos[4]. Regarding hepatotoxicity of grade 3 or higher, the instructions recommend dosage reduction and discontinuation to allow recovery of liver functioning. The objective of this study was to confirm that the distinction between true hepatotoxicity and hyperbilirubinemia alone does not require dosage changes or discontinuation of treatment for hyperbilirubinemia caused by nilotinib alone.

PRESENT STATUS OF NILOTINIB TREATMENT

Nilotinib is a molecularly targeted kinase inhibitor that competitively binds to the platelet-derived growth factor C-kit and ABL kinases, thus it is effective for treating CML. A five-year follow-up survey showed that the 5-year cumulative rate of complete cytogenetic remission was 87%, the estimated 5-year survival rate was 89%, and the rate of progression-free survival was 93%[5]. Up to 98% of the patients achieved hematological remission, and cytogenetic response was achieved in 86% of the patients at the chronic stage after treatment. After 1, 2, 3, and 4 years of continuous treatment, only 3.4%, 7.5%, 4.8%, and 1.5% of the patients, respectively, were expected to experience deterioration. Nilotinib is also considered an effective second-line treatment for imatinib-resistant cases[6,7].

HEPATOTOXICITY OF NILOTINIB

Common adverse reactions to nilotinib include dermatological, digestive, and cardiovascular side effects. Abnormal liver function is a common side effect in nilotinib-treated patients, and a considerable proportion of the patients show elevated levels of liver enzymes and bilirubin, based on analyses of phase II and III trials (Table 1). Up to 70% of the patients exhibit elevated levels of liver enzymes[8], and most patients are asymptomatic, presenting with grade 1–2 abnormal liver function. This typically occurs within 10–14 d of initial treatment, and only 4%–9% of the patients demonstrate a five-fold upper limit of normal (ULN) alanine aminotransferase (ALT) or aspartate aminotransferase (AST) activity, which can be improved by dosage adjustment and treatment discontinuation.

Table 1 Incidence of biochemical abnormalities across the second-line and frontline nilotinib trials.
Patients (%) Second-line trials
Frontline trials
Phase 2
Phase 3
Phase 2
Phase 3

2101[35] (n = 321)
ENACT[36,37] (n = 1422)
MDACC[38] (n = 61)
GIMEMA[17] (n = 76)
ENESTnd[39] 300 mg twice daily (n = 279)
ENESTnd[39] 400 mg twice daily (n = 277)
All gradesGrade 3/4All gradesGrade 3/4All gradesGrade 3/4All gradesGrade 3/4All gradesGrade 3/4All gradesGrade 3/4
Bilirubin elevation (total)72 72543975316534628
ALT elevation694142480428664739
AST elevation55391460293401483

In a real-world study of nilotinib, a marked elevation of ALT and AST levels was not common, and the incidence of an increase to more than five-fold the ULN was 1%–4%. Concurrently, the manifestation of jaundice was mainly attributed to indirect bilirubin levels and self-remission, and it was attributed to Gilbert syndrome (GS)[9]. However, in a nilotinib phase I and II study spanning 36 mo and conducted in Japan, 29% of the patients showed elevated bilirubin levels, 24% showed elevated ALT levels, but only 3% showed over five-fold the ULN[10]. Severe liver failure caused by nilotinib treatment is rare. For example, a 34-year-old woman with CML developed progressive jaundice (bilirubin 14.5 mg/dL, ALT 1856 U/L, and ALP 254 U/L) after 8 mo of nilotinib treatment. She was subsequently diagnosed with liver failure and underwent liver transplantation.

COMPARISON OF HEPATOTOXICITY INDUCED BY NILOTINIB AND IMATINIB

Nilotinib is clinically used because of resistance to or side effects of imatinib treatments. In the ENESTnd trial, a phase 3 randomized trial including 846 patients[11], two different doses of nilotinib showed ALT activity rates of over five-fold the ULN at 4% and 9%, respectively, compared with 3% with imatinib. Only two patients showed relatively severe liver dysfunction (data not reported), indicating that the frequency of liver dysfunction due to nilotinib treatment was higher than that of imatinib. During the 5-year follow-up of the same group of patients, the rate of abnormal liver function was 50%–60%, but liver failure was implicated in none of the reported 50 cases of death[12]. A 47-year-old woman with CML presented with liver function deterioration (bilirubin level 20 mg/dL, alanine aminotransferase level 828 U/L prothrombin time 24 s) during imatinib therapy administrated before liver transplantation, and the postoperative treatment with nilotinib did not result in liver damage[13]. In a group of 88 imatinib-resistant CML patients who received nilotinib for 3 years, 14% showed elevated ALT and ALP levels. Interestingly, five patients with imatinib-induced hepatotoxicity did not exhibit any recurrence of nilotinib-induced hepatotoxicity[14].

THE MAIN SYMPTOM OF NILOTINIB-INDUCED HEPATOTOXICITY IS UNCONJUGATED HYPERBILIRUBINEMIA

Elevated levels of bilirubin are a common adverse drug reactions to nilotinib treatment, which occurs in 3%–16% of the patients, but constitutes no evident pathological significance[15]. In a previous study, 119 patients with CML with imatinib resistance were treated with nilotinib; nine patients subsequently showed elevated levels of indirect bilirubin, and three showed elevated ALT levels. None of the increased parameters influenced the hematological remission effect[16].

In studies on nilotinib-induced hepatotoxicity, hyperbilirubinemia is the most common etiology of hepatotoxicity and is the main reason reducing or discontinuing nilotinib (Table 1). Hyperbilirubinemia is mainly characterized by the presence of unconjugated bilirubin. The initial median time of increase was 18 d, and the duration was approximately 8 d. A spontaneous decrease in bilirubin levels may occur without medication or phototherapy.

In a case reported by Cortes et al[17], a 39-year-old man with CML was administered with low-dose (300 mg/d) nilotinib for continuous treatment in the frontline GIMEMA trial, and his bilirubin levels increased gradually, resulting in the development of grade-4 hyperbilirubinemia. Ten days after drug discontinuation, the hyperbilirubinemia was resolved. Moreover, the original dose of nilotinib was continued, and hyperbilirubinemia development was observed along with four recurrences. Bilirubinemia was maintained at grade 2–3, and notably, the patients demonstrated a complete molecular response 1 year after treatment. Therefore, hyperbilirubinemia is considered a benign condition[18].

NILOTINIB-INDUCED HYPERBILIRUBINEMIA IS ASSOCIATED WITH URIDINE DIPHOSPHATE GLUCURONOSYLTRANSFERASE (UGT1A1) ACTIVITY

UGT1A1 is a membrane protein that binds to the endoplasmic reticulum. It is an enzyme that plays a crucial role in phase II biotransformation of several endogenous and exogenous substances[19]. The UGT1A1 gene is predominantly expressed in the human liver, but also in bile tissue, the large intestine, and the stomach, and it is responsible for bilirubin binding[20]. UGT1A1 is a key enzyme involved in the metabolism of bilirubin. UGT1A1 expression is correlated with UGT1A1 activity, which leads to abnormal synthesis of unconjugated bilirubin in the blood and excretion of conjugated bilirubin in vitro[21], resulting in hyperbilirubinemia[22]. Hyperbilirubinemia has been observed in patients treated with TKIs. A correlation between the inhibition of UGT1A1 activity and an increase in bilirubin level was observed in patients treated with TKI. The gene polymorphism of UGT1A1 is the main reason underlying the function of the UGT1A1 gene[23].

The promoter region of the UGT1A1 gene exhibits a certain polymorphism, and its atypical TATA box region contains 5–8 TA repeats. The most common genotypes included six TA repeats[24]. UGT1A1 expression decreases with increasing TA repeats. A variant of UGT1A1 containing an untypical TATA box region of the UGT1A1 * 28 promoter contains seven TA repeats; it is associated with decreased UGT1A1 expression and frequently causes hyperbilirubinemia of unknown etiology[25]. Furthermore, new variants have been reported, including UGT1A1 *6/*6, *6/*28, and *28/*28[26].

Nilotinib inhibited UGT1A1 activity in a dose-and time-dependent manner. Abumiya et al[27] investigated 34 CML patients treated with low and high dosages of nilotinib, and hyperbilirubinemia was more common in patients treated with high dosages and over long periods. The proportion of patients harboring UGT1A1 *6/*6 and *6/*28 genotypes (poor metabolizers) was also higher[27]. This suggests that variations in the UGT1A1 gene should be considered when administering nilotinib.

MECHANISM UNDERLYING NILOTINIB-INDUCED HYPERBILIRUBINEMIA

Nilotinib inhibits the activities of cytochrome P450 enzymes CYP3A4, CYP2C8, CYP2C9, CYP2D6, and UGT1A1[28]. Fujita et al[29] have proven nilotinib to be an effective non-competitive drug for human UGT1A1 in vitro. In this study, SN-38 was used as a substrate, and human liver microsome and recombinant human UGT1A1 were used as enzyme sources to study the inhibitory effect of nilotinib on UGT1A1-catalyzed SN-38 glucuronidation. The results showed that nilotinib exerted a noncompetitive inhibitory effect on SN-38 glucuronization of human liver microsome UGT1A1 and recombinant human UGT1A1 with K (i) values of 0.286 ± 0.0094 and 0.079 ± 0.0029 μm, respectively.

UGT1A1 GENOTYPE DOES NOT AFFECT NILOTINIB EFFICACY AND SAFETY

TKIs are currently used for the treatment of CML. Nilotinib can increase the level of bilirubin by inhibiting UGT1A1 activity[30,31] In imatinib- and dasatinib-treated GS patients, elevated levels of bilirubin may occur; thus, it is not known whether such a condition of hyperbilirubinemia affects the efficacy and safety of nilotinib. We previously reported the case of a 24-year-old CML patient with unconjugated hyperbilirubinemia treated with nilotinib[32]. Reduction and discontinuation of treatment can improve bilirubin levels immediately, but complete cytogenetic response (CCyR) worsens. CCyR can be achieved using a normal dose, but hyperbilirubinemia without ALT and other abnormal enzyme activities can be induced again. After conducting repeated trials four times, a normal dose of nilotinib was administered, and no pathological damage was observed in liver pathology. Although high bilirubin (grade 3–4) levels persisted, the patient achieved continuous CCyR.

In a 16-year retrospective study[33], long-term hematological toxicity and hepatocellular toxicity of imatinib, nilotinib, and dasatinib were observed. The effect of the GS genotype on progression-free survival was evaluated. One hundred and five patients with CML-CP who were consecutively treated with either first- or second-generation TKIs were evaluated. Gilbert's syndrome genotypes were distributed as follows: 17 (16.2%) patients with 7/7, 44 (41.9%) patients with 6/7, and the remaining cases with wild-type genotypes. The results showed that there was no difference in either the major molecular response or complete cytogenetic response observed among the different GS genotypes. Hyperbilirubinemia was observed in 26 patients, and grade 3/4 was observed in 9 patients. However, no true endothelial toxicity was observed in patients who continued to use TKIs.

Therefore, the European LeukemiaNet recommendations state that hyperbilirubinemia caused by TKIs is more common during nilotinib treatment and that this characteristic is the most frequently reported laboratory adverse event, rather than representing a true liver injury. This phenomenon is more common in the UGT1A1-mutated population and should be monitored closely, while dose adjustment has not been suggested[34].

This type of hyperbilirubinemia does not necessitate dose adjustment to achieve improvements, but bilirubin levels may spontaneously decline, a phenomenon which has been recognized by several scholars[18].

CONCLUSION

In conclusion, nilotinib is a commonly used drug in the treatment of CML, during which hyperbilirubinemia is commonly observed, and hyperbilirubinemia with indirect bilirubin elevation is not accompanied by abnormal hepatocyte enzymes. This abnormality is not real drug-induced liver damage, but the variation in UGT1A1 leads to inhibition of UGT1A1 activity, resulting in disorders of the bilirubin metabolism[34]. Therefore, neither dose modification nor discontinuation is recommended for patients with hyperbilirubinemia at more than three-fold the ULN, and the benefits of maintaining the original dose to CML outweigh the disadvantages caused by dose reduction or treatment discontinuation. A different abnormality, mainly accompanied by elevated levels of liver enzymes, should follow the standard of reduction or drug discontinuation at more than three-fold the ULN.

Footnotes

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

Peer-review model: Single blind

Specialty type: Gastroenterology and hepatology

Country/Territory of origin: China

Peer-review report’s scientific quality classification

Grade A (Excellent): 0

Grade B (Very good): 0

Grade C (Good): C, C

Grade D (Fair): D

Grade E (Poor): 0

P-Reviewer: Cengiz M, Nabil A S-Editor: Liu M L-Editor: Wang TQ P-Editor: Liu M

References
1.  Lyman GH, Henk HJ. Association of Generic Imatinib Availability and Pricing With Trends in Tyrosine Kinase Inhibitor Use in Patients With Chronic Myelogenous Leukemia. JAMA Oncol. 2020;6:1969-1971.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 8]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
2.  Talpaz M, Shah NP, Kantarjian H, Donato N, Nicoll J, Paquette R, Cortes J, O'Brien S, Nicaise C, Bleickardt E, Blackwood-Chirchir MA, Iyer V, Chen TT, Huang F, Decillis AP, Sawyers CL. Dasatinib in imatinib-resistant Philadelphia chromosome-positive leukemias. N Engl J Med. 2006;354:2531-2541.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1298]  [Cited by in F6Publishing: 1240]  [Article Influence: 68.9]  [Reference Citation Analysis (0)]
3.  Chow EJ, Doody DR, Wilkes JJ, Becker LK, Chennupati S, Morin PE, Winestone LE, Henk HJ, Lyman GH. Adverse events among chronic myelogenous leukemia patients treated with tyrosine kinase inhibitors: a real-world analysis of health plan enrollees. Leuk Lymphoma. 2021;62:1203-1210.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 1]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
4.  Teo YL, Ho HK, Chan A. Risk of tyrosine kinase inhibitors-induced hepatotoxicity in cancer patients: a meta-analysis. Cancer Treat Rev. 2013;39:199-206.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 66]  [Cited by in F6Publishing: 74]  [Article Influence: 6.2]  [Reference Citation Analysis (0)]
5.  Druker BJ, Guilhot F, O'Brien SG, Gathmann I, Kantarjian H, Gattermann N, Deininger MW, Silver RT, Goldman JM, Stone RM, Cervantes F, Hochhaus A, Powell BL, Gabrilove JL, Rousselot P, Reiffers J, Cornelissen JJ, Hughes T, Agis H, Fischer T, Verhoef G, Shepherd J, Saglio G, Gratwohl A, Nielsen JL, Radich JP, Simonsson B, Taylor K, Baccarani M, So C, Letvak L, Larson RA; IRIS Investigators. Five-year follow-up of patients receiving imatinib for chronic myeloid leukemia. N Engl J Med. 2006;355:2408-2417.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2588]  [Cited by in F6Publishing: 2488]  [Article Influence: 138.2]  [Reference Citation Analysis (0)]
6.  Hussain S, Usman Shaikh M. Response and Adverse Effects of Nilotinib in Imatinib-resistant Chronic Myeloid Leukemia Patients: Data From a Developing Country. Clin Ther. 2015;37:2449-2457.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 3]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
7.  Takahashi N, Miura M, Kuroki J, Mitani K, Kitabayashi A, Sasaki O, Kimura H, Imai K, Tsukamoto N, Noji H, Kondo T, Motegi M, Kato Y, Mita M, Saito H, Yoshida C, Torimoto Y, Kimura T, Wano Y, Nomura J, Yamamoto S, Mayama K, Honma R, Sugawara T, Sato S, Shinagawa A, Abumiya M, Niioka T, Harigae H, Sawada K. Multicenter phase II clinical trial of nilotinib for patients with imatinib-resistant or -intolerant chronic myeloid leukemia from the East Japan CML study group evaluation of molecular response and the efficacy and safety of nilotinib. Biomark Res. 2014;2:6.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 20]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
8.  Hughes TP, Hochhaus A, Kantarjian HM, Cervantes F, Guilhot F, Niederwieser D, le Coutre PD, Rosti G, Ossenkoppele G, Lobo C, Shibayama H, Fan X, Menssen HD, Kemp C, Larson RA, Saglio G. Safety and efficacy of switching to nilotinib 400 mg twice daily for patients with chronic myeloid leukemia in chronic phase with suboptimal response or failure on front-line imatinib or nilotinib 300 mg twice daily. Haematologica. 2014;99:1204-1211.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 36]  [Cited by in F6Publishing: 38]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
9.  Deremer DL, Ustun C, Natarajan K. Nilotinib: a second-generation tyrosine kinase inhibitor for the treatment of chronic myelogenous leukemia. Clin Ther. 2008;30:1956-1975.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 115]  [Cited by in F6Publishing: 118]  [Article Influence: 7.9]  [Reference Citation Analysis (0)]
10.  Usuki K, Tojo A, Maeda Y, Kobayashi Y, Matsuda A, Ohyashiki K, Nakaseko C, Kawaguchi T, Tanaka H, Miyamura K, Miyazaki Y, Okamoto S, Oritani K, Okada M, Usui N, Nagai T, Amagasaki T, Wanajo A, Naoe T. Efficacy and safety of nilotinib in Japanese patients with imatinib-resistant or -intolerant Ph+ CML or relapsed/refractory Ph+ ALL: a 36-month analysis of a phase I and II study. Int J Hematol. 2012;95:409-419.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 14]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
11.  Kantarjian HM, Hochhaus A, Saglio G, De Souza C, Flinn IW, Stenke L, Goh YT, Rosti G, Nakamae H, Gallagher NJ, Hoenekopp A, Blakesley RE, Larson RA, Hughes TP. Nilotinib vs imatinib for the treatment of patients with newly diagnosed chronic phase, Philadelphia chromosome-positive, chronic myeloid leukaemia: 24-month minimum follow-up of the phase 3 randomised ENESTnd trial. Lancet Oncol. 2011;12:841-851.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 354]  [Cited by in F6Publishing: 391]  [Article Influence: 30.1]  [Reference Citation Analysis (0)]
12.  Hochhaus A, Saglio G, Hughes TP, Larson RA, Kim DW, Issaragrisil S, le Coutre PD, Etienne G, Dorlhiac-Llacer PE, Clark RE, Flinn IW, Nakamae H, Donohue B, Deng W, Dalal D, Menssen HD, Kantarjian HM. Long-term benefits and risks of frontline nilotinib vs imatinib for chronic myeloid leukemia in chronic phase: 5-year update of the randomized ENESTnd trial. Leukemia. 2016;30:1044-1054.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 500]  [Cited by in F6Publishing: 547]  [Article Influence: 68.4]  [Reference Citation Analysis (0)]
13.  Perini GF, Santos FP, Funke V, Ruiz J, Neto BH, Hamerschlak N. Nilotinib post-liver transplantation for acute hepatic failure related to imatinib. Leuk Res. 2009;33:e234-e235.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in F6Publishing: 13]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
14.  Koren-Michowitz M, le Coutre P, Duyster J, Scheid C, Panayiotidis P, Prejzner W, Rowe JM, Schwarz M, Goldschmidt N, Nagler A. Activity and tolerability of nilotinib: a retrospective multicenter analysis of chronic myeloid leukemia patients who are imatinib resistant or intolerant. Cancer. 2010;116:4564-4572.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 19]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
15.  Breccia M, Alimena G. Nilotinib therapy in chronic myelogenous leukemia: the strength of high selectivity on BCR/ABL. Curr Drug Targets. 2009;10:530-536.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 5]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
16.  Kantarjian H, Giles F, Wunderle L, Bhalla K, O'Brien S, Wassmann B, Tanaka C, Manley P, Rae P, Mietlowski W, Bochinski K, Hochhaus A, Griffin JD, Hoelzer D, Albitar M, Dugan M, Cortes J, Alland L, Ottmann OG. Nilotinib in imatinib-resistant CML and Philadelphia chromosome-positive ALL. N Engl J Med. 2006;354:2542-2551.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 998]  [Cited by in F6Publishing: 930]  [Article Influence: 51.7]  [Reference Citation Analysis (0)]
17.  Cortes JE, Jones D, O'Brien S, Jabbour E, Konopleva M, Ferrajoli A, Kadia T, Borthakur G, Stigliano D, Shan J, Kantarjian H. Nilotinib as front-line treatment for patients with chronic myeloid leukemia in early chronic phase. J Clin Oncol. 2010;28:392-397.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 184]  [Cited by in F6Publishing: 184]  [Article Influence: 13.1]  [Reference Citation Analysis (0)]
18.  Rosti G, Castagnetti F, Gugliotta G, Palandri F, Baccarani M. Physician's guide to the clinical management of adverse events on nilotinib therapy for the treatment of CML. Cancer Treat Rev. 2012;38:241-248.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 26]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
19.  van der Mey D, Gerisch M, Jungmann NA, Kaiser A, Yoshikawa K, Schulz S, Radtke M, Lentini S. Drug-drug interaction of atazanavir on UGT1A1-mediated glucuronidation of molidustat in human. Basic Clin Pharmacol Toxicol. 2021;128:511-524.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 7]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
20.  Varughese LA, Lau-Min KS, Cambareri C, Damjanov N, Massa R, Reddy N, Oyer R, Teitelbaum U, Tuteja S. DPYD and UGT1A1 Pharmacogenetic Testing in Patients with Gastrointestinal Malignancies: An Overview of the Evidence and Considerations for Clinical Implementation. Pharmacotherapy. 2020;40:1108-1129.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 13]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
21.  Hulshof EC, Deenen MJ, Guchelaar HJ, Gelderblom H. Pre-therapeutic UGT1A1 genotyping to reduce the risk of irinotecan-induced severe toxicity: Ready for prime time. Eur J Cancer. 2020;141:9-20.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 35]  [Article Influence: 8.8]  [Reference Citation Analysis (0)]
22.  Li Z, Song L, Hao L. The role of UGT1A1 (c.-3279 T > G) gene polymorphisms in neonatal hyperbilirubinemia susceptibility. BMC Med Genet. 2020;21:218.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 4]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
23.  Qosa H, Avaritt BR, Hartman NR, Volpe DA. In vitro UGT1A1 inhibition by tyrosine kinase inhibitors and association with drug-induced hyperbilirubinemia. Cancer Chemother Pharmacol. 2018;82:795-802.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 30]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
24.  Bahr TM, Agarwal AM, Christensen RD. Does heterozygosity for UGT1A1 *28 convey increased risk for severe neonatal jaundice? J Perinatol. 2021;41:658-660.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 1]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
25.  Saeki M, Saito Y, Jinno H, Sai K, Ozawa S, Kurose K, Kaniwa N, Komamura K, Kotake T, Morishita H, Kamakura S, Kitakaze M, Tomoike H, Shirao K, Tamura T, Yamamoto N, Kunitoh H, Hamaguchi T, Yoshida T, Kubota K, Ohtsu A, Muto M, Minami H, Saijo N, Kamatani N, Sawada JI. Haplotype structures of the UGT1A gene complex in a Japanese population. Pharmacogenomics J. 2006;6:63-75.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 68]  [Cited by in F6Publishing: 71]  [Article Influence: 3.9]  [Reference Citation Analysis (0)]
26.  Fukui T, Mitsufuji H, Kubota M, Inaoka H, Hirose M, Iwabuchi K, Masuda N, Kobayashi H. Prevalence of topoisomerase I genetic mutations and UGT1A1 polymorphisms associated with irinotecan in individuals of Asian descent. Oncol Lett. 2011;2:923-928.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 6]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
27.  Abumiya M, Takahashi N, Niioka T, Kameoka Y, Fujishima N, Tagawa H, Sawada K, Miura M. Influence of UGT1A1 6, 27, and 28 polymorphisms on nilotinib-induced hyperbilirubinemia in Japanese patients with chronic myeloid leukemia. Drug Metab Pharmacokinet. 2014;29:449-454.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 31]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
28.  Kim MJ, Lee JW, Oh KS, Choi CS, Kim KH, Han WS, Yoon CN, Chung ES, Kim DH, Shin JG. The tyrosine kinase inhibitor nilotinib selectively inhibits CYP2C8 activities in human liver microsomes. Drug Metab Pharmacokinet. 2013;28:462-467.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 9]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
29.  Fujita K, Sugiyama M, Akiyama Y, Ando Y, Sasaki Y. The small-molecule tyrosine kinase inhibitor nilotinib is a potent noncompetitive inhibitor of the SN-38 glucuronidation by human UGT1A1. Cancer Chemother Pharmacol. 2011;67:237-241.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 36]  [Cited by in F6Publishing: 37]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
30.  Ha VH, Jupp J, Tsang RY. Oncology Drug Dosing in Gilbert Syndrome Associated with UGT1A1: A Summary of the Literature. Pharmacotherapy. 2017;37:956-972.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 17]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
31.  Perry HM Jr, Kopp SJ, Perry EF, Erlanger MW. Hypertension and associated cardiovascular abnormalities induced by chronic barium feeding. J Toxicol Environ Health. 1989;28:373-388.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 23]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
32.  Tan Y, Ye Y, Zhou X. Nilotinib-induced liver injury: A case report. Medicine (Baltimore). 2020;99:e22061.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 6]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
33.  Iurlo A, Bucelli C, Cattaneo D, Levati GV, Viani B, Tavazzi D, Consonni D, Baldini L, Cappellini MD. UGT1A1 genotype does not affect tyrosine kinase inhibitors efficacy and safety in chronic myeloid leukemia. Am J Hematol. 2019;94:E283-E285.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 3]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
34.  Steegmann JL, Baccarani M, Breccia M, Casado LF, García-Gutiérrez V, Hochhaus A, Kim DW, Kim TD, Khoury HJ, Le Coutre P, Mayer J, Milojkovic D, Porkka K, Rea D, Rosti G, Saussele S, Hehlmann R, Clark RE. European LeukemiaNet recommendations for the management and avoidance of adverse events of treatment in chronic myeloid leukaemia. Leukemia. 2016;30:1648-1671.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 323]  [Cited by in F6Publishing: 309]  [Article Influence: 38.6]  [Reference Citation Analysis (0)]
35.  le Coutre PD, Giles FJ, Hochhaus A, Apperley JF, Ossenkoppele GJ, Blakesley R, Shou Y, Gallagher NJ, Baccarani M, Cortes J, Kantarjian HM. Nilotinib in patients with Ph+ chronic myeloid leukemia in accelerated phase following imatinib resistance or intolerance: 24-month follow-up results. Leukemia. 2012;26:1189-1194.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 69]  [Cited by in F6Publishing: 73]  [Article Influence: 5.6]  [Reference Citation Analysis (0)]
36.  Nicolini FE, Masszi T, Shen Z, Gallagher NJ, Jootar S, Powell BL, Dorlhiac-Llacer PE, Zheng M, Szczudlo T, Turkina A. Expanding Nilotinib Access in Clinical Trials (ENACT), an open-label multicenter study of oral nilotinib in adult patients with imatinib-resistant or -intolerant chronic myeloid leukemia in accelerated phase or blast crisis. Leuk Lymphoma. 2012;53:907-914.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 23]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
37.  Nicolini FE, Turkina A, Shen ZX, Gallagher N, Jootar S, Powell BL, De Souza C, Zheng M, Szczudlo T, le Coutre P. Expanding Nilotinib Access in Clinical Trials (ENACT): an open-label, multicenter study of oral nilotinib in adult patients with imatinib-resistant or imatinib-intolerant Philadelphia chromosome-positive chronic myeloid leukemia in the chronic phase. Cancer. 2012;118:118-126.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 47]  [Cited by in F6Publishing: 52]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
38.  Rosti G, Palandri F, Castagnetti F, Breccia M, Levato L, Gugliotta G, Capucci A, Cedrone M, Fava C, Intermesoli T, Cambrin GR, Stagno F, Tiribelli M, Amabile M, Luatti S, Poerio A, Soverini S, Testoni N, Martinelli G, Alimena G, Pane F, Saglio G, Baccarani M; GIMEMA CML Working Party. Nilotinib for the frontline treatment of Ph(+) chronic myeloid leukemia. Blood. 2009;114:4933-4938.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 159]  [Cited by in F6Publishing: 160]  [Article Influence: 10.7]  [Reference Citation Analysis (0)]
39.  Saglio G, Kim DW, Issaragrisil S, le Coutre P, Etienne G, Lobo C, Pasquini R, Clark RE, Hochhaus A, Hughes TP, Gallagher N, Hoenekopp A, Dong M, Haque A, Larson RA, Kantarjian HM; ENESTnd Investigators. Nilotinib vs imatinib for newly diagnosed chronic myeloid leukemia. N Engl J Med. 2010;362:2251-2259.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1228]  [Cited by in F6Publishing: 1176]  [Article Influence: 84.0]  [Reference Citation Analysis (0)]