Open Access

Dose adjustment for tyrosine kinase inhibitors in non‑small cell lung cancer patients with hepatic or renal function impairment (Review)

  • Authors:
    • Dehua Zhao
    • Jing Chen
    • Xiaoqing Long
    • Jisheng Wang
  • View Affiliations

  • Published online on: November 27, 2020     https://doi.org/10.3892/or.2020.7870
  • Pages: 413-426
  • Copyright: © Zhao et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

In recent years, a number of tyrosine kinase inhibitors (TKIs) have been approved for the treatment of non‑small cell lung cancer. These novel treatments exhibit improved efficacy and toxicity when compared to conventional chemotherapy agents. TKIs are administered orally, which has the advantages of improved flexibility and convenience for the patients. However, challenges have arisen in the use of these novel agents. Prescribing drugs for patients with hepatic or renal function impairment poses a challenge for clinicians due to the large pharmacokinetic variability in each individual patient. Moreover, several TKIs have been shown to cause laboratory test abnormalities normally associated with hepatic or renal injury. The aim of the present review was to discuss the effects of hepatic and renal function impairment on the pharmacokinetic variability of 17 TKIs and their potential hepatotoxicity and nephrotoxicity, and to recommend dose adjustment for patients with hepatic or renal impairment.

Introduction

Lung cancer remains the leading cause of cancer-related mortality, and non-small-cell lung cancer (NSCLC) represents ~85% of all cases (1). NSCLC has been shown to be driven by various activated oncogenes, such as epidermal growth factor receptor (EGFR) and anaplastic lymphoma kinase (ALK) (24). The identification of genetic driver alterations in patients with NSCLC and the use of effective targeted agents have been recommended by the National Comprehensive Cancer Network and European Society for Medical Oncology guidelines, offering new promising strategies for patients with NSCLC (5,6). To date, a number of small-molecule targeted drugs have been developed for the treatment of NSCLC, most of which are tyrosine kinase inhibitors (TKIs). Compared with traditional chemotherapy drugs, TKIs are associated with significantly improved clinical outcomes and reduced treatment-associated toxicity (7). As TKIs are given orally at a fixed dose for long periods of time, they represent an easy and convenient drug regimen for patients with cancer. However, administration of TKIs is also associated with certain risks, such as a large variability in pharmacokinetics (PK), particularly in patients with concurrent hepatic impairment (HI) or renal impairment (RI) (8,9).

HI and RI are common in patients with cancer, and they are intrinsic factors that may affect the absorption, distribution, metabolism and excretion of orally administered drugs (8,9). As a result of the alterations in drug PK, effective drug exposure may increase or decrease, leading to suboptimal efficacy or substantial toxicity. In addition, certain TKIs may induce hepatic or renal toxicities. Thus, understanding the effects of HI and RI on PK variability of specific anticancer drugs and their potential hepatotoxicity and nephrotoxicity is crucial for selecting the appropriate doses and dosing intervals, thus insuring maximum efficacy and minimal toxicity. The aim of the present review was to summarize the potential hepatotoxicity and nephrotoxicity of 17 TKIs and their PK variability in patients with HI or RI. Furthermore, based on available evidence from drug labels and published literature, dose recommendations for the 17 TKIs are provided for NSCLC patients with varying degrees of HI or RI.

TKIs approved for the treatment of NSCLC

As mentioned above, the identification of genetic driver alterations has created new therapeutic interventions for patients with NSCLC (5,6). Driver mutations identified in NSCLC include EGFR mutations, ALK rearrangement, c-ros oncogene (ROS1) rearrangement, B-RAF proto-oncogene (BRAF) V600E mutations, neurotrophin tyrosine receptor kinase gene fusions, high-level MET amplification or MET exon 14 skipping mutations, and RET rearrangements (5). As a number of the identified driver mutations activate tyrosine kinase signaling pathways, strategies to inhibit these pathways by TKIs have been developed and approved for the treatment of NSCLC (1056). The TKIs approved for the treatment of patients with NSCLC and their PK properties are listed in Table I.

Table I.

Pharmacokinetics properties of TKIs approved for the treatment of patients with non-small cell lung cancer.

Table I.

Pharmacokinetics properties of TKIs approved for the treatment of patients with non-small cell lung cancer.

Metabolic enzymes Excretion


TKIsTargetsPB (%)Vd/Vss (l)MajorMinorTransportersUrinary (%)Fecal (%)
AfatinibEGFR, HER2/4952,770aCYP450FMO3P-gp, BCRP485
AlectinibALK, RET>99475aCYP3A4NANA<0.598
BrigatinibALK, ROS1, IGF-1R, FLT-3, EGFR91153aCYP2C8, CYP3A4CYP3A5P-gp, BCRP2565
CabozantinibMET, VEGFR-1/2/3, AXL, RET, ROS1, TYRO3, MER, KIT, TRKB, FLT-3, TIE-2≥99.7319CYP3A4CYP2C9MRP22754
CeritinibALK, IGF-1R, InsR, ROS1974,230CYP3ANAP-gp, BCRP1.392
CrizotinibALK, c-Met, ROS1, RON911,772aCYP3ANAP-gp, OATP1B1, OATP1B3, OATP2B1, OCT22263
DabrafenibBRAF V600E/V600K/V600D, CRAF, SIK1, NEK11, LIMK199.770.3CYP2C8, CYP3A4NAP-gp, BCRP2371
DacomitinibEGFR, HER2/4, DDR1/2, EPHA6, LCK, MNK1981,889aCYP2D6CYP3A4, CYP2C9P-gp, BCRP  379
EntrectinibTRKA/B/C, ROS1, ALK, JAK2, ACK1>99551CYP3A4NANA  383
ErlotinibEGFR93232CYP3A4CYP1A2, CYP1A1P-gp  883
GefitinibEGFR901,400aCYP3A4CYP2D6, CYP3A5P-gp, OATP1B3<486
LarotrectinibTRKA/B/C, TNK2,7048aCYP3A4, CYP3A5NAP-gp, BCRP3958
LorlatinibALK, ROS1, TYK1, FER, FPS, TRKA/B/C, FAK, FAK2, ACK66305aCYP3A4, UGT1A4CYP2C8, CYP2C19, CYP3A5, UGT1A3NA4841
OsimertinibEGFR, HER2/3/4, ACK1, BLK95918aCYP3A4, CYP3A5NAP-gp, BCRP1468
TrametinibMEK1/2, BRAF V60097.4214 CarboxylesterasesNAP-gp, BSEP<20>80
VandetanibEGFR, VEGFR, RET, BRK, TIE2, EPH, SRC947,450CYP3A4FMO1/3OATP1B1, OATP1B32544
VemurafenibBRAF V600E, CRAF, ARAF, SRMS, ACK1, MAP4K5, FGR>99106Non-CYP enzymesCYP3A4P-gp, BCRP  194

a Steady state volume of distribution (Vss). TKIs, tyrosine kinase inhibitors; PB, plasma protein binding; Vd, apparent volume of distribution; NA, not applicable/not available; HER, human epidermal growth factor receptor; DDR, discoidin domain receptor tyrosine kinase; EPHA, ephrin type-A receptor; LCK, leukocyte-specific protein-tyrosine kinase; MNK, MAPK-interacting serine/threonine-protein kinase; ACK, activated Cdc42-associated kinase; BLK, B lymphoid tyrosine kinase; C-met, mesenchymal-epithelial transition factor; RON, tyrosine kinase receptor macrophage-stimulating receptor; IGF-1R, insulin-like growth factor 1 receptor; InsR, insulin receptors; RET, Rearranged during transfection proto-oncogene gene; FLT, FMS-like tyrosine kinase; TYK, Tyrosine kinase; FER, Fes-related kinase; TRK, tropomyosin-related kinase; FAK, focal adhesion kinase; JAK, Janus kinase; SIK, salt-inducible kinase; NEK, never in mitosis gene A-related expressed kinase; LIMK, LIM kinase; MEK, mitogen-activated protein kinase kinase; VEGFR, vascular endothelial growth factor; SRC, Rous sarcoma oncogene; UGT, UDP-glucuronosyltransferase; FOM, flavin-containing monooxygenase; P-gp, P-glycoprotein; BCRP, breast cancer resistance protein; OATP, organic anion transporting peptide; OCT, organic cation transporters; BSEP, bile salt export pump transporter; MRP, multidrug resistance-associated protein.

Dose adjustment for patients with HI

The liver is involved in the absorption, distribution and elimination of most drugs and their metabolites (8). HI does not only affect the hepatic metabolism or biliary excretion, but may also affect protein binding (PB), intestinal enzymes and transporters, which may ultimately affect drug absorption and distribution (8,57,58). The effect of HI on the absorption of orally administered drugs is mainly the result of a reduced first-pass effect, leading to an increased bioavailability of oral drugs in patients with HI (59). Similarly, HI may increase the distribution of many drugs by decreasing the PB. Mechanisms underlying the decreased binding of drugs to plasma proteins include low levels of plasma proteins, accumulation of endogenous substances [e.g., bilirubin (Bil)], and qualitative changes in plasma proteins (60). As most drugs are metabolized by the liver, impaired hepatic function may decrease their metabolism by reducing the levels and activity of hepatic enzymes and transporters and, consequently, alter the exposure to drugs and their metabolites (61). In addition to decreased metabolizing capacity and decreased PB, reduced biliary excretion may also occur in patients with HI (62). Moreover, HI may also affect kidney function, leading to an accumulation of a drug and its metabolites, even when the liver is not primarily responsible for their elimination (63).

The extent of alterations in PK is associated with the degree of HI (58). The US Food and Drug Administration (FDA) and European Medicines Agency (EMA) recommend using the Child-Pugh (C-P) classification to categorize the degree of HI (64,65). However, studies have often used other grading systems to assess liver function (66), and the results from these different grading systems are not interchangeable and, hence, are difficult to compare with one another. Therefore, multiple HI descriptions are presented in our dose recommendations. The classification of HI by C-P score and National Cancer Institute (NCI) criteria are listed in Table II.

Table II.

Classification of hepatic impairment by Child-Pugh score and NCI.

Table II.

Classification of hepatic impairment by Child-Pugh score and NCI.

DegreeChild-Pugh classification (points)NCI classification
MildA (56)TBil ≤ULN and AST >ULN, or TBil >1-1.5× ULN and any AST
ModerateB (79)TBil >1.5 3× ULN and any AST
SevereC (1015)TBil >3 10× ULN and any AST

[i] NCI, National Cancer Institute; TBil, total bilirubin; AST, aspartate aminotransferase; ULN, upper limit of normal.

Taking the aforementioned points into account, HI may increase the bioavailability and distribution of orally administered drugs, reduce metabolizing capacity, biliary and renal excretion, consequently increasing the drug exposure (8,58). Therefore, dose adjustment should be considered in patients with HI. The dose adjustment recommendations for TKIs in NSCLC patients with HI are summarized in Table III.

Table III.

Dose adjustment recommendations for TKIs in cases of HI.

Table III.

Dose adjustment recommendations for TKIs in cases of HI.

Dose adjustment recommendations (Refs.)

TKIsALT/AST/BilNCI classificationC-P classification
AfatinibNKNKC-P A and B: No dose adjustment (10,11)
C-P C: Close monitoring (10); not recommended (11)
AlectinibNKMild and moderate HI: No dose adjustment (12)C-P A and B: No dose adjustment (1214)
Severe HI: NKC-P C: 450 mg bid (1214)
BrigatinibNKNKC-P A and B: No dose adjustment (15,16)
C-P C: 60% dose reduction (15)
CabozantinibNKMild HI: No dose adjustment (17,18)C-P A and B: 66% dose reduction (1719,20,21)
Moderate and severe HI: NKC-P C: Not recommended (1719)
CeritinibNKNKC-P A and B: No dose adjustment (22,23)
C-P C: 300 mg qd (22,23)
CrizotinibALT/AST >5× ULN with TBil ≤1.5× ULN,Mild HI: No dose adjustment (24,25)NK
treatment suspension (20); ALT/AST >3× ULNModerate HI: 200 mg bid (2426)
with TBil >1.5× ULN, permanent discontinuation (20)Severe HI: 250 mg qd (2426)
DabrafenibNKMild HI: No dose adjustment (2729)NK
Moderate and severe HI: Use with caution (28)
DacomitinibNKMild and moderate HI: No dose adjustment (30)C-P A and B: No dose adjustment (3032)
Severe HI: Not recommended (31)C-P C: Not recommended (31)
EntrectinibNKMild HI: No dose adjustment (33)NK
Moderate and severe HI: NK
ErlotinibALT/AST >5× ULN, not recommended (11); ASTSevere HI: Close monitoring (34)C-P A and B: Close monitoring (34); use with
≥3× ULN, or DBil >17 µmol/l, 50% caution (35); no dose adjustment (36)
dose reduction (37) C-P C: Not recommended (35); close monitoring (34)
GefitinibALT/AST >5× ULN, treatment suspension (38)Severe HI: Discontinuation (38)C-P A: No dose adjustment (39)
C-P B: Close monitoring (38)
C-P C: Use with caution (40); close monitoring (38)
LarotrectinibNKNKC-P A: No dose adjustment (41,42)
C-P B and C: 50% dose reduction (41,42)
LorlatinibNKMild HI: No dose adjustment (43,44)NK
Moderate and severe HI: Not recommended (44)
OsimertinibNKMild and moderate HI: No dose adjustment (45,46)C-P A and B: No dose adjustment (4547)
Severe HI: Not recommended (45)C-P C: NK
TrametinibNKMild HI: No dose adjustment (4850)NK
Moderate and severe HI: Use with caution (49)
VandetanibNKMild HI: No dose adjustment (51,52)C-P A: No dose adjustment (53) C-P B and
Moderate and severe HI: Not recommended (51,52)C: Not recommended (51); no dose adjustment (52,53)
VemurafenibNKMild and moderate HI: No dose adjustment (54,56)NK
Severe HI: Close monitoring (55)

[i] TKIs, tyrosine kinase inhibitors; HI, hepatic impairment; AST, aspartate aminotransferase; ALT, alanine aminotransferase; TBil, total bilirubin; DBil, direct bilirubin; NK, not known; C-P, Child-Pugh; NCI, National Cancer Institute; ULN, upper limit of normal; bid, twice daily; qd, once daily.

Afatinib

Afatinib is metabolized only to a minor extent (~9%) and, in the circulation, forms covalent adducts with plasma proteins. Excretion of afatinib is primarily via the feces (10,11). As shown in PK studies, exposure to afatinib was not significantly altered in patients with C-P A and B; therefore, dose adjustment in this population is not deemed necessary (10,11). Subjects with C-P C have not been studied; therefore, the recommendation is to use afatinib with caution or not at all in this patient population (10,11).

Alectinib

Alectinib is eliminated primarily by the liver (12,13). In a clinical study, the combined area under the plasma drug concentration-time curve (AUC) of alectinib and its major metabolite (M4) increased by 36 and 76% in subjects with C-P B and C, respectively (14). The combined Cmax of alectinib and M4 increased by 16% in patients with C-P B and decreased by 2% in patients with C-P C (14). Therefore, dose adjustment is not necessary for patients with C-P A and B, but is necessary for patients with CP-C. In these patients, the dose should be reduced to 450 mg twice daily (1214).

Brigatinib

Brigatinib is eliminated primarily via the liver (15,16). The brigatinib AUC was by 37% higher in subjects with C-P C compared to subjects with normal hepatic function (NHF), while subjects with C-P A and B had a similar brigatinib AUC compared to subjects with NHF (15,16). Based on these data, dose adjustment is not required for patients with C-P A and B (15,16), whereas the dose should be reduced by ~40% (i.e., from 180 to 120 mg, or from 90 to 60 mg) in patients with C-P C (16).

Cabozantinib

Based on a population PK analysis, no clinically significant differences in cabozantinib exposure were observed between subjects with mild HI (based on the NCI criteria) and those with NHF (20). In a HI study, the cabozantinib AUC and Cmax increased by 81 and 10%, respectively, in subjects with C-P A (21); for subjects with C-P B, the cabozantinib AUC increased by 63% and Cmax decreased by 29% (21). As cabozantinib exposure was significantly altered in C-P A and B, dose reduction is necessary for these patients (1721). Cabozantinib is not recommended for use in patients with C-P C, since the safety and efficacy have not been characterized in this population (17,18).

Ceritinib

Ceritinib is primarily eliminated by the liver (22,23). A clinical study was conducted to evaluate the effect of HI on ceritinib PK. The AUC of ceritinib was increased by 18, 2 and 66% in subjects with C-P A, B and C, respectively, compared with subjects with NHF (22,23). Based on these data, dose adjustment is not required for patients with C-P A and B, but the ceritinib dose should be reduced by approximately one-third in patients with C-P C (22,23).

Crizotinib

Crizotinib is eliminated primarily by the liver (24,25). Following oral crizotinib administration at doses of 250 mg twice daily, patients with mild HI (based on the NCI criteria) exhibited similar crizotinib exposure at steady state compared to patients with NHF, with AUC and Cmax decreasing by 9% (26). Therefore, dose adjustment is not required for patients with mild HI (2426). Patients with moderate HI exhibited higher crizotinib exposure compared to patients with NHF, with AUC and Cmax increasing by 50 and 44%, respectively (26). The crizotinib AUC decreased by 35% and Cmax decreased by 27% in patients with severe HI following crizotinib 250 mg orally once daily compared with patients with NHF following crizotinib 250 mg orally twice daily (26). Based on these results, crizotinib 200 mg twice daily is recommended for patients with moderate HI, and 250 mg once daily for patients with severe HI (2426).

Dabrafenib

Based on a population PK analysis, the effect of mild HI (based on the NCI criteria) on the concentration of dabrafenib or its metabolites was not significant (29). Therefore, dose adjustment is not required for patients with mild HI (2729). No data are available for patients with moderate to severe HI. However, considering that dabrafenib is mainly eliminated by the liver, patients with moderate to severe HI should be treated with caution (28).

Dacomitinib

Dacomitinib is eliminated primarily by the liver (30,31). In a dedicated HI trial, the AUC and Cmax of dacomitinib were unchanged in subjects with C-P A, whereas they decreased by 15 and 20%, respectively, in subjects with C-P B, as compared to subjects with NHF (32). Based on this trial, C-P A and B exerted no clinically important effects on the PK of dacomitinib. In addition, based on a population PK analysis, dose adjustment is not required for patients with mild and moderate HI (according to the NCI criteria) (30,31). The effect of severe HI (both NCI and C-P criteria) on dacomitinib PK is unknown, and treatment in this patient population is not recommended (31).

Entrectinib

Entrectinib is eliminated primarily via the liver (33). In a population PK analysis, no clinically significant differences in the PK of entrectinib were observed in patients with mild HI (based on the NCI criteria) compared with subjects with NHF (33). Therefore, dose adjustment is not required in this patient population (33). The impact of moderate to severe HI on the PK of entrectinib is unknown; therefore, there are currently no dose recommendations for these populations.

Erlotinib

Erlotinib is primarily eliminated by the liver (34,35). The PK and safety profiles of erlotinib in patients with C-P B were found to be similar to those with NHF; therefore, dose adjustment is not required for patients with C-P B (36). No data are available regarding the effect of C-P C on the PK of erlotinib; therefore, erlotinib must be used with caution, or not at all, in patients with CP-C (34,35). In a phase I study, the erlotinib clearance was reduced and dose-limiting toxicities were increased in patients with HI [aspartate aminotransferase ≥3× upper limit of normal (ULN), or direct Bil >17 µmol/l] (37). Therefore, a 50% dose reduction was recommended for patients with this level of HI (37).

Gefitinib

Gefitinib is cleared primarily by the liver (38,39). In a study of non-cancer patients with HI due to cirrhosis, the exposure to gefitinib was increased by 40, 263 and 166% in patients with C-P A, B and C, respectively, compared to healthy subjects (40). However, the PK of gefitinib is highly variable in subjects with HI due to cirrhosis and, considering the low dose reduction rate observed when gefitinib was administered at a dose 2-fold higher than the recommended dose, no dose adjustment was recommended in patients with HI due to cirrhosis (3840). In case of patients with cancer, the exposure of gefitinib was similar between patients with moderate and severe HI (according to the NCI criteria) when compared to patients with NHF (40). Based on these results, dose adjustment may not be necessary for patients with HI; however, considering its hepatotoxicity, close monitoring is required for patients with C-P B and C due to liver metastases (3840).

Larotrectinib

Larotrectinib is eliminated primarily via the liver (41,42). In a PK study, the larotrectinib AUC increased by 30, 100 and 220%, and Cmax increased by 10, 10 and 50% in patients with C-P A, B and C, respectively, when compared to subjects with NHF (41,42). Therefore, dose adjustment is not required for patients with C-P A, but the starting dose of larotrectinib should be reduced by 50% in patients with C-P B and C (41,42).

Lorlatinib

Lorlatinib is primarily metabolized in the liver and excreted in both the urine (48%) and feces (41%) (43,44). Based on a population PK analysis, no clinically meaningful differences in lorlatinib PK were observed in patients with mild HI (based on the NCI criteria) (43). Therefore, dose adjustment is not required in patients with mild HI (43,44). The effect of moderate to severe HI on lorlatinib PK is unknown; therefore, lorlatinib is not recommended for use in these patients (44).

Osimertinib

Osimertinib is eliminated primarily via the liver (45,46). In a clinical trial, the osimertinib AUC and Cmax decreased by 36.7 and 48.6%, respectively, in patients with C-P A, and by 31.6 and 39.3%, respectively, in patients with C-P B, when compared to patients with NHF (47). In terms of safety, there were no apparent differences in safety between patients with NHF and those with C-P A or B (47). In a population PK analysis, the PK of osimertinib exhibited no clinically significant differences in patients with mild or moderate HI (according to the NCI criteria), compared to patients with NHF (45,46). Therefore, based on the population PK analysis and clinical study results, dose adjustment is not necessary for patients with mild and moderate HI (both NCI and C-P criteria) (4547). Since no study has been conducted in patients with severe HI, osimertinib is not recommended for use in this population (46).

Trametinib

Trametinib is eliminated primarily via the liver (48,49). Formal clinical studies have not been conducted to assess the effect of organ impairment on the PK of trametinib. A population PK analysis demonstrated that mild HI did not affect the PK of trametinib (50); therefore, dose adjustment is not required for this patient population (4850). The PK of trametinib has not been investigated in patients with moderate to severe HI; therefore, trametinib should be administered with caution in this patient population (49).

Vandetanib

A PK study demonstrated that the vandetanib AUC increased by 4% in patients with C-P A and decreased by 6 and 7% in patients with C-P B and C, respectively, compared to patients with NHF (53). Given that exposure to vandetanib was not altered in subjects with HI, no dose adjustment appears to be necessary in patients with C-P A, B and C (5153).

Vemurafenib

Vemurafenib is eliminated primarily via the liver (54,55). In clinical trials, the apparent clearance of vemurafenib was similar between patients with mild to moderate HI (based on the NCI criteria) and those with NHF, indicating that dose adjustment is not required for patients with mild to moderate HI (54,56). As few patients with severe HI were enrolled in clinical trials, close monitoring is required when vemurafenib is administered to patients with severe HI (55).

Dose adjustment for patients with RI

The kidney and the liver are the two main organs responsible for the elimination of drugs. Impaired renal function may decrease the excretion of a drug or its metabolites that are primarily or partly eliminated via the kidneys (9,58). In addition, RI may also affect the non-renal disposition of drugs that are eliminated by the liver via alterations in the expression and activity of drug-metabolizing enzymes and transporters in the liver (67,68). Although the most obvious changes with RI affect the elimination of drugs as well as their metabolites, RI may also be associated with other changes, such as absorption and PB (9). Alterations in drug absorption in patients with RI may result from delayed gastric emptying, changes in gut motility, changes in gastric pH, decrease in first-pass effect, and vomiting or diarrhea (69). Alterations in PB are a result of hypoalbuminemia and accumulation of endogenous substances (e.g., uremic toxins) (69).

The extent of PK alterations depends on the degree of RI (9,58). For example, the AUC of afatinib increased by 22 and 50% in patients with moderate and severe RI, respectively, when compared to patients with normal renal function (NRF) (70). There are several methods currently used to categorize the degree of RI in patients, among which estimated glomerular filtration rate (eGFR) is the most commonly used in clinical practice and has been included in the FDA and EMA guidelines (71). Therefore, eGFR is used as a measure of renal function in order to make dose recommendations.

In conclusion, RI not only affects the PK of renally eliminated drugs, but may also affect drugs not eliminated by the kidneys (58,67). If a drug's PK is significant altered in patients with RI, a dose adjustment is required to ensure its efficacy and safety. TKIs are considered safer than chemotherapy drugs, but may still cause serious side effects when their systemic concentrations and exposure are increased (72). Therefore, dose adjustments are required for certain TKIs in patients with RI. The dose adjustment recommendations for TKIs in NSCLC patients with RI are summarized in Table IV.

Table IV.

Dose adjustment recommendations for TKIs in cases of renal impairment.

Table IV.

Dose adjustment recommendations for TKIs in cases of renal impairment.

Dose adjustment recommendations (Refs.)

ESRDd

TKIsMild impairmentaModerate impairmentbSevere impairmentcNot on HDRequiring HD
AfatinibNo dose adjustment (10,11)No dose adjustment (10,11,70)30 mg qd (10)Not recommended (11)30 mg qd (73,74)
AlectinibNo dose adjustment (12,13)No dose adjustment (12,13)Withhold (12) No dose adjustment (13)Treatment suspension (12)50% dose reduction (75)
BrigatinibNo dose adjustment (15,16)No dose adjustment (15,16)50% dose reduction (15)NKNK
CabozantinibNo dose adjustment (17,18,21) Use with caution (19)No dose adjustment (17,18,21) Use with caution (19)Not recommended (19)Not recommended (19)NK
CeritinibNo dose adjustment (22,23,76)No dose adjustment (22,23,76)Use with caution (23)Use with caution (23)NK
CrizotinibNo dose adjustment (24,25,77,78)No dose adjustment (24,25,77,78)250 mg qd (24,25)250 mg qd (79)250 mg qd (80)
DabrafenibNo dose adjustment (2729)No dose adjustment (2729)Use with caution (28)Use with caution (28)75 mg qd (81)
DacomitinibNo dose adjustment (30,31)No dose adjustment (30,31)NKNKNK
EntrectinibNo dose adjustment (33)No dose adjustment (33)NKNKNK
ErlotinibNo dose adjustment (35)No dose adjustment (35)Not recommended (35) No dose adjustment (37)Not recommended (35)No dose adjustment (82,83)
GefitinibNo dose adjustment (39)No dose adjustment (39)eGFR >20 ml/min, no dose adjustment; ≤20 ml/min, use with caution (39)Use with caution (15) No dose adjustment (84)No dose adjustment (8588)
LarotrectinibNo dose adjustment (41,42)No dose adjustment (41,42)No dose adjustment (41,42)No dose adjustment (41,42)No dose adjustment (41,42)
LorlatinibNo dose adjustment (43,44)No dose adjustment (43,44)Not recommended (44)Not recommended (44)NK
OsimertinibNo dose adjustment (45,46)No dose adjustment (45,46)Use with caution (46) No dose adjustment (89)Use with caution (46)No dose adjustment (9092)
TrametinibNo dose adjustment (4850)No dose adjustment (4850)Use with caution (49)Use with caution (49)0.5 mg qd (81)
VandetanibNo dose adjustment (5153)200 mg qd (51) Not recommended (52)200 mg qd (51) Not recommended (52)200 mg qd (51) Not recommended (52)NK
VemurafenibNo dose adjustment (5456)No dose adjustment (5456)Close monitoring (55)Close monitoring (55)720 mg bid (93)

a eGFR 60-89 ml/min.

b eGFR 30-59 ml/min.

c eGFR 15-29 ml/min.

d eGFR <15 ml/min. TKIs, tyrosine kinase inhibitors; NK, not known; qd, once daily; bid, twice daily; ESRD, end-stage renal disease; eGFR, estimated glomerular filtration rate; HD, hemodialysis.

Afatinib

Compared with patients with NRF, the afatinib AUC and Cmax increased by 50 and 22%, respectively, in subjects with severe RI and increased by 22 and 1%, respectively, in subjects with moderate RI (70). Based on these results, dose adjustment is not required for patients with mild to moderate RI, but a 25% dose reduction is recommended for patients with severe RI (10,11,70). In case reports, afatinib at a dose of 30 mg once daily was tolerated and effective for patients with end-stage renal disease (ESRD) undergoing hemodialysis (HD) (73,74).

Alectinib

From a patient population PK analysis, mild to moderate RI exerted no clinically meaningful effect on the exposure of alectinib and its major metabolite (M4), suggesting that dose adjustment is not required in patients with mild to moderate RI (12,13). The PK of alectinib have not been studied in patients with severe RI, but a case report demonstrated that a patient with ESRD undergoing HD was safely treated with alectinib at a dose of 600 mg daily. The patient exhibited no disease progression for 34 months (75).

Brigatinib

The brigatinib AUC was 86% higher in subjects with severe RI compared to subjects with NRF (15,16). Based on a population PK analysis, the PK of brigatinib was similar between patients with NRF and those with mild to moderate RI (15,16). Thus, dose adjustment is not required for patients with mild to moderate RI, but the dose should be reduced by ~50% in patients with severe RI (15).

Cabozantinib

Results from a study in patients with RI indicated that the cabozantinib AUC and Cmax were 30 and 19% higher, respectively, for subjects with mild RI, and 2 and 6% higher, respectively, for subjects with moderate RI compared to subjects with NRF (21). Based on the small exposure increase with mild to moderate RI, dose adjustment is not required for patients with mild to moderate RI (1721). Since patients with severe RI have not been studied, cabozantinib is not recommended for use in this patient population (19).

Ceritinib

Based on patient population PK analysis, the ceritinib AUC in patients with mild to moderate RI was predicted to increase by 9 and 19%, respectively, compared to patients with NRF (76). These differences are not considered to be clinically relevant; hence, dose adjustment is not deemed necessary in patients with mild to moderate RI (22,23,76). Caution is recommended in patients with severe RI since there are no published PK data available for ceritinib treatment in this population (23).

Crizotinib

Patients with mild and moderate RI had a similar crizotinib exposure compared to patients with NRF (77). Similarly, a population PK analysis revealed that mild to moderate RI did not significantly affect the PK of crizotinib (78). Due to the small size of the increases in crizotinib exposure (5-15%), dose adjustment is not required for patients with mild to moderate RI (24,25,77,78). However, patients with severe RI exhibited increases in crizotinib AUC and Cmax of 79 and 34%, respectively, compared to subjects with NRF (77). Thus, crizotinib at a dose of 250 mg once daily is recommended for patients with severe RI (24,25,77). A case report demonstrated that crizotinib 250 mg once daily was tolerated and effective for a patient with ESRD not undergoing HD (79). In another case report, treatment with crizotinib 250 mg once daily in a patient with ESRD undergoing HD was well-tolerated, without any significant adverse events (80).

Dabrafenib

Based on a patient population PK analysis, mild to moderate RI did not exert an effect on the concentrations of dabrafenib or its metabolites; therefore, dose adjustment is not required for patients with mild to moderate RI (2729). The effect of severe RI on dabrafenib PK has not been established; therefore, dabrafenib should be administered with caution to patients with severe RI (28). A case report revealed that a patient with ESRD undergoing HD continued dabrafenib 75 mg once daily and trametinib 0.5 mg once daily for 9 months, and this dose was well-tolerated and effective for the patient (81).

Dacomitinib

Based on population PK analysis, patients with mild to moderate RI had comparable dacomitinib exposure to those with NRF, indicating that dose adjustment is not required in these patients (30,31). The PK of dacomitinib has not been characterized in patients with severe RI; therefore, there are no dose adjustment recommendations for this patient group (30,31).

Entrectinib

In a population PK analysis, no clinically significant differences in the PK of entrectinib were observed in patients with mild to moderate RI; therefore, dose adjustment is not required in patients with mild to moderate RI (33). Sufficient data for severe RI are currently lacking; therefore, dose recommendations cannot be made for these patients (33).

Erlotinib

Based on PK data, erlotinib was almost unaffected by RI; therefore, dose adjustment is not required in patients with mild to moderate RI (35,37). A small study compared the PK of erlotinib and its active metabolite (OSI-420) in 3 patients with ESRD undergoing HD and 5 patients with NRF. Renal function and HD exerted only minor effects on the PK of erlotinib. No serious adverse events were reported in any of the cases, and 1 of the HD patients achieved partial response, indicating that dose adjustment is not required in patients with ESRD undergoing HD (82). A phase I and PK study was conducted to investigate the dose and PK of erlotinib in patients with HI or RI (37). The results demonstrated that patients with RI (creatinine 1.6-5.0 mg/dl) tolerated 150 mg of erlotinib daily and appear to have a clearance similar to that of patients with NRF (37). In addition, a case report revealed that erlotinib was safe and effective for a patient with ESRD undergoing HD (83).

Gefitinib

RI is unlikely to significantly affect the PK of gefitinib, since <4% of gefitinib and its metabolites are excreted by the kidney (38,39). Therefore, the EMA recommended that dose adjustments are not required in patients with eGFR >20 ml/min/1.73 m2, but gefitinib should be administered with caution to patients with eGFR ≤20 ml/min1.73 m2 (39). For patients with ESRD, several case reports revealed that gefitinib administered as 250 mg once daily was safe and effective for patients with ESRD not on HD, or patients undergoing HD or peritoneal dialysis (8488).

Larotrectinib

Following oral administration of a single 100-mg dose of larotrectinib to subjects with ESRD, the larotrectinib AUC increased by 46% and Cmax increased by 25% when compared to subjects with NRF (41,42). Therefore, dose adjustment is not required for patients with RI of any severity (41,42).

Lorlatinib

Based on population PK analysis, no clinically meaningful differences in lorlatinib PK were observed in patients with mild to moderate RI, suggesting that dose adjustment is not required in patients with mild to moderate RI (43,44). The effect of severe RI on lorlatinib PK is unknown; therefore, use of lorlatinib in this patient population is not recommended (44).

Osimertinib

No clinical studies have been conducted to evaluate the effect of RI on the PK of osimertinib. Based on a population PK analysis, the osimertinib exposure was similar among patients with mild to severe RI and those with NRF (45,46,89). Patients with ESRD have not been analyzed, but three case reports revealed that osimertinib at a dose of 80 mg orally once daily was safe and effective for patients with ESRD undergoing HD (9092). Based on these results, dose adjustment is not required for patients with mild to severe RI, or patients with ESRD undergoing HD (45,46,8992).

Trametinib

A population PK analysis demonstrated that mild to moderate RI did not affect the PK of trametinib; therefore, dose adjustment is not required for these patient populations (4850). As the effect of severe RI on the PK of trametinib is unknown, caution is suggested when trametinib is administered to patients with severe RI (49). In one case report, administration of dabrafenib 75 mg once daily combined with trametinib 0.5 mg once daily was well-tolerated and effective for a patient with ESRD undergoing HD (81).

Vandetanib

In a PK study, the AUC and Cmax of vandetanib increased by 46 and 7%, 62 and 9%, and 79 and 11% in patients with mild, moderate and severe RI, respectively, when compared to subjects with NRF (53). Clinical data suggested that dose adjustment is not required in patients with mild RI. The FDA and EMA recommendations differ in regards to patients with moderate to severe RI (51,52). The FDA recommends reducing the vandetanib dose in these patients (51), whereas the EMA indicates that vandetanib is not recommended for these patients due to limited data availability (52).

Vemurafenib

In a population PK analysis, the clearance of vemurafenib in patients with mild to moderate RI was similar to that in patients with NRF (56), indicating that dose adjustment is not required for patients with mild to moderate RI (5456). The effect of severe RI on vemurafenib PK is unknown; therefore; close monitoring is required when vemurafenib is administered to patients with severe RI (55). Based on one case report, administration of vemurafenib at a dose of 720 mg twice daily was safe and effective for a patient with ESRD undergoing HD (93).

Potential, hepatotoxicity and nephrotoxicity of TKIs

Some TKIs may cause hepatic and renal toxicity (94,95), with the most common hepatic toxicities induced by TKIs include increases in the hepatic alkaline phosphatase and Bil. The most common renal toxicities induced by TKIs included proteinuria (with increased serum creatinine), acute kidney injury, chronic kidney disease and renal failure (95). To reduce the risk of hepatic and renal side effects, the use of potentially hepatotoxic or nephrotoxic TKIs requires specific monitoring and, when available, specific prevention methods in patients who already have abnormal hepatic or renal function (9496). The incidence, severity and pattern of hepatic and renal toxicities may differ among different TKIs. The present review summarized the potential hepatotoxicity and nephrotoxicity of 17 TKIs in order to provide information for clinicians and pharmacists when prescribing these drugs to patients with HI or RI. The potential hepatotoxic and nephrotoxic effects of TKIs are listed in Table V.

Table V.

Potential hepatotoxic and nephrotoxic effects of TKIs.

Table V.

Potential hepatotoxic and nephrotoxic effects of TKIs.

TKIs (Refs.)HepatotoxicityNephrotoxicity
Afatinib (10,11,98)Any grade: ALT elevation (20.1-54%),Any grade: Creatinine clearance decreased (49%)
AST elevation (15.1-46%),Grade 3/4: Creatinine clearance decreased (2%)
ALP elevation (34-51%), Bil elevation (16%)Acute renal failure (0.3%)
Grade 3/4: ALT elevation (2%),
AST elevation (0.4-3%), ALP elevation (2-3%),
Bil elevation (1%) Fatal HI (0.2%)
Alectinib (12,13,99,100)Any grade: ALT elevation (14%),Any grade: RI (12%), creatinine increased (7.2%),
AST elevation (15%), ALP elevation (6.2%),AKI (1%)
Bil elevation (18%) Grade 3/4:Grade 3/4: RI (3.9%), creatinine increased (0.7%),
ALT elevation (3.7%), AST elevation (3.7%),AKI (1%)
ALP elevation (0.2%), Bil elevation (3.2%)
Brigatinib (15,16,101)Any grade: ALT elevation (40%),Any grade: Creatinine increased (2%)
AST elevation (65%), ALP elevation (29%),Grade 3/4: Creatinine increased (0%)
Bil elevation (18%) Grade 3/4:
ALT elevation (2.7%), AST elevation (0%),
ALP elevation (0.9%), Bil elevation (3.2%)
Cabozantinib (1719,102)Any grade: ALT elevation (86%),Proteinuria (1-10%), dysuria (1-10%),
AST elevation (86%), ALP elevation (52%),hematuria (1-10%)
Bil elevation (25%) Grade 3/4:Acute renal failure (<1%)
ALT elevation (6%), AST elevation (3%),
ALP elevation (3%), Bil elevation (2%)
Ceritinib (22,23,103)Any grade: ALT elevation (80-91%),Any grade: Creatinine increased (22.1-77%)
AST elevation (75-86%),Grade 3/4: Creatinine increased (0.5-4.2%)
ALP elevation (81%), Bil elevation (15%)Renal failure (2%), RI (1%)
Grade 3/4: ALT elevation (27-34%),
AST elevation (13-21%), ALP elevation (12%),
Bil elevation (0.5-1%)
Crizotinib (24,25,101)Any grade: ALT elevation (76-79%),Any grade: eGFR decreased (76%), creatinine
AST elevation (61-66%), Bil elevation (4.7%)increased (8-23%)
Grade 3/4: ALT elevation (15-17%),Grade 3/4: eGFR decreased (3.6%), creatinine
AST elevation (8-9%), Bil elevation (0%)increased (0.7-2%)
Fatal hepatotoxicity (0.1%),Renal cysts (3-5%), acute renal failure (<1%),
Hepatic failure (1%)renal failure (<1%)
DabrafenibAny grade: ALT elevation (48%),Any grade: Creatinine increased (21%),
(27,28,104,105)AST elevation (57%), ALP elevation (38%)Grade 3/4: Creatinine increased (1.1%)
Grade 3/4: ALT elevation (5%),Renal failure (<1%),
AST elevation (6%), ALP elevation (1%)
Dacomitinib (30,31,106)Any grade: ALT elevation (40%),Any grade: Creatinine increased (24%)
AST elevation (35%), ALP elevation (22%),Grade 3/4: Creatinine increased (0%)
Bil elevation (16%) Grade 3/4:
ALT elevation (1.4%), AST elevation (0.5%),
ALP elevation (0.5%), Bil elevation (0.5%)
Entrectinib (33,107)Any grade: ALT elevation (36%),Any grade: Creatinine increased (73%)
AST elevation (42%), ALP elevation (25%)Grade 3/4: Creatinine increased (2.1%)
Grade 3/4: ALT elevation (2.8%),
AST elevation (2.5%), ALP elevation (0.9%)
Erlotinib (34,35,108,109)Any grade: ALT/AST elevation (35-45%)Severe acute renal failure (0.5%),
Grade 3/4: ALT/AST elevation (10-14%)renal disorders (3.1%)
Hepatic failure, Hepatorenal syndromeGrade 3/4: Renal disorders (1.1%)
Gefitinib (38,39,106,110)Any grade: ALT elevation (34-38%%),Any grade: Creatinine increased (1.5-16%)
AST elevation (40-57%), Bil elevation (22%)Grade 3/4: Creatinine increased (0.5%)
Grade 3/4: ALT elevation (2.4-13%),AKI or nephrotic syndrome (few cases)
AST elevation (2-8%), Bil elevation (0.5%)
Larotrectinib (41,42,111)Any grade: ALT elevation (45%),Any grade: Creatinine increased (13%)
AST elevation (45%), ALP elevation (30%)Grade 3/4: Creatinine increased (0%)
Grade 3/4: ALT elevation (3%),
AST elevation (3%), ALP elevation (3%)
Lorlatinib (43,44,112)Any grade: ALT elevation (28%),Not reported
AST elevation (37%), ALP elevation (24%)
Grade 3/4: ALT elevation (2.1%),
AST elevation (2.1), ALP elevation (1%)
Osimertinib (45,46,113)Any grade: ALT elevation (6-21%),Any grade: Creatinine increased (12.2%)
AST elevation (5-22%), Bil elevation (14%)Grade 3/4: Creatinine increased (0%)
Grade 3/4: ALT elevation (0.7%),
ALT elevation (1.1%), Bil elevation (0%)
TrametinibAny grade: ALT elevation (39%),Any grade: Creatinine increased (21%)
(48,49,104,105)AST elevation (60%), ALP elevation (24%)Grade 3/4: Creatinine increased (1.1%)
Grade 3/4: ALT elevation (3%),
AST elevation (2%), ALP elevation (2%)
Vandetanib (51,52,114)Any grade: ALT elevation (51%),Any grade: Creatinine increased (16%)
Bil elevation (13%) Grade 3/4:Grade 3/4: Creatinine increased (0%)
ALT elevation (2%), Bil elevation (0%)
Vemurafenib (54,55,115)Any grade: ALT elevation (67%),Grade 1/2: Creatinine increased (26-86%)
AST elevation (71%), ALP elevation (69%),Grade 3/4: Creatinine increased (1.2-9.1%)
Bil elevation (33%) Grade 3/4:
ALT elevation (11%), AST elevation (7%),
ALP elevation (7%), Bil elevation (2%)

[i] TKIs, tyrosine kinase inhibitors; AST, aspartate aminotransferase; ALT, alanine aminotransferase; Bil, bilirubin; ALP, alkaline phosphatase; HI, hepatic impairment; RI, renal impairment; AKI, acute kidney injury; eGFR, estimated glomerular filtration rate.

When patients do not exhibit HI and RI prior to TKI treatment but develop TKI-induced HI or RI, the dose modification schedule is based on the grade of the adverse events. For example, if patients experience grade 3 RI during alectinib treatment, the recommendation is to temporarily suspend alectinib until serum creatinine recovers to ≤1.5 times the ULN, then resume at a reduced dose; alectinib should be permanently discontinued if patients experienced grade 4 RI during treatment.

Discussion

Current evidence suggests that severe RI or ESRD may significantly affect the PK of drugs that are primarily eliminated by the liver, whereas the effects are not significant for mild and moderate RI (67,68,94). As shown in the present review, most TKIs are primarily eliminated via the liver, indicating that the effects of mild and moderate RI on the PK of most TKIs are not clinically relevant. It was herein demonstrated that, apart from vandetanib, dose adjustment is not required for the other 16 TKIs in patients with mild or moderate RI. However, for patients with severe RI or ESRD, it is recommended that TKIs be administered with caution and dose reductions be carefully established; otherwise, TKIs should not be used at all.

Since most TKIs are eliminated by the liver, the effects of HI on the PK of TKIs may be more significant compared with the effects of RI (8,58,97). According to the present review, 1 TKI (cabozantinib) requires dose reduction in patients with mild HI; 3 TKIs (crizotinib, larotrectinib, cabozantinib) require dose reduction and 2 TKIs (lorlatinib, vandetanib) are not recommended for use in patients with moderate HI; 5 TKIs (crizotinib, ceritinib, alectinib, brigatinib, larotrectinib) require dose reduction and 11 TKIs are recommended to be used with caution or not at all, in patients with severe HI.

For certain TKIs (e.g., vandetanib), dose adjustment recommendations differ between the FDA and EMA (51,52). In order to provide more data to help physicians decide on individual treatment options, and taking into consideration the regulatory laws of each country, both recommendations were included in this review. Moreover, a number of clinical trials have excluded patients with ESRD undergoing HD, and only a few case reports have reported the safety and efficiency of certain TKIs in patients with ESRD undergoing HD; those case reports were included in this review in order to provide more information on drug administration in patients ESRD undergoing HD.

As some TKIs can induce hepatic and renal toxicities, it is important to monitor hepatic and renal function in patients receiving potentially hepatotoxic or nephrotoxic drugs (98115). If prescription of a hepatotoxic or nephrotoxic drug is necessary for patients with HI or RI, and there are no better tolerated alternatives, preventive methods, including hepatic and renal function monitoring, adverse reactions monitoring and appropriate dosage adjustment, should be considered.

To date, there are several methods available for selecting an appropriate dose in patients with HI or RI, such as physiologically based pharmacokinetics (PBPK) models, two-stage PK studies, and population PK studies (50,56,116,117). However, the physiological bases of the alterations in the absorption, distribution, metabolism and excretion characteristics in patients with HI or RI have not been fully elucidated, and the PK of a drug may display high interindividual variability in patients with HI or RI. Therefore, dose recommendations cannot be made with a high level of granularity and precision (116). To overcome interindividual variability in PK and confirm the attainment of target concentrations, therapeutic drug monitoring may be the best option for patients with HI or RI, and should be considered early after initiation of therapy (118). In addition, close monitoring of potential adverse reactions is required for patients with HI or RI.

Conclusion

The PK profile of a drug for individuals with HI or RI may lead to either increased or decreased drug exposure, necessitating dose adjustment. However, the number of clinical trials that provide data from patients with HI or RI is scarce, particularly for patients with severe HI or RI, making it is difficult to select an appropriate TKI dosage. The aim of the present review was to aid clinicians by providing dose recommendations for 17 TKIs in patients with varying degrees of HI or RI. The described dose adjustment is a preventive method for reduce the risk of toxicities for patients who already have abnormal hepatic or renal function. When patients experience TKI-induced HI or RI, the dose modification schedule is based on the grade of adverse events.

Acknowledgements

Not applicable.

Funding

No funding was received.

Availability of data and materials

All data generated or analyzed during the present study are included in this published article.

Authors' contributions

DZ and JW designed the study and revised the manuscript. DZ, JC and XL performed the literature search. DZ drafted the manuscript. All the authors have read and approved the final version of this manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

All the authors declare that they have no competing interests.

References

1 

Ferlay J, Steliarova-Foucher E, Lortet-Tieulent J, Rosso S, Coebergh JW, Comber H, Forman D and Bray F: Cancer incidence and mortality patterns in Europe: Estimates for 40 countries in 2012. Eur J Cancer. 49:1374–403. 2013. View Article : Google Scholar : PubMed/NCBI

2 

Gridelli C, Rossi A, Carbone DP, Guarize J, Karachaliou N, Mok T, Petrella F, Spaggiari L and Rosell R: Non-small-cell lung cancer. Nat Rev Dis Primers. 1:150092015. View Article : Google Scholar : PubMed/NCBI

3 

Sgambato A, Casaluce F, Maione P and Gridelli C: Targeted therapies in non-small cell lung cancer: A focus on ALK/ROS1 tyrosine kinase inhibitors. Expert Rev Anticancer Ther. 18:71–80. 2018. View Article : Google Scholar : PubMed/NCBI

4 

Lee DH: Treatments for EGFR-mutant non-small cell lung cancer (NSCLC): The road to a success, paved with failures. Pharmacol Ther. 174:1–21. 2017. View Article : Google Scholar : PubMed/NCBI

5 

Ettinger DS, Wood DE, Aggarwal C, Aisner DL, Akerley W, Bauman JR, Bharat A, Bruno DS, Chang JY, Chirieac LR, et al: NCCN guidelines insights: Non-small cell lung cancer, version 1.2020. J Natl Compr Canc Netw. 17:1464–1472. 2019. View Article : Google Scholar : PubMed/NCBI

6 

Planchard D, Popat S, Kerr K, Novello S, Smit EF, Faivre-Finn C, Mok TS, Reck M, Van Schil PE, Hellmann MD, et al: Metastatic non-small cell lung cancer: ESMO clinical practice guidelines for diagnosis, treatment and follow-up. Ann Oncol. 29 (Suppl 4):iv192–iv237. 2018. View Article : Google Scholar

7 

Holleman MS, van Tinteren H, Groen HJ, Al MJ and Uyl-de Groot CA: First-line Tyrosine Kinase inhibitors in EGFR mutation-positive non-small-cell lung cancer: A network meta-analysis. Onco Targets Ther. 12:1413–1421. 2019. View Article : Google Scholar : PubMed/NCBI

8 

Verbeeck RK: Pharmacokinetics and dosage adjustment in patients with hepatic dysfunction. Eur J Clin Pharmacol. 64:1147–1161. 2008. View Article : Google Scholar : PubMed/NCBI

9 

Verbeeck RK and Musuamba FT: Pharmacokinetics and dosage adjustment in patients with renal dysfunction. Eur J Clin Pharmacol. 65:757–773. 2009. View Article : Google Scholar : PubMed/NCBI

10 

US Food and Drug Administration. Label, . simplehttps://www.accessdata.fda.gov/drugsatfda_docs/label/2019/201292s015lbl.pdfFeb 2–2020

11 

European Medicines Agency. Product information, . simplehttps://www.ema.europa.eu/en/documents/product-information/giotrif-epar-product-information_en.pdfFeb 2–2020

12 

US Food and Drug Administration. Label, . simplehttps://www.accessdata.fda.gov/drugsatfda_docs/label/2018/208434s004lbl.pdfFeb 2–2020

13 

European Medicines Agency. Product information, . simplehttps://www.ema.europa.eu/en/documents/product-information/alecensa-epar-product-information_en.pdfFeb 2–2020

14 

Morcos PN, Cleary Y, Sturm-Pellanda C, Guerini E, Abt M, Donzelli M, Vazvaei F, Balas B, Parrott N and Yu L: Effect of hepatic impairment on the pharmacokinetics of alectinib. J Clin Pharmacol. 58:1618–1628. 2018. View Article : Google Scholar : PubMed/NCBI

15 

US Food and Drug Administration. Label, . simplehttps://www.accessdata.fda.gov/drugsatfda_docs/label/2018/208772s004lbl.pdfFeb 2–2020

16 

European Medicines Agency. Product information, . simplehttps://www.ema.europa.eu/en/documents/product-information/alunbrig-epar-product-information_en.pdfFeb 2–2020

17 

US Food and Drug Administration. Label, . simplehttps://www.accessdata.fda.gov/drugsatfda_docs/label/2020/203756s008lbl.pdfFeb 2–2020

18 

US Food and Drug Administration. Label, . simplehttps://www.accessdata.fda.gov/drugsatfda_docs/label/2020/208692s007lbl.pdfFeb 2–2020

19 

European Medicines Agency. Product information, . simplehttps://www.ema.europa.eu/en/documents/product-information/cometriq-epar-product-information_en-0.pdfFeb 2–2020

20 

Nguyen L, Chapel S, Tran BD and Lacy S: Updated population pharmacokinetic model of cabozantinib integrating various cancer types including hepatocellular carcinoma. J Clin Pharmacol. 59:1551–1561. 2019. View Article : Google Scholar : PubMed/NCBI

21 

Nguyen L, Holland J, Ramies D, Mamelok R, Benrimoh N, Ciric S, Marbury T, Preston RA, Heuman DM, Gavis E and Lacy S: Effect of renal and hepatic impairment on the pharmacokinetics of cabozantinib. J Clin Pharmacol. 56:1130–1140. 2016. View Article : Google Scholar : PubMed/NCBI

22 

US Food and Drug Administration. Label, . simplehttps://www.accessdata.fda.gov/drugsatfda_docs/label/2019/205755s016lbl.pdfFeb 2–2020

23 

European Medicines Agency. Product information, . simplehttps://www.ema.europa.eu/en/documents/product-information/zykadia-epar-product-information_en.pdfFeb 2–2020

24 

US Food and Drug Administration. Label, . simplehttps://www.accessdata.fda.gov/drugsatfda_docs/label/2011/202570s000lbl.pdfFeb 2–2020

25 

European Medicines Agency. Product information, . simplehttps://www.ema.europa.eu/en/documents/product-information/xalkori-epar-product-information_en.pdfFeb 2–2020

26 

El-Khoueiry AB, Sarantopoulos J, O'Bryant CL, Ciombor KK, Xu H, O'Gorman M, Chakrabarti J, Usari T and El-Rayes BF: Evaluation of hepatic impairment on pharmacokinetics and safety of crizotinib in patients with advanced cancer. Cancer Chemother Pharmacol. 81:659–670. 2018. View Article : Google Scholar : PubMed/NCBI

27 

US Food and Drug Administration. Label, . simplehttps://www.accessdata.fda.gov/drugsatfda_docs/label/2020/202806s015lbl.pdfFeb 2–2020

28 

European Medicines Agency. Product information, . simplehttps://www.ema.europa.eu/en/documents/product-information/tafinlar-epar-product-information_en.pdfFeb 2–2020

29 

Ouellet D, Gibiansky E, Leonowens C, O'Hagan A, Haney P, Switzky J and Goodman VL: Population pharmacokinetics of dabrafenib, a BRAF inhibitor: Effect of dose, time, covariates, and relationship with its metabolites. J Clin Pharmacol. 54:696–706. 2014. View Article : Google Scholar : PubMed/NCBI

30 

US Food and Drug Administration. Label, . simplehttps://www.accessdata.fda.gov/drugsatfda_docs/label/2018/211288s000lbl.pdfFeb 2–2020

31 

European Medicines Agency. Product information, . simplehttps://www.ema.europa.eu/en/documents/product-information/vizimpro-epar-product-information_en.pdfFeb 2–2020

32 

Giri N, Masters JC, Plotka A, Liang Y, Boutros T, Pardo P, O'Connell J and Bello C: Investigation of the impact of hepatic impairment on the pharmacokinetics of dacomitinib. Invest New Drugs. 33:931–941. 2015. View Article : Google Scholar : PubMed/NCBI

33 

US Food and Drug Administration. Label, . simplehttps://www.accessdata.fda.gov/drugsatfda_docs/label/2019/212725s000lbl.pdfMay 3–2020

34 

US Food and Drug Administration. Label, . simplehttps://www.accessdata.fda.gov/drugsatfda_docs/label/2016/021743s025lbl.pdfFeb 2–2020

35 

European Medicines Agency. Product information, . simplehttps://www.ema.europa.eu/en/documents/product-information/tarceva-epar-product-information_en.pdfFeb 2–2020

36 

O'Bryant CL, Haluska P, Rosen L, Ramanathan RK, Venugopal B, Leong S, Boinpally R, Franke A, Witt K, Evans J, et al: An open-label study to describe pharmacokinetic parameters of erlotinib in patients with advanced solid tumors with adequate and moderately impaired hepatic function. Cancer Chemother Pharmacol. 69:605–612. 2012. View Article : Google Scholar : PubMed/NCBI

37 

Miller AA, Murry DJ, Owzar K, Hollis DR, Lewis LD, Kindler HL, Marshall JL, Villalona-Calero MA, Edelman MJ, Hohl RJ, et al: Phase I and pharmacokinetic study of erlotinib for solid tumors in patients with hepatic or renal dysfunction: CALGB 60101. J Clin Oncol. 25:3055–3060. 2007. View Article : Google Scholar : PubMed/NCBI

38 

US Food and Drug Administration. Label, . simplehttps://www.accessdata.fda.gov/drugsatfda_docs/label/2005/021399s008lbl.pdfFeb 2–2020

39 

European Medicines Agency. Product information, . simplehttps://www.ema.europa.eu/en/documents/product-information/gefitinib-mylan-epar-product-information_en.pdfFeb 2–2020

40 

Horak J, White J, Harris AL, Verrill M, Carmichael J, Holt A, Cantarini M, Macpherson M, Swaisland A, Swaisland H and Twelves C: The effect of different etiologies of hepatic impairment on the pharmacokinetics of gefitinib. Cancer Chemother Pharmacol. 68:1485–1495. 2011. View Article : Google Scholar : PubMed/NCBI

41 

US Food and Drug Administration. Label, . simplehttps://www.accessdata.fda.gov/drugsatfda_docs/label/2018/210861s000lbl.pdfFeb 2–2020

42 

European Medicines Agency. Product information, . simplehttps://www.ema.europa.eu/en/documents/product-information/vitrakvi-epar-product-information_en.pdfFeb 2–2020

43 

US Food and Drug Administration. Label, . simplehttps://www.accessdata.fda.gov/drugsatfda_docs/label/2018/210868s000lbl.pdfFeb 2–2020

44 

European Medicines Agency. Product information, . simplehttps://www.ema.europa.eu/en/documents/product-information/lorviqua-epar-product-information_en.pdfFeb 2–2020

45 

US Food and Drug Administration. Label, . simplehttps://www.accessdata.fda.gov/drugsatfda_docs/label/2019/208065s013lbl.pdfFeb 2–2020

46 

European Medicines Agency. Product information, . simplehttps://www.ema.europa.eu/en/documents/product-information/tagrisso-epar-product-information_en.pdfFeb 2–2020

47 

Grande E, Harvey RD, You B, Batlle JF, Galbraith H, Sarantopoulos J, Ramalingam SS, Mann H, So K, Johnson M and Vishwanathan K: Pharmacokinetic study of osimertinib in cancer patients with mild or moderate hepatic impairment. J Pharmacol Exp Ther. 369:291–299. 2019. View Article : Google Scholar : PubMed/NCBI

48 

US Food and Drug Administration. Label, . simplehttps://www.accessdata.fda.gov/drugsatfda_docs/label/2020/204114s014lbl.pdfFeb 2–2020

49 

European Medicines Agency. Product information, . simplehttps://www.ema.europa.eu/en/documents/product-information/mekinist-epar-product-information_en.pdfFeb 2–2020

50 

Ouellet D, Kassir N, Chiu J, Mouksassi MS, Leonowens C, Cox D, DeMarini DJ, Gardner O, Crist W and Patel K: Population pharmacokinetics and exposure-response of trametinib, a MEK inhibitor, in patients with BRAF V600 mutation-positive melanoma. Cancer Chemother Pharmacol. 77:807–817. 2016. View Article : Google Scholar : PubMed/NCBI

51 

US Food and Drug Administration. Label, . simplehttps://www.accessdata.fda.gov/drugsatfda_docs/label/2018/022405s014lbl.pdfFeb 2–2020

52 

European Medicines Agency. Product information, . simplehttps://www.ema.europa.eu/en/documents/product-information/caprelsa-epar-product-information_en.pdfFeb 2–2020

53 

Weil A, Martin P, Smith R, Oliver S, Langmuir P, Read J and Molz KH: Pharmacokinetics of vandetanib in subjects with renal or hepatic impairment. Clin Pharmacokinet. 49:607–618. 2010. View Article : Google Scholar : PubMed/NCBI

54 

US Food and Drug Administration. Label, . simplehttps://www.accessdata.fda.gov/drugsatfda_docs/label/2017/202429s016lbl.pdfFeb 2–2020

55 

European Medicines Agency. Product information, . simplehttps://www.ema.europa.eu/en/documents/product-information/zelboraf-epar-product-information_en.pdfFeb 2–2020

56 

Zhang W, Heinzmann D and Grippo JF: Clinical pharmacokinetics of vemurafenib. Clin Pharmacokinet. 56:1033–1043. 2017. View Article : Google Scholar : PubMed/NCBI

57 

Field KM and Michael M: Part II: Liver function in oncology: Towards safer chemotherapy use. Lancet Oncol. 9:1181–1190. 2008. View Article : Google Scholar : PubMed/NCBI

58 

Krens SD, Lassche G, Jansman FG, Desar IM, Lankheet NA, Burger DM, van Herpen CM and van Erp NP: Dose recommendations for anticancer drugs in patients with renal or hepatic impairment. Lancet Oncol. 20:e200–e207. 2019. View Article : Google Scholar : PubMed/NCBI

59 

Pond SM and Tozer TN: First-pass elimination: Basic concepts and clinical consequences. Clin Pharmacokinet. 9:1–25. 1984. View Article : Google Scholar : PubMed/NCBI

60 

Chtioui H and Buclin T: Pharmacokinetics in hepatic impairment: Mind the protein binding. J Hepatol. 63:1539–1540. 2015. View Article : Google Scholar : PubMed/NCBI

61 

Villeneuve JP and Pichette V: Cytochrome P450 and liver diseases. Curr Drug Metab. 5:273–282. 2004. View Article : Google Scholar : PubMed/NCBI

62 

Zollner G, Fickert P, Silbert D, Fuchsbichler A, Marschall HU, Zatloukal K, Denk H and Trauner M: Adaptive changes in hepatobiliary transporter expression in primary biliary cirrhosis. J Hepatol. 38:717–727. 2003. View Article : Google Scholar : PubMed/NCBI

63 

Proulx NL, Akbari A, Garg AX, Rostom A, Jaffey J and Clark HD: Measured creatinine clearance from timed urine collections substantially overestimates glomerular filtration rate in patients with liver cirrhosis: A Systematic review and individual patient Meta-analysis. Nephrol Dial Transplant. 20:1617–1622. 2005. View Article : Google Scholar : PubMed/NCBI

64 

European Medicines Agency, . Guideline on the evalution of the pharmacokinetics of medicinal products in patients with impaired hepatic function. simplehttps://www.ema.europa.eu/documents/scientific-guideline/guideline-evaluation-pharmacokineticsmedicinal-products-patients-impaired-hepatic-function_en.pdfJan 20–2020

65 

US Food and Drug Administration, . Guidance for industry: Pharmacokinetics in patients with impaired hepatic function: Study design, data analysis, and impact on dosing and labeling. simplehttps://www.fda.gov/downloads/drugs/guidancecomplianceregulatoryinformation/guidances/ucm072123.pdfJan 20–2020

66 

Mansfield AS, Rudek MA, Vulih D, Smith GL, Harris PJ and Ivy SP; NCI Organ Dysfunction Working Group, : The effect of hepatic impairment on outcomes in phase I clinical trials in cancer subjects. Clin Cancer Res. 22:5472–5479. 2016. View Article : Google Scholar : PubMed/NCBI

67 

Fujita K, Matsumoto N, Ishida H, Kubota Y, Iwai S, Shibanuma M and Kato Yo: Decreased disposition of anticancer drugs predominantly eliminated via the liver in patients with renal failure. Curr Drug Metab. 20:361–376. 2019. View Article : Google Scholar : PubMed/NCBI

68 

Kim AH, Yoon S, Lee Y, Lee J, Bae E, Lee H, Kim DK, Lee S, Yu KS, Jang IJ and Cho JY: Assessment of Hepatic Cytochrome P450 3A activity using metabolic markers in patients with renal impairment. J Korean Med Sci. 33:e2982018. View Article : Google Scholar : PubMed/NCBI

69 

Matzke GR, Dowling TC, Marks SA and Murphy JE: Influence of kidney disease on drug disposition: An assessment of industry studies submitted to the FDA for new chemical entities 1999-2010. J Clin Pharmacol. 56:390–398. 2016. View Article : Google Scholar : PubMed/NCBI

70 

Wiebe S, Schnell D, Külzer R, Gansser D, Weber A, Wallenstein G, Halabi A, Conrad A and Wind S: Influence of renal impairment on the pharmacokinetics of afatinib: An Open-Label, Single-Dose Study. Eur J Drug Metab Pharmacokinet. 42:461–469. 2017. View Article : Google Scholar : PubMed/NCBI

71 

Inker LA, Astor BC, Fox CH, Isakova T, Lash JP, Peralta CA, Kurella Tamura M and Feldman HI: KDOQI US commentary on the 2012 KDIGO clinical practice guideline for the evaluation and management of CKD. Am J Kidney Dis. 63:713–735. 2014. View Article : Google Scholar : PubMed/NCBI

72 

Shah RR and Shah DR: Safety and tolerability of epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors in oncology. Drug Saf. 42:181–198. 2019. View Article : Google Scholar : PubMed/NCBI

73 

Imai H, Kaira K, Naruse I, Hayashi H, Iihara H, Kita Y, Mizusaki N, Asao T, Itoh Y, Sugiyama T, et al: Successful afatinib treatment of advanced non-small-cell lung cancer patients undergoing hemodialysis. Cancer Chemother Pharmacol. 79:209–213. 2017. View Article : Google Scholar : PubMed/NCBI

74 

Bersanelli M, Tiseo M, Artioli F, Lucchi L and Ardizzoni A: Gefitinib and Afatinib treatment in an advanced Non-small cell lung cancer (NSCLC) Patient Undergoing Hemodialysis. Anticancer Res. 34:3185–3188. 2014.PubMed/NCBI

75 

Suzuki S, Haratani K, Takahama T, Watanabe S, Takegawa N, Hayashi H, Takeda M, Yonesaka K and Nakagawa K: Safety and Efficacy of alectinib in a patient with advanced NSCLC Undergoing Hemodialysis. J Thorac Oncol. 14:e50–e52. 2019. View Article : Google Scholar : PubMed/NCBI

76 

Hong Y, Passos VQ, Huang PH and Lau YY: Population pharmacokinetics of ceritinib in adult patients with tumors characterized by genetic abnormalities in anaplastic lymphoma kinase. J Clin Pharmacol. 57:652–662. 2017. View Article : Google Scholar : PubMed/NCBI

77 

Tan W, Yamazaki S, Johnson TR, Wang R, O'Gorman MT, Kirkovsky L, Boutros T, Brega NM and Bello A: Effects of renal function on crizotinib pharmacokinetics: Dose recommendations for patients with ALK-Positive Non-Small cell lung cancer. Clin Drug Investig. 37:363–373. 2017. View Article : Google Scholar : PubMed/NCBI

78 

Wang E, Nickens DJ, Bello A, Khosravan R, Amantea M, Wilner KD, Parivar K and Tan W: Clinical implications of the pharmacokinetics of crizotinib in populations of patients with non-small cell lung cancer. Clin Cancer Res. 22:5722–5728. 2016. View Article : Google Scholar : PubMed/NCBI

79 

Kothari S, Ud-Din N, Lisi M and Coyle T: Crizotinib in anaplastic lymphoma kinasepositive anaplastic large cell lymphoma in the setting of renal insufficiency: A case report. J Med Case Rep. 10:1762016. View Article : Google Scholar : PubMed/NCBI

80 

Song SH, Ryu JW, Jwa HY, Ha CW, Kim H, Jo JM and Han SH: ALK-positive lung cancer diagnosed with abdominal lymph nodes in a patient receiving hemodialysis. Thorac Cancer. 10:2188–2191. 2019. View Article : Google Scholar : PubMed/NCBI

81 

Park JJ, Boddy AV, Liu X, Harris D, Lee V, Kefford RF and Carlino MS: Pharmacokinetics of dabrafenib in a patient with metastatic melanoma undergoing haemodialysis. Pigment Cell Melanoma Res. 30:68–71. 2017. View Article : Google Scholar : PubMed/NCBI

82 

Togashi Y, Masago K, Fukudo M, Terada T, Ikemi Y, Kim YH, Fujita S, Irisa K, Sakamori Y, Mio T, et al: Pharmacokinetics of Erlotinib and its active metabolite OSI-420 in patients with non-small cell lung cancer and chronic renal failure who are undergoing hemodialysis. J Thorac Oncol. 5:601–605. 2010. View Article : Google Scholar : PubMed/NCBI

83 

Gridelli C, Maione P, Galetta D and Rossi A: Safety profile of Erlotinib in patients with advanced non-small cell lung cancer with chronic renal failure. J Thorac Oncol. 2:96–98. 2007. View Article : Google Scholar : PubMed/NCBI

84 

Rossi A, Maione P, Del Gaizo F, Guerriero C, Castaldo V and Gridelli C: Safety profile of gefitinib in advanced non-small cell lung cancer elderly patients with chronic renal failure: Two clinical cases. Lung Cancer. 47:421–423. 2005. View Article : Google Scholar : PubMed/NCBI

85 

Yamaguchi T, Isogai S, Okamura T, Uozu S, Mieno Y, Hoshino T, Goto Y, Hayashi M, Nakanishi T and Imaizumi K: Pharmacokinetics of Gefitinib in a patient with non-small cell lung cancer undergoing continuous ambulatory peritoneal dialysis. Case Rep Oncol. 8:78–82. 2015. View Article : Google Scholar : PubMed/NCBI

86 

Luo J, Ni L, Wang M, Zhong W, Xiao Y, Zheng K and Hu P: Pharmacokinetic analysis of gefitinib in a patient with advanced non-small cell lung cancer undergoing hemodialysis. Thorac Cancer. 7:251–253. 2016. View Article : Google Scholar : PubMed/NCBI

87 

Shinagawa N, Yamazaki K, Asahina H, Agata J, Itoh T and Nishimura M: Gefitinib administration in a patient with lung cancer undergoing hemodialysis. Lung Cancer. 58:422–424. 2007. View Article : Google Scholar : PubMed/NCBI

88 

Del Conte A, Minatel E, Schinella D, Baresic T, Basso SM and Lumachi F: Complete metabolic remission with gefitinib in a hemodialysis patient with bone metastases from non-small cell lung cancer. Anticancer Res. 34:319–322. 2014.PubMed/NCBI

89 

Yamada H, Satoh H, Hida N, Nakaizumi T, Terashima H and Hizawa N: Osimertinib for an older de novo T790M patient with chronic kidney disease. Geriatr Gerontol Int. 18:503–504. 2018. View Article : Google Scholar : PubMed/NCBI

90 

Tamura T, Takagi Y, Okubo H, Yamaguchi S, Kikkawa Y, Hashimoto I, Kaburagi T, Miura M, Satoh H and Hizawa N: Plasma concentration of osimertinib in a non-small cell lung cancer patient with chronic renal failure undergoing hemodialysis. Lung Cancer. 112:225–226. 2017. View Article : Google Scholar : PubMed/NCBI

91 

Iwafuchi Y, Saito I and Narita I: Efficacy and safety of osimertinib in a hemodialysis patient with advanced non-small cell lung cancer. Ther Apher Dial. 21:416–417. 2017. View Article : Google Scholar : PubMed/NCBI

92 

Matsunashi A, Fujimoto D, Hosoya K, Irie K, Fukushima S and Tomii K: Osimertinib in a patient with non-small cell lung cancer and renal failure undergoing hemodialysis: a case report. Invest New Drugs. 38:1192–1195. 2020. View Article : Google Scholar : PubMed/NCBI

93 

Iddawela M, Crook S, George L, Lakkaraju A, Nanayakkara N, Hunt R and Adam WR: Safety and efficacy of Vemurafenib in end stage renal failure. BMC Cancer. 13:5812013. View Article : Google Scholar : PubMed/NCBI

94 

Porta C, Cosmai L, Gallieni M, Pedrazzoli P and Malberti F: Renal effects of targeted anticancer therapies. Nat Rev Nephrol. 11:354–370. 2015. View Article : Google Scholar : PubMed/NCBI

95 

Qian J, Zhang X, Zhang B, Yan B, Wang L, Gu P, Wang W, Wang H and Han B: Tyrosine kinase inhibitor-related hepatotoxicity in patients with advanced lung adenocarcinoma: A real-world retrospective study. Cancer Manag Res. 12:3293–3299. 2020. View Article : Google Scholar : PubMed/NCBI

96 

Jhaveri KD, Wanchoo R, Sakhiya V, Ross DW and Fishbane SL: Adverse renal effects of novel molecular oncologic targeted therapies: A narrative review. Kidney Int Rep. 2:108–123. 2016. View Article : Google Scholar : PubMed/NCBI

97 

González J, Quiroga M, Escudero-Vilaplana V, Collado-Borrell R, Herranz-Alonso A and Sanjurjo Sáez M: Posology adjustments of oral antineoplastic agents for special populations: Patients with renal impairment, hepatic impairment and hematologic toxicities. Expert Opin Drug Saf. 17:553–572. 2018. View Article : Google Scholar : PubMed/NCBI

98 

Park K, Tan EH, O'Byrne K, Zhang L, Boyer M, Mok T, Hirsh V, Yang JC, Lee KH, Lu S, et al: Afatinib versus gefitinib as first-line treatment of patients with EGFR mutation-positive non-small-cell lung cancer (LUX-Lung 7): A phase 2B, open-label, randomised controlled trial. Lancet Oncol. 17:577–589. 2016. View Article : Google Scholar : PubMed/NCBI

99 

Camidge DR, Dziadziuszko R, Peters S, Mok T, Noe J, Nowicka M, Gadgeel SM, Cheema P, Pavlakis N, de Marinis F, et al: Updated efficacy and safety data and impact of the EML4-ALK fusion variant on the efficacy of Alectinib in untreated Alk-positive advanced non-small cell lung cancer in the global phase III ALEX Study. J Thorac Oncol. 14:1233–1243. 2019. View Article : Google Scholar : PubMed/NCBI

100 

Zhu V and Ou SH: Safety of alectinib for the treatment of metastatic ALK-rearranged non-small cell lung cancer. Expert Opin Drug Saf. 16:509–514. 2017. View Article : Google Scholar : PubMed/NCBI

101 

Camidge DR, Kim HR, Ahn MJ, Yang JC, Han JY, Lee JS, Hochmair MJ, Li JY, Chang GC, Lee KH, et al: Brigatinib versus Crizotinib in ALK-positive Non-small-cell lung cancer. N Engl J Med. 379:2027–2039. 2018. View Article : Google Scholar : PubMed/NCBI

102 

Choueiri TK, Halabi S, Sanford BL, Hahn O, Michaelson MD, Walsh MK, Feldman DR, Olencki T, Picus J, Small EJ, et al: Cabozantinib versus Sunitinib as initial targeted therapy for patients with metastatic renal cell carcinoma of poor or intermediate risk: The alliance A031203 CABOSUN trial. J Clin Oncol. 35:591–597. 2017. View Article : Google Scholar : PubMed/NCBI

103 

Kim DW, Mehra R, Tan DSW, Felip E, Chow LQM, Camidge DR, Vansteenkiste J, Sharma S, De Pas T, Riely GJ, et al: Intracranial and whole-body response of ceritinib in ALK inhibitor-naïve and previously ALK inhibitor-treated patients with ALK-rearranged non-small-cell lung cancer (NSCLC): Updated results from the phase 1, multicentre, open-label ASCEND-1 trial. Lancet Oncol. 17:452–463. 2016. View Article : Google Scholar : PubMed/NCBI

104 

Knispel S, Zimmer L, Kanaki T, Ugurel S, Schadendorf D and Livingstone E: The safety and efficacy of dabrafenib and trametinib for the treatment of melanoma. Expert Opin Drug Saf. 17:73–87. 2018. View Article : Google Scholar : PubMed/NCBI

105 

Planchard D, Smit EF, Groen HJM, Mazieres J, Besse B, Helland Å, Giannone V, D'Amelio AM Jr, Zhang P, Mookerjee B and Johnson BE: Dabrafenib plus trametinib in patients with previously untreated BRAFV600E-mutant metastatic non-small-cell lung cancer: An open-label, phase 2 trial. Lancet Oncol. 18:1307–1316. 2017. View Article : Google Scholar : PubMed/NCBI

106 

Wu YL, Cheng Y, Zhou X, Lee KH, Nakagawa K, Niho S, Tsuji F, Linke R, Rosell R, Corral J, et al: Dacomitinib versus gefitinib as first-line treatment for patients with EGFR-mutation-positive non-small-cell lung cancer (ARCHER 1050): A randomised, open-label, phase 3 trial. Lancet Oncol. 18:1454–1466. 2017. View Article : Google Scholar : PubMed/NCBI

107 

Doebele RC, Drilon A, Paz-Ares L, Siena S, Shaw AT, Farago AF, Blakely CM, Seto T, Cho BC, Tosi D, et al: Entrectinib in patients with advanced or metastatic NTRK fusionpositive solid tumours: Integrated analysis of three phase 1-2 trials. Lancet Oncol. 21:271–282. 2020. View Article : Google Scholar : PubMed/NCBI

108 

Choi HD and Chang MJ: Eye, hepatobiliary, and renal disorders of erlotinib in patients with non-small-cell lung cancer: A meta-analysis. PLoS One. 15:e02348182020. View Article : Google Scholar : PubMed/NCBI

109 

Schacher-Kaufmann S and Pless M: Acute fatal liver toxicity under Erlotinib. Case Rep Oncol. 3:182–188. 2010. View Article : Google Scholar : PubMed/NCBI

110 

Hsiue EH, Lee JH, Lin CC and Yang JC: Safety of gefitinib in non-small cell lung cancer treatment. Expert Opin Drug Saf. 15:993–1000. 2016. View Article : Google Scholar : PubMed/NCBI

111 

Laetsch TW, DuBois SG, Mascarenhas L, Turpin B, Federman N, Albert CM, Nagasubramanian R, Davis JL, Rudzinski E, Feraco AM, et al: Larotrectinib for paediatric solid tumours harbouring NTRK gene fusions: Phase 1 results from a multicentre, open-label, phase 1/2 study. Lancet Oncol. 19:705–714. 2018. View Article : Google Scholar : PubMed/NCBI

112 

Shaw AT, Solomon BJ, Chiari R, Riely GJ, Besse B, Soo RA, Kao S, Lin CC, Bauer TM, Clancy JS, et al: Lorlatinib in advanced ROS1-positive non-small-cell lung cancer: A multicentre, open-label, single-arm, phase 1-2 trial. Lancet Oncol. 20:1691–1701. 2019. View Article : Google Scholar : PubMed/NCBI

113 

Cao Y, Qiu X, Xiao G, Hu H and Lin T: Effectiveness and safety of osimertinib in patients with metastatic EGFR T790M-positive NSCLC: An observational real-world study. PLoS One. 14:e02215752019. View Article : Google Scholar : PubMed/NCBI

114 

Tsang VH, Robinson BG and Learoyd DL: The safety of vandetanib for the treatment of thyroid cancer. Expert Opin Drug Saf. 15:1107–1113. 2016. View Article : Google Scholar : PubMed/NCBI

115 

Brose MS, Cabanillas ME, Cohen EE, Wirth LJ, Riehl T, Yue H, Sherman SI and Sherman EJ: Vemurafenib in patients with BRAF(V600E)-positive metastatic or unresectable papillary thyroid cancer refractory to radioactive iodine: A non-randomised, multicentre, open-label, phase 2 trial. Lancet Onco. 17:1272–1282. 2016. View Article : Google Scholar

116 

Borella E, Poggesi I and Magni P: Prediction of the effect of renal impairment on the pharmacokinetics of new drugs. Clin Pharmacokinet. 57:505–514. 2018. View Article : Google Scholar : PubMed/NCBI

117 

Hsueh CH, Hsu V, Zhao P, Zhang L, Giacomini KM and Huang SM: PBPK modeling of the effect of reduced kidney function on the pharmacokinetics of drugs excreted renally by organic anion transporters. Clin Pharmacol Ther. 103:485–492. 2018. View Article : Google Scholar : PubMed/NCBI

118 

Solassol I, Pinguet F and Quantin X: FDA- and EMA-approved tyrosine kinase inhibitors in advanced EGFR-mutated non-small cell lung cancer: Safety, tolerability, plasma concentration monitoring, and management. Biomolecules. 9:6682019. View Article : Google Scholar

Related Articles

Journal Cover

February-2021
Volume 45 Issue 2

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zhao D, Chen J, Long X and Wang J: Dose adjustment for tyrosine kinase inhibitors in non‑small cell lung cancer patients with hepatic or renal function impairment (Review). Oncol Rep 45: 413-426, 2021
APA
Zhao, D., Chen, J., Long, X., & Wang, J. (2021). Dose adjustment for tyrosine kinase inhibitors in non‑small cell lung cancer patients with hepatic or renal function impairment (Review). Oncology Reports, 45, 413-426. https://doi.org/10.3892/or.2020.7870
MLA
Zhao, D., Chen, J., Long, X., Wang, J."Dose adjustment for tyrosine kinase inhibitors in non‑small cell lung cancer patients with hepatic or renal function impairment (Review)". Oncology Reports 45.2 (2021): 413-426.
Chicago
Zhao, D., Chen, J., Long, X., Wang, J."Dose adjustment for tyrosine kinase inhibitors in non‑small cell lung cancer patients with hepatic or renal function impairment (Review)". Oncology Reports 45, no. 2 (2021): 413-426. https://doi.org/10.3892/or.2020.7870