Skip to main content
British Journal of Clinical Pharmacology logoLink to British Journal of Clinical Pharmacology
. 2015 Jan 20;79(2):241–253. doi: 10.1111/bcp.12496

Metabolism-related pharmacokinetic drug−drug interactions with tyrosine kinase inhibitors: current understanding, challenges and recommendations

Yi Ling Teo 1, Han Kiat Ho 1, Alexandre Chan 1,2,
PMCID: PMC4309630  PMID: 25125025

Abstract

Drug−drug interactions (DDIs) occur when a patient's response to the drug is modified by administration or co-exposure to another drug. The main cytochrome P450 (CYP) enzyme, CYP3A4, is implicated in the metabolism of almost all of the tyrosine kinase inhibitors (TKIs). Therefore, there is a substantial potential for interaction between TKIs and other drugs that modulate the activity of this metabolic pathway. Cancer patients are susceptible to DDIs as they receive many medications, either for supportive care or for treatment of toxicity. Differences in DDI outcomes are generally negligible because of the wide therapeutic window of common drugs. However for anticancer agents, serious clinical consequences may occur from small changes in drug metabolism and pharmacokinetics. Therefore, the objective of this review is to highlight the current understanding of DDIs among TKIs, with a focus on metabolism, as well as to identify challenges in the prediction of DDIs and provide recommendations.

Keywords: cytochrome P450, drug−drug interactions, drug metabolism, pharmacokinetics, tyrosine kinase inhibitors

Introduction

Tyrosine kinases are a major family of proteins frequently dysregulated (either through somatic mutations or overexpression) in various cancers. Their critical role in the control of cancer phenotypes, coupled to the presence of suitable binding domains for small molecules, led to the development of many tyrosine kinase inhibitors (TKIs) as anti-cancer agents. As we are able to achieve a better control of the disease over the longer lifespan of a patient, these TKIs are now being considered as chronic medications as they are used over a long period of time. The increasing number of therapies has improved the prognosis for the disease but has augmented the challenge of evaluating patients for potential drug interactions during therapy 1.

Drug−drug interactions (DDIs) occur when a patient's pharmacological or clinical response to the drug is modified by administration or co-exposure to another drug. Pharmacokinetic interactions occur when one drug influences the pharmacokinetic processes such as absorption, distribution, metabolism and excretion, of another drug. Altered metabolism is among the most complex of these processes by which drug–drug interactions can occur, and induction or inhibition of hepatic enzymes by drugs are often implicated. The clinical consequences of enzyme induction or inhibition depend on the pharmacological and toxic effect of both the parent drug and its metabolite(s). For example, if the parent compound is more active than its metabolite, inhibition of metabolism increases the exposure to the drug and also its therapeutic and/or toxic effects. However, if the parent compound is a pro-drug, inhibition of metabolism may result in a decrease in therapeutic efficacy. More recently, another paradigm of interaction arises when the metabolite is more toxic, and hence induction of metabolism down this pathway can exacerbate toxicity.

Central to the metabolism of drugs is the cytochrome P450 (CYP) family of enzymes. This consists of numerous enzymes that are responsible for the phase I metabolism of many drugs, nutrients, endogenous substances, and environmental toxins. The main CYP enzyme, CYP3A4, is responsible for the metabolism of more than 50% of all drugs in the market. It is also implicated in the metabolism of almost all of the TKIs. Therefore, there is a substantial potential for interaction between TKIs and other drugs that modulate the activity of this metabolic pathway.

A recent study revealed that co-prescription of drugs that induce or inhibit metabolic pathways used by TKIs was high. Overall co-prescribing rates for DDI drugs that may decrease TKIs effectiveness ranged from 23–57%, while co-prescribing rates with drugs that may increase TKI toxicity ranged from 24–74% 2. In another study which studied the pattern of DDIs in cancer patients, the frequency of at least one potential DDI occurring was 63%. Among them, almost 62% of the identified DDIs were considered as major, where the effects of the interaction may result in serious consequences such as hospitalization, therapeutic failure, permanent injury or even death 3. Although many studies highlighted the problem of frequent DDIs among TKIs, these studies did not address the clinical consequences of the potential DDIs, such as increased toxicity or therapeutic failure. In some cases, these combinations could have been intentionally prescribed, where physicians may have knowingly prescribed a potentially interacting combination because they considered the potential benefits to outweigh the risks or because the patient had tolerated the combination in the past 4. As these TKIs are relatively new to the market, the scientific evidence that supports their DDI is limited. Therefore, it is not unexpected to observe that oncology professionals are unable to identify TKI DDI pairs which that might have a high probability of causing deleterious effects in cancer patients 5.

Cancer patients are susceptible to DDIs as they receive many medications, either for supportive care or for treatment of therapy-induced toxicity 6. For instance, an observational study highlighted that patients were receiving on average 6.8 drugs in addition to sunitinib. Among them, antihypertensive drugs were most commonly prescribed, followed by analgesics, anti-emetics and thyroid substitution therapy 7. In certain cases, a cancer patient's pharmacokinetic parameters may be also altered, for example, oedema affecting volume of distribution or impaired drug absorption due to malnutrition or mucositis. These issues may also affect the consequences of DDIs. Since most cancers typically occur at a later age, these patients may also be receiving other drugs for the management of their comorbidities. Differences in DDI outcomes are generally negligible because of the wide therapeutic windows of common drugs. However, in cancer chemotherapy with anti-cancer drugs, serious clinical consequences may occur from small changes in drug metabolism and pharmacokinetics 8. Studies conducted by our group have exhibited the increase in risk of toxicity as a consequence of DDIs 9. Sunitinib, a multi-kinase inhibitor used in the first line treatment of metastatic renal cell carcinoma, has been associated with dose-limiting dermatological toxicities such as hand-foot skin reaction (HFSR), rash and dry skin 10. Sunitinib undergoes metabolism by CYP3A4 to form an active metabolite, SU12662 (N-desethyl sunitinib). A recent study has demonstrated that sunitinib is more dermatotoxic than SU12662, suggesting that patients who receive concomitant CYP3A4 inhibitors would be at a higher risk of dermatological toxicities, due to a lower ability to metabolize sunitinib 11. Consequently, this may result in an increased risk of non-compliance, dose reduction or therapy discontinuation, thereby leading to suboptimal therapy.

Due to the substantial potential for interaction between TKIs and other drugs that modulate the activity of metabolic pathways, unwanted clinical consequences may occur from small changes in drug metabolism and pharmacokinetics in cancer patients. Furthermore, it is a challenge to determine the clinical effects of the DDI due to the large inter-patient variability in the pharmacokinetics of the TKIs. Therefore, the objective of this review is to highlight the current understanding of DDIs among TKIs, with a specific focus on DDIs involving metabolism, and to identify challenges in the prediction of DDIs and provide some possible recommendations.

Methods

A search was conducted to identify all small molecule TKIs approved by the FDA from January 2000 to February 2014. A comprehensive literature search of articles involving TKIs was performed using the PubMed and Scopus databases. Meetings abstracts presented at the American Association for Cancer Research (AACR) and the American Society of Clinical Oncology (ASCO) were also reviewed. The search was conducted by using the generic names of all the identified TKIs (afatinib, axitinib, bosutinib, cabozantinib, crizotinib, dasatinib, erlotinib, gefitinib, imatinib, lapatinib, nilotinib, pazopanib, ponatinib, regorafenib, sorafenib, sunitinib, vandetanib), and terms such as ‘drug interaction’, ‘metabolism’ and ‘pharmacokinetics’. The search was limited to English language articles published between January 1995 and February 2014.

Results and discussion

Overview of TKIs (Table 1)

Table 1.

Overview of FDA-approved TKIs

Year of FDA approval Indication(s) Targets FDA black box warning Dosing administration Reference
Afatinib (Gilotrif) 2013 Metastatic NSCLC with EGFR mutations EGFR, HER2, HER4 40 mg once daily 52
Axitinib (Inlyta) 2012 Advanced RCC VEGFR-1, VEGFR-2, and VEGFR-3 5 mg twice daily 53
Bosutinib (Bosulif) 2012 CML Bcr-Abl, Src 500 mg once daily 16
Cabozantinib (Cometriq) 2012 Thyroid cancer RET, MET, VEGFR-1, −2 and −3, KIT, TRKB, FLT-3, AXL, and TIE-2 Haemorrhage 140 mg once daily 54
Crizotinib (Xalkori) 2011 ALK+ NSCLC ALK, MET, RON 250 mg twice daily 55
Dasatinib (Sprycel) 2006 CML Ph+ ALL Bcr-Abl, Src 100 mg once daily 56
Erlotinib (Tarceva) 2004 NSCLC Metastatic pancreatic cancer EGFR 100–150 mg once daily 57
Gefitinib (Iressa) 2003 NSCLC EGFR 250 mg once daily 58
Imatinib (Gleevec) 2001 CML GIST Ph+ ALL Bcr-Abl 300–800 mg once daily* 59
Lapatinib (Tykerb) 2007 Metastatic breast cancer EGFR, HER2 Hepatotoxicity 1250–1500 mg once daily* 60
Nilotinib (Tasigna) 2007 CML Bcr-Abl QT prolongation 300 mg twice daily 61
Pazopanib (Votrient) 2009 RCC Soft tissue sarcoma VEGFR-1, VEGFR-2 and VEGFR-3 Hepatotoxicity 800 mg once daily 62
Ponatinib (Iclusig) 2012 CML Ph+ ALL Bcr-Abl Arterial thrombosis and hepatotoxicity 45 mg once daily 63
Regorafenib (Stivarga) 2012 Metastatic colorectal cancer GIST VEGFR2 and TIE2 Hepatotoxicity 160 mg once daily 64
Sorafenib (Nexavar) 2005 RCC Unresectable HCC KIT, FLT-3, VEGFR-2, VEGFR-3 and PDGFR-B 400 mg twice daily 65
Sunitinib (Sutent) 2006 RCC GIST pNET PDGFR (α,β) VEGFR (1, 2, 3), KIT, FLT3, CSF-1R, RET Hepatotoxicity 37.5–50 mg once daily* 66
Vandetanib (Caprelsa) 2011 Thyroid Cancer EGFR, VEGFR, RET QT prolongation 800 mg once daily 67
*

Dosing administration depends on indication. ALK+, anaplastic lymphoma kinase; ALL, acute lymphoblastic leukemia; CML, chronic myeloid leukemia; FDA, Food and Drug Administration; GIST, gastrointestinal stromal tumour; HCC, hepatocellular carcinoma; NSCLC, non-small-cell lung cancer; Ph+ ALL, Philadelphia chromosome-positive acute lymphoid leukemia; pNET, progressive, well-differentiated pancreatic neuroendocrine u; RCC, renal cell carcinoma.

There are currently 17 FDA-approved TKIs (as of 31 March 2014) and they are indicated for various malignancies ranging from solid tumours such as breast and lung cancers to haematological malignancies like chronic myeloid leukemia.

Metabolism profile of TKIs (Table 2)

Table 2.

Metabolism profile of FDA-approved TKIs

% of dose recovered (% recovered unchanged) Metabolism
Feces Urine Major CYPs Minor CYPs & others Induces Inhibits
Afatinib 85 (NR) 4 (NR) Negligible
Axitinib 41 (12) 23 (ND) CYP3A4 CYP1A2 CYP1A2
CYP3A5 CYP2C19 CYP2C8
UGT1A1
Bosutinib 91 (NR) 3 (NR) CYP3A4
Cabozantinib 54 (NR) 27 (NR) CYP3A4 CYP2C9 CYP1A1 CYP2C8
CYP2C9
CYP2C19
CYP3A4
Crizotinib 63 (53) 22 (2) CYP3A4 CYP3A
CYP3A5 CYP2B6
Dasatinib 85 (19) 4 (<1) CYP3A4 FMO-3 CYP3A4
UGT
Erlotinib 83 (1) 8 (<1) CYP3A4 CYP1A2 CYP1A1
CYP1A1 CYP3A4
CYP2C8
Gefitinib 86 (NR) 4 (NR) CYP3A4 CYP2C19
CYP2D6 CYP2D6
Imatinib 68 (20) 13 (5) CYP3A4 CYP1A2 CYP2C8
CYP2D6 CYP2C9
CYP2C9 CYP3A4/5
CYP2C19 CYP2D6
Lapatinib (27) (<2) CYP3A4 CYP2C19 CYP3A
CYP3A5 CYP2C8 CYP2C8
Nilotinib 93 (69) N.R. CYP3A4 CYP2C8 CYP2B6 CYP3A4
CYP2C8 CYP2C8
CYP2C9 CYP2C9
CYP2D6
Pazopanib Majority in faeces 4 CYP3A4 CYP1A2 CYP1A2
CYP2C8 CYP3A4
CYP2B6
CYP2C8
CYP2C9
CYP2C19
CYP2D6
CYP2E1
Ponatinib 87 (NR) 5 (NR) CYP3A4 CYP2C8
CYP2D6
CYP3A5
Regorafenib 71 (47) 19 (2) CYP3A4 UGT1A9 CYP2C8
CYP2C9
CYP2B6
CYP3A4
CYP2C19
Sorafenib 77 (51) 19 (ND) CYP3A4 UGT1A9 CYP2B6
CYP2C8
CYP2C9
CYP2C19
CYP2D6
CYP3A4
Sunitinib 61 (NR) 16 (NR) CYP3A4
Vandetanib 44 (NR) 25 (NR) CYP3A4 FMO-1
FMO-3

All information was obtained from product information labels 68,69. ND, not detected; NR, not reported.

Almost all of the TKIs undergo metabolism by CYP enzymes. CYP3A4 is the CYP enzyme involved in the metabolism of the majority of the TKIs. Some of these TKIs, including imatinib, sunitinib and dasatinib, form an active metabolite upon metabolism. These TKIs can also act as an inducer or inhibitor to the CYP enzymes. All TKIs are primarily excreted in the faeces. However, the percentage of unchanged drug recovered in the faeces and urine varies widely between the TKIs. This could be likely due to the varying degree of absorption of the drug or due to differences in the extent of drug metabolism.

Potential effect of enzyme inducers/inhibitors on the pharmacokinetics of TKIs (Table 3)

Table 3.

Potential effect of enzyme inhibitors/inducers on the pharmacokinetics of TKIs

Changes in PK of TKI Recommendations Reference
Afatinib Unlikely Unlikely 52
Axitinib Ketoconazole: ↑ 1.5× Cmax, ↑ 2× AUC of axitinib Strong 3A4/5 inhibitors → Avoid; Consider alternative agents; Consider ↓ dose of axitinib by half 53,70,71
Rifampicin: ↓ 71% Cmax, ↓ 79% AUC of axitinib Strong 3A4/5 inducers → Avoid; Consider alternative agents
Bosutinib Ketoconazole: ↑ 5.2× Cmax, ↑ 8.6× AUC of bosutinib Strong 3A inhibitors → Avoid 13,16
Rifampicin: ↓ 86% Cmax, ↓ 94% AUC of bosutinib Strong 3A inducers → Avoid
Cabozantinib Ketoconazole: ↑ 38% AUC of cabozantinib Strong 3A4 inhibitors → Avoid; Consider ↓ daily dose of cabozantinib by 40 mg 54
Rifampicin: ↓ 77% AUC of cabozantinib Strong 3A4 inducers → Avoid; Consider ↑ daily dose of cabozantinib by 40 mg
Crizotinib Ketoconazole: ↑ 1.4× Cmax, ↑ 3.2× AUC of crizotinib Strong 3A4 inhibitors → Avoid 55
Rifampicin: ↓ 69% Cmax, ↓ 82% AUC of crizotinib Strong 3A4 inducers → Avoid
Dasatinib Ketoconazole: ↑ 4× Cmax, ↑ 5× AUC of dasatinib Strong 3A4 inhibitors → Avoid; Consider alternative agents; Consider ↓ dose of dasatinib to 20 mg daily (for patients taking 100 mg) or 40 mg daily (for patients taking 140 mg) 56,72
Rifampicin: ↓ 81% Cmax, ↓ 82% AUC of dasatinib Strong 3A4 inducers → Consider alternative agents; Consider ↑ dose of dasatinib
Erlotinib Ketoconazole: ↑ 67% AUC of erlotinib Ciprofloxacin: ↑ 17% Cmax, ↑ 39% AUC of erlotinib Strong 3A4 inhibitors → Use with caution 57
Rifampicin: ↓ 58% AUC of erlotinib Strong 3A4 inducers → Consider alternative agents; consider ↑ dose of erlotinib (up to maximum of 450 mg)
Gefitinib Itraconazole: ↑ 51% Cmax, ↑ 78% AUC of gefitinib Strong 3A4 inhibitors → Use with caution 22,58,73
Rifampicin: ↓ 65% Cmax, ↓ 83% AUC of gefitinib Phenytoin: ↓ 26% Cmax, ↓ 47% AUC of gefitinib Strong 3A4 inducers → Consider ↑ dose of gefitinib to 500 mg daily
Imatinib Ketoconazole: ↑ 26% Cmax, ↑ 40% AUC of imatinib Gemfibrozil*: ↓ 56% Cmax, ↓ 48% AUC of N-desmethylimatinib Strong 3A4 inhibitors → Use with caution 12,59,7476
Rifampicin: ↓ 54% Cmax, ↓ 74% AUC of imatinib EIAEDs: ↓ 68% Ctrough of imatinib Strong 3A4 inducers → Consider alternative agents
Lapatinib Ketoconazole: ↑ 114% Cmax, ↑ 257% AUC of lapatinib Strong 3A4 inhibitor → Avoid; Consider ↓ dose of lapatinib to 500 mg daily 60,77
Carbamazepine: ↓ 59% Cmax, ↓ 72% AUC of lapatinib Strong 3A4 inducers → Avoid; Consider ↑ dose of lapatinib up to 4500 mg daily (for HER2+ metastatic breast cancer) or 5500 mg daily (for HR+, HER2+ breast cancer)
Nilotinib Ketoconazole: ↑ 1.8× Cmax, ↑ 3× AUC of nilotinib Strong 3A4 inhibitors → Avoid; Consider ↓ dose of nilotinib to 300 mg daily (in resistant or intolerant Ph+ CML) or 200 mg daily (newly diagnosed Ph+ CML-CP) 61,78
Rifampicin: ↓ 64% Cmax, ↓ 80% AUC of nilotinib Strong 3A4 inducers → Avoid; Consider alternative agents
Pazopanib Ketoconazole: ↑ 45% Cmax, ↑ 66% AUC of pazopanib Strong 3A4 inhibitors → Avoid; Consider ↓ dose of pazopanib to 400 mg 62,79
Strong 3A4 inducers → Pazopanib should not be used
Ponatinib Ketoconazole: ↑ 47% Cmax, ↑ 78% AUC of ponatinib Strong 3A4 inhibitors → Consider ↓ dose of ponatinib to 30 mg daily 63,80
Regorafenib Ketoconazole: ↑ AUC of regorafenib Strong 3A inhibitors → Avoid 64
Rifampicin: ↓ AUC of regorafenib Strong 3A4 inducers → Avoid
Sorafenib Ketoconazole: no change in AUC of sorafenib 65
Rifampicin: ↓ 37% AUC of sorafenib Strong 3A4 inducers → Consider ↑ dose of sorafenib
Sunitinib Ketoconazole: ↑ 49% Cmax, ↑ 51% AUC of combined sunitinib and N-desethyl sunitinib Ritonavir: ↓ 48% Cmax, ↓ 40% AUC of N-desethyl sunitinib Strong 3A4 inhibitor → Consider alternative agents; Consider ↓ dose reduction of sunitinib to a minimum of 37.5 mg (GIST & RCC) or 25 mg (pNET) 66,81
Rifampicin: ↓ 23% Cmax, ↓ 50% AUC of combined sunitinib and N-desethyl sunitinib Efavirenz: ↑ 410% Cmax, ↑ 390% AUC of N-desethyl sunitinib Strong 3A4 inducers → Consider alternative agents; Consider ↑ dose of sunitinib to a maximum of 87.5 mg (GIST & RCC) or 62.5 mg (pNET)
Vandetanib Itraconazole: ↑ 9% AUC of vandetanib 15,67
Rifampicin: ↓ 40% AUC of vandetanib Strong 3A4 inducers → Avoid
*

Gemfibrozil inhibits the CYP2C8-mediated formation of N-desmethylimatinib (equipotent metabolite of parent imatinib). †Ritonavir inhibits the CYP3A4-mediated formation of N-desethyl sunitinib (equipotent metabolite of parent sunitinib). ‡Efavirenz induces the CYP3A4-mediated formation of N-desethyl sunitinib (equipotent metabolite of parent sunitinib). AUC, area under the curve; Cmax, maximum concentration; Ctrough, trough concentration; EIAEDs, enzyme-inducing anti-epileptic drugs; GIST, gastrointestinal stromal tumour; Ph+ CML, Philadelphia chromosome-positive chronic myeloid leukemia; Ph+ CML-CP, Philadelphia chromosome-positive chronic myeloid leukemia in chronic phase; PK, pharmacokinetics; pNET, progressive, well-differentiated pancreatic neuroendocrine tumours; RCC, renal cell carcinoma; TKI, tyrosine kinase inhibitor.

As most of these TKIs are substrates of the CYP3A4 enzyme, inducers and inhibitors of this enzyme can affect the exposure to these TKIs. The most common inducer and inhibitor used for the study of the potential pharmacokinetic interaction are rifampicin and ketoconazole, respectively. As expected, ketoconazole increases the exposure of TKIs due to the inhibition of metabolism. However, the extent to which the exposure is increased varied widely between the TKIs. For instance, concomitant ketoconazole can result in a slight increase of imatinib area under the curve (AUC) by 40% 12 but for bosutinib, the increase in AUC is more than eight times with concomitant ketoconazole 13. It is also interesting to note that exposure to TKIs is much increased by concomitant ketoconazole, as CYP3A4 is the only enzyme involved in the TKI's metabolic pathway. This is also supported by a report by Scripture et al. whereby drug interactions are likely to be significant when drug elimination occurs primarily through a single metabolic pathway 14. Similarly for rifampicin, the combination of drugs resulted in a decrease in exposure to the TKIs. However, the extent to which the exposure is decreased is not as large as that observed with ketoconazole. The decrease in TKI AUC ranged from 40% for vandetanib 15 to 94% for bosutinib 16. Among all the TKIs, sorafenib seems to be an exception, where studies have consistently demonstrated that clinically important interactions between sorafenib and drugs metabolized primarily by CYPs 3A4, 2C19, or 2D6 are not expected 17,18. Although co-administration of sorafenib with capecitabine has been shown to result in a mild increase in capecitabine exposure, these findings were not statistically significant and the mechanism of interaction is unclear 19. As strong CYP3A4 inducers and inhibitors generally cause a change in exposure, the concomitant use of such agents with TKIs is not recommended and should be avoided if possible. If such combinations must be used, manufacturers generally recommend that dose increase or decrease may be considered and that patients should be monitored closely following any changes in dosages.

Effect of TKIs as enzyme inducers/inhibitors on the pharmacokinetics of other drugs

While the effect of enzyme inhibitors or inducers (such as ketoconazole and rifampicin) on the pharmacokinetics of TKI has been extensively studied, the reciprocal effect of a TKI acting as an enzyme inducer or inhibitor has been comparatively less investigated. The ability for TKIs to increase or decrease plasma concentrations of non-anticancer drugs is mainly unclear, especially within in vivo conditions. Concomitant imatinib and simvastatin has resulted in a two-fold increase in simvastatin's maximum plasma concentraction (Cmax) and a three-fold increase in simvastatin AUC. This is likely due to inhibition by imatinib of the CYP3A4 enzyme, which is responsible for the metabolism of simvastatin to other metabolites. This also suggests that in the presence of imatinib, plasma concentrations of standard doses of drugs which are degraded by the CYP3A4 enzyme may also be increased. As such, caution is required when imatinib is administered with other CYP3A4 substrates with a narrow therapeutic window 20. Imatinib has also been shown to increase the exposure to metoprolol (23% increase in metoprolol AUC) when both agents are used together, due to the inhibition of the CYP2D6 enzyme by imatinib 21. Although the combination of gefitinib and metoprolol also resulted in an increased exposure to metoprolol (35% increase in metoprolol AUC), this change was not statistically significant. Despite this, gefitinib has a potential to increase plasma concentrations of CYP2D6 substrates and caution should be exercised when using CYP2D6 substrates that have a narrow therapeutic window 22. Concomitant use of two TKIs has also been investigated in certain cases. Clearance of erlotinib was markedly enhanced by sorafenib when the two agents were given concurrently, although the potential mechanism for this seeming interaction is not obvious 23. In another example, co-administration of lapatinib and pazopanib led to an increase in pazopanib exposure, and it has been suggested that this might be the result of inhibition of CYP3A4 and/or cellular transporters such as ABCB1 and ABCG2 by lapatinib 24. The effect of lapatinib on the clearance of vinorelbine has also been studied, where lapatinib resulted in a lower clearance of vinorelbine due to the inhibition of CYP3A4 25.

Applicability of in vitro and in vivo data within clinical practice

The majority of the available pharmacokinetic information results from in vitro data, preclinical animal studies or from small phase I studies which evaluated healthy volunteers who were administered a single dose of the drugs. Emerging methods include creating a simulator where in vivo clearances can be predicted from their in vitro data. For instance, the impact of co-administration of ketoconazole was simulated and the predicted two-fold increase in erlotinib exposure was found to be consistent with the results of a clinical study 26. However, in most cases, the prediction may not be entirely accurate, especially when most of these studies evaluate DDIs in the form of two interacting drugs, and these results may not be realistic where multiple drugs are used concurrently. In addition, several reasons have been proposed to highlight the inability of clinical interactions to be predicted accurately. Firstly, it is not always possible to determine the therapeutic concentration of a new drug and its metabolites in specific tissues. To complicate the issue further, the multiplicity of enzymes and transporters involved in the disposition of the said drugs and the intricacy of the pathways and interactions, in addition to overlapping substrate specificities of these proteins, results in complex and sometimes perplexing pharmacokinetic interactions with multidrug regimens. Large differences in genotype and expression level of each of these contributors can lead to a very complex influence on actual drug disposition. There can also be compensatory responses when one enzyme or transporter is inhibited, ‘cushioning’ any resulting change in metabolism. Each drug has a different level of dependence on intrinsic clearance for its overall clearance. Drugs with a high extraction ratio may be less sensitive to enzyme inhibition and induction, as their clearance is limited by blood flow rather than intrinsic activity. This makes it very challenging to test all of them in an in vitro system. Furthermore, the clinical significance of an interaction is unknown even if the in vitro or in vivo effect was established. Moreover, underlying disease states may influence the occurrence of an interaction that is unaccounted for by in vitro studies or by studies involving healthy volunteers alone 27,28. Endogenous CYP isoforms expressed in tumour cells also contributes to the metabolism of active drug, thereby playing a role in altering the half-life and kinetics of the administered TKI 29. In summary, it is complex and challenging to extrapolate these preliminary results to routine clinical practice, where TKIs are used to treat patients with cancer, many of whom are receiving multiple drugs and many of whom have impaired renal or hepatic function 30.

Formation of reactive intermediates/metabolites and implications for toxicity

Several TKIs such as dasatinib, erlotinib, gefitinib, imatinib, lapatinib, nilotinib, pazopanib, sorafenib and sunitinib undergo bioactivation to form reactive intermediates, which has implications in the generation of idiosyncratic adverse drug reactions (ADR) 31. One TKI whose metabolism and implications for toxicity has been extensively studied is lapatinib. Lapatinib has been shown to be extensively metabolized, as exemplified by diverse biotransformations to form metabolites. A number of the metabolites could potentially form reactive electrophilic intermediates that could contribute to hepatotoxicity 32. It is also worthy of note that the daily dose of these TKIs is high. For example the daily dose of lapatinib is more than 1000 mg. A high daily dose of more than 50 mg has been demonstrated to be a risk factor for ADRs 33; thereby setting 13 out of the 17 approved TKIs at risk. Due to the high dose, there would be high amounts of these reactive intermediates or metabolites generated, thus increasing the risk for toxicity 34. A recent study also further demonstrates that a dose more than 100 mg daily and being a substrate of CYP450 enzymes are two important predictors of drug-induced liver injury 35.

Actual drug−drug interaction cases involving TKIs as documented in the literature (Table 4)

Table 4.

Actual drug−drug interaction cases involving TKIs as documented in the literature

TKI Interacting drug Event Remarks Recommendations Reference
Gefitinib Anastrozole Liver toxicity Routinely monitor liver transaminases in all patients treated with gefitinib 82
Warfarin Coagulation abnormalities (Prothrombin time [PT] and international normalized ratio [INR] abnormalities) Gefitinib could inhibit the metabolism of warfarin, which is a substrate of CYP1A2, CYP2C9, and CYP3A4. The degree of the inhibitory effect of gefitinib on CYP enzymes varies from patient to patient. This may in part explain the variability of the PT-INR values observed on the coadministration of gefitinib and warfarin Close monitoring of PT-INR is recommended for patients receiving gefitinib and warfarin, especially during the first 2 weeks after the beginning of warfarin therapy. Appropriate adjustment of the warfarin dose should be done if an altered response to warfarin is observed. 83
Imatinib Voriconazole Severe pustular eruption Plasma concentrations of imatinib markedly elevated during simultaneous administration with voriconazole, possibly due to inhibition of imatinib metabolism by voriconazole Use of imatinib in association with CYP3A4 inhibitors has to be considered with caution. When such an association is considered, the monitoring of imatinib plasma levels in patients may be of help for identifying individuals with high imatinib concentrations who are at risk of developing toxicity, including skin lesions. 36
Amlodipine Peripheral neuropathy Amlodipine inhibits CYP3A4, which could increase imatinib concentrations Therapeutic monitoring of plasma imatinib levels may be useful to investigate unexpected imatinib toxicity. 84
Phenytoin AUC of imatinib was decreased by about 80%. After phenytoin was discontinued and the imatinib dose was increased to 500 mg daily, a complete haematological response was observed. 85
Levothyroxine Hypothyroidism Mechanism unclear Evaluate thyroid function in hypothyroid patients on tyrosine kinase inhibitors 86
Lapatinib Dexamethasone Hepatotoxicity Concomitant use may cause an increase in metabolizing capacity by dexamethasone, which in turn increases the formation of lapatinib-derived RM thereby elevating the risk of toxicity Clinicians should be aware of this risk when considering the use of this combination and follow through with close monitoring where necessary. 9
Pazopanib Simvastatin Transaminase elevations As pazopanib and statins are substrates for the same key metabolizing enzymes e.g. CYP3A4 and drug transporters, it is plausible that concomitant administration of pazopanib and statins may alter their systemic and/or hepatic exposures, leading to increased toxicities such as liver injury In addition to implementing the recommended dose modification guidelines for pazopanib, discontinuation of simvastatin should be considered to manage the risk of liver injury in cancer patients receiving both medications 37
Sorafenib Prednisolone Serum concentration of sorafenib was gradually increased following tapering of prednisolone, possibly due to prednisolone inducing sorafenib metabolism Therapeutic drug monitoring could be useful during sorafenib therapy in combination with prednisolone and for determining the optimal dosage of sorafenib. 87
Sunitinib Levothyroxine Hypothyroidism Mechanism unclear Evaluate thyroid function in hypothyroid patients on tyrosine kinase inhibitors 86

There have been several reports demonstrating actual DDI cases in clinical practice. The events reported were potentially fatal ones such as hepatotoxicity and anticoagulation abnormalities. The concomitant use of imatinib and voriconazole resulted in markedly elevated concentrations of imatinib (between 3500–4700 ng ml−1), than compared with when imatinib was used alone (2000 ng ml−1). The raised plasma concentrations resulted in severe pustular eruption in the patient, and this was deemed due to the inhibition of imatinib metabolism by voriconazole 36. Transaminase elevations have also been observed in drug pairs involving pazopanib and simvastatin. As both drugs are substrates of the same enzymes and transporters, it is plausible that the concomitant administration of pazopanib and statins may alter systemic and/or hepatic exposures, leading to increased toxicities such as liver injury 37. In a study implicating interaction between lapatinib and dexamethasone, both in vitro and clinical results points to the increased risk of hepatotoxicity when the combination of drugs was used together. It has been proposed that the concomitant use may cause an increase in the metabolism of lapatinib by dexamethasone, thereby increasing the formation of lapatinib-derived reactive metabolites, and leading to toxicity 9. However, this list of DDI cases is not exhaustive as there remains a possibility that many DDI cases remains unreported in the literature. Even if the drug pair is not listed in this table, it does not indicate that it is entirely safe for use. Healthcare professionals should still be aware of the potential interactions that may occur, and be vigilant in monitoring those patients who are receiving any potentially-interacting drug pairs.

Challenges

The large amount of published research into drug interactions might have overwhelmed most healthcare practitioners. As a clinician cannot know all potential clinically significant drug interactions, this emphasizes the need for practical identification and management systems 6. The inefficiency of updating and maintaining drug labels poses a serious threat to patients. For example, despite the recent evidence supporting a larger contribution of CYP2C8 and a less significant role for CYP3A4 in the metabolism of imatinib, drug labels continue to warn against potential interactions with CYP3A4 inhibitors but fail to mention any risk with a CYP2C8 inhibitor 38. Few databases and software programs are capable of checking for all potential interactions among multiple medications. More importantly, the absence of reported interactions does not guarantee a lack of interaction 39.

Recommendations

The majority of the phase I trials have evaluated two drug combinations of therapeutic interest, rather than combinations hypothesized to have a DDI. When DDI studies were performed with a clear rationale, the probability of identifying a DDI increased from 8% to 32%. This demonstrates the importance of understanding the mechanism behind a DDI and the value to which this translates clinically and suggests that DDI studies should only be performed when there is a pre-specified plausible hypothesis 40.

Most pharmacokinetic studies would report changes in TKI exposure as a result of the enzyme inhibitor or inducer. However it would be useful if the investigators also provided information regarding its potential effect on toxicity or efficacy 41.

It is imperative that comprehensive and accurate information be collected on the use of medications by patients, to increase awareness and familiarity with potential DDIs to ensure patient safety and to aid the development of optimal therapy 42,43. To reduce the potential for unexpected drug interactions during therapy, the patient's medical history should be taken thoroughly and interactively, and updated periodically 1. Patients who have risk factors for potential DDIs should also be monitored more closely. Risk factors for potential DDI include liver function status, age, tumour type 44, number and type of medications received 45 and using drugs that are metabolized exclusively by only one CYP isoform 46.

When drugs with potential DDIs are considered for use with TKIs, clinicians should also consider alternative agents that have no or less interaction potential. However, it is also important to note that in some cases, switching to an alternative agent does not have any significant difference on the pharmacokinetic profile. For instance, despite azithromycin having a low potential for interaction, there was no significant effect of oral clarithromycin or azithromycin on the pharmacokinetic profile of sunitinib after single administration 47. Because we do not know the clinical effects of these potential interactions when drugs with potential DDIs need to be used with TKIs (e.g. in cases where there is a compelling indication for the potential interacting drug to be used), more intense patient monitoring for interactions is needed. Scripture et al. have provided a valuable summary of the conditions where drug interactions are likely to be clinically significant, such as when drug elimination occurs primarily through a single metabolic pathway or when one or both of the interacting drugs has a steep dose−response curve or a narrow therapeutic range etc 14.

Utilization of therapeutic drug monitoring (TDM) in DDIs

The prerequisites of being a candidate for therapeutic drug monitoring (TDM) in clinical practice include long term therapy, significant inter-individual but limited intra-individual PK variability, narrow therapeutic index, a well-defined exposure−response (efficacy/toxicity) relationship and availability of appropriate bio-analytical methods for quantification 4851. As most of the TKIs fulfil the traditional criteria for a TDM programme, the role of TDM in TKI therapy is increasingly being studied. Furthermore, as TKIs have the potential to be involved in multiple interactions (e.g. drug−drug, drug−food and drug−herb) involving pharmacokinetic or pharmacodynamic pathways, TDM could complement clinical evaluation by providing additional information on efficacy, adherence and toxicity 51.

The application of TDM may be useful in DDIs for several reasons: for monitoring of patient when high risk drug pairs cannot be avoided, for diagnosis of DDIs and for dose adjustments 49. In such cases, the changes in drug concentrations, together with the patient's response and toxicity, could be used together to make an informed decision, on whether the drug pair can be continued safely or whether dose adjustments should be performed. For example, in a recent study conducted by our group, we have demonstrated that sunitinib is more dermatotoxic than its active metabolite, SU12662 11. If sunitinib is used concomitantly with a CYP3A4 inducer, the total effective plasma concentration (sunitinib and SU12662) may still be above the therapeutic target. However, patients may experience less toxicity due to the lesser accumulation of the parent drug.

Although TDM of TKIs is still in its infancy, there is increasing evidence that dose adjustments based on pharmacokinetic targets would help to increase efficacy and reduce toxicity of TKIs, and might be beneficial for patients treated with most of the TKIs 50. Currently, target plasma concentrations of TKIs and their respective dose-adaptation strategy are only available for several of the FDA-approved TKIs. Recommended therapeutic targets for efficacy are available for crizotinib, erlotinib, gefitinib, imatinib, nilotinib, pazopanib and sunitinib, and targets for safety are available for dasatinib and sunitinib 50. Thus, application of TDM may be limited to those TKIs which have a recommended target and may be challenging in those TKIs which have a lack of information. Although there are recommended therapeutic targets available for some of the TKIs, no prospective studies have been conducted to validate these targets, and thus applications of TDM may best be reserved for individual situations relating to a lack of therapeutic response, severe or unexpected toxicities, drug−drug interactions or treatment adherence 51. In summary, future research should focus on the role and benefits of TDM in TKI therapy, especially those with a well-established dose−response relationship and well-established pharmacokinetic targets. Prospective, randomized studies should be performed to confirm the benefits of implementation of TDM, such as reductions in toxicity and/or improvement in outcomes.

Summary and recommendations

As we are able to achieve a better control of the disease over the longer lifespan of a patient, TKIs are now being considered as chronic medications taken in an outpatient setting. Due to the large inter-patient variability in pharmacokinetics of these TKIs, any potential DDI could have serious consequences in a patient's therapy. Thus far, there have been numerous phase I and in vitro studies conducted to evaluate DDIs. This information has been incorporated into drug labels or drug information databases, to warn prescribers of the risk of such DDIs. However, as previously mentioned, it is somewhat challenging to extrapolate results from phase I and in vitro studies to routine clinical practice. During the drug development phase, potentially clinically relevant drug interactions are not usually detected. Only after receiving regulatory approval and after widespread usage, would a new DDI typically surface. Therefore, healthcare professionals, especially physicians and pharmacists, play a vital role in identifying these new interactions. Perhaps scheduled drug utilization reviews could be conducted routinely to identify any common DDI pairs. Prospective data could then be collected regarding these DDI pairs, to identify any increase in toxicity or lack of efficacy events. Where possible, investigators could also subsequently recreate the interaction in an in vitro system to determine the mechanisms which may be involved. However, such research takes time and in the meantime, suspected DDI pairs would have to be used cautiously in patients. Nevertheless, using both clinical and in vitro data to validate claims of potential DDIs would ensure accuracy of data as well as clinical relevance. The utilization of TDM in DDIs may be useful when high risk drug pairs cannot be avoided, especially during initiation of therapy or when there is lack of response or occurrence of severe toxicities. Instead of withholding a beneficial drug therapy from a patient, the high risk drug pair may be continued safely with regular monitoring if the patient is not experiencing excessive toxicity, even when the drug concentrations are increased. In these situations, using additional information on drug concentrations together with patient's response and toxicity data, a more informed decision can be made.

Competing Interests

All authors have completed the Unified Competing Interest form at http://www.icmje.org/coi_disclosure.pdf (available on request from the corresponding author) and declare no support from any organization for the submitted work, no financial relationships with any organizations that might have an interest in the submitted work in the previous 3 years and no other relationships or activities that could appear to have influenced the submitted work.

References

  1. Tyler T. Drug interactions in metastatic breast cancer. J Oncol Pharm Pract. 2011;17:236–245. doi: 10.1177/1078155210379167. [DOI] [PubMed] [Google Scholar]
  2. Bowlin SJ, Xia F, Wang W, Robinson KD, Stanek EJ. Twelve-month frequency of drug-metabolizing enzyme and transporter-based drug−drug interaction potential in patients receiving oral enzyme-targeted kinase inhibitor antineoplastic agents. Mayo Clin Proc. 2013;88:139–148. doi: 10.1016/j.mayocp.2012.10.020. [DOI] [PubMed] [Google Scholar]
  3. Hadjibabaie M, Badri S, Ataei S, Moslehi AH, Karimzadeh I, Ghavamzadeh A. Potential drug−drug interactions at a referral hematology-oncology ward in Iran: a cross-sectional study. Cancer Chemother Pharmacol. 2013;71:1619–1627. doi: 10.1007/s00280-013-2162-5. [DOI] [PubMed] [Google Scholar]
  4. Ko Y, Tan SLD, Chan A, Wong YP, Yong WP, Ng RCH, Lim SW, Salim A. Prevalence of the coprescription of clinically important interacting drug combinations involving oral anticancer agents in Singapore: a retrospective database study. Clin Ther. 2012;34:1696–1704. doi: 10.1016/j.clinthera.2012.06.025. [DOI] [PubMed] [Google Scholar]
  5. Chan A, Tan SH, Wong CM, Yap KYL, Ko Y. Clinically significant drug−drug interactions between oral anticancer agents and nonanticancer agents: a delphi survey of oncology pharmacists. Clin Ther. 2009;31:2379–2386. doi: 10.1016/j.clinthera.2009.11.008. [DOI] [PubMed] [Google Scholar]
  6. Lees J, Chan A. Polypharmacy in elderly patients with cancer: clinical implications and management. Lancet Oncol. 2011;12:1249–1257. doi: 10.1016/S1470-2045(11)70040-7. [DOI] [PubMed] [Google Scholar]
  7. Kruse V, Somers A, Van Bortel L, De Both A, Van Belle S, Rottey S. Sunitinib for metastatic renal cell cancer patients: observational study highlighting the risk of important drug−drug interactions. J Clin Pharm Ther. 2014;39:259–265. doi: 10.1111/jcpt.12134. [DOI] [PubMed] [Google Scholar]
  8. Fujita KI. Cytochrome P450 and anticancer drugs. Curr Drug Metab. 2006;7:23–37. doi: 10.2174/138920006774832587. [DOI] [PubMed] [Google Scholar]
  9. Teo YL, Saetaew M, Chanthawong S, Yap YS, Yong Chan EC, Ho HK, Chan A. Effect of CYP3A4 inducer dexamethasone on hepatotoxicity of lapatinib: clinical and in vitro evidence. Breast Cancer Res Treat. 2012;133:703–711. doi: 10.1007/s10549-012-1995-7. [DOI] [PubMed] [Google Scholar]
  10. Rosenbaum S, Wu S, Newman M, West D, Kuzel T, Lacouture M. Dermatological reactions to the multitargeted tyrosine kinase inhibitor sunitinib. Support Care Cancer. 2008;16:557–566. doi: 10.1007/s00520-008-0409-1. [DOI] [PubMed] [Google Scholar]
  11. Teo YL, Chong XJ, Chue XP, Chau NM, Tan MH, Kanesvaran R, Wee HL, Ho HK, Chan A. Role of sunitinib and SU12662 on dermatological toxicities in metastatic renal cell carcinoma patients: in vitroin vivo, and outcomes investigation. Cancer Chemother Pharmacol. 2013;73:381–388. doi: 10.1007/s00280-013-2360-1. [DOI] [PubMed] [Google Scholar]
  12. Dutreix C, Peng B, Mehring G, Hayes M, Capdeville R, Pokorny R, Seiberling M. Pharmacokinetic interaction between ketoconazole and imatinib mesylate (Glivec) in healthy subjects. Cancer Chemother Pharmacol. 2004;54:290–294. doi: 10.1007/s00280-004-0832-z. [DOI] [PubMed] [Google Scholar]
  13. Abbas R, Hug BA, Leister C, Burns J, Sonnichsen D. Effect of ketoconazole on the pharmacokinetics of oral bosutinib in healthy subjects. J Clin Pharmacol. 2011;51:1721–1727. doi: 10.1177/0091270010387427. [DOI] [PubMed] [Google Scholar]
  14. Scripture CD, Figg WD. Drug interactions in cancer therapy. Nat Rev Cancer. 2006;6:546–558. doi: 10.1038/nrc1887. [DOI] [PubMed] [Google Scholar]
  15. Martin P, Oliver S, Robertson J, Kennedy SJ, Read J, Duvauchelle T. Pharmacokinetic drug interactions with vandetanib during coadministration with rifampicin or itraconazole. Drugs R D. 2011;11:37–51. doi: 10.2165/11586980-000000000-00000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  16. 2013. Bosulif (bosutinib) prescribing information. Pfizer;
  17. Flaherty KT, Lathia C, Frye RF, Schuchter L, Redlinger M, Rosen M, O'Dwyer PJ. Interaction of sorafenib and cytochrome P450 isoenzymes in patients with advanced melanoma: a phase I/II pharmacokinetic interaction study. Cancer Chemother Pharmacol. 2011;68:1111–1118. doi: 10.1007/s00280-011-1585-0. [DOI] [PubMed] [Google Scholar]
  18. Takimoto CH, Awada A. Safety and anti-tumor activity of sorafenib (Nexavar®) in combination with other anti-cancer agents: a review of clinical trials. Cancer Chemother Pharmacol. 2008;61:535–548. doi: 10.1007/s00280-007-0639-9. [DOI] [PubMed] [Google Scholar]
  19. Infante JR, Jones SF, Bendell JC, Greco FA, Yardley DA, Lane CM, Spigel DR, Hainsworth JD, Burris Iii HA. A drug interaction study evaluating the pharmacokinetics and toxicity of sorafenib in combination with capecitabine. Cancer Chemother Pharmacol. 2012;69:137–144. doi: 10.1007/s00280-011-1674-0. [DOI] [PubMed] [Google Scholar]
  20. O'Brien SG, Meinhardt P, Bond E, Beck J, Peng B, Dutreix C, Mehring G, Milosavljev S, Huber C, Capdeville R, Fischer T. Effects of imatinib mesylate (ST1571, Glivec) on the pharmacokinetics of simvastatin, a cytochrome P450 3A4 substrate, in patients with chronic myeloid leukaemia. Br J Cancer. 2003;89:1855–1859. doi: 10.1038/sj.bjc.6601152. [DOI] [PMC free article] [PubMed] [Google Scholar]
  21. Wang Y, Zhou L, Dutreix C, Leroy E, Yin Q, Sethuraman V, Riviere GJ, Yin OQP, Schran H, Shen ZX. Effects of imatinib (Glivec) on the pharmacokinetics of metoprolol, a CYP2D6 substrate, in Chinese patients with chronic myelogenous leukaemia. Br J Clin Pharmacol. 2008;65:885–892. doi: 10.1111/j.1365-2125.2008.03150.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  22. Swaisland HC, Ranson M, Smith RP, Leadbetter J, Laight A, McKillop D, Wild MJ. Pharmacokinetic drug interactions of gefitinib with rifampicin, itraconazole and metoprolol. Clin Pharmacokinet. 2005;44:1067–1081. doi: 10.2165/00003088-200544100-00005. [DOI] [PubMed] [Google Scholar]
  23. Peereboom DM, Ahluwalia MS, Ye X, Supko JG, Hilderbrand SL, Phuphanich S, Nabors LB, Rosenfeld MR, Mikkelsen T, Grossman SA. NABTT 0502: a phase II and pharmacokinetic study of erlotinib and sorafenib for patients with progressive or recurrent glioblastoma multiforme. Neuro Oncol. 2013;15:490–496. doi: 10.1093/neuonc/nos322. [DOI] [PMC free article] [PubMed] [Google Scholar]
  24. De Jonge MJA, Hamberg P, Verweij J, Savage S, Suttle AB, Hodge J, Arumugham T, Pandite LN, Hurwitz HI. Phase I and pharmacokinetic study of pazopanib and lapatinib combination therapy in patients with advanced solid tumors. Invest New Drugs. 2013;31:751–759. doi: 10.1007/s10637-012-9885-8. [DOI] [PubMed] [Google Scholar]
  25. Brain E, Isambert N, Dalenc F, Diéras V, Bonneterre J, Rezai K, Jimenez M, Mefti-Lacheraf F, Cottura E, Tresca P, Vanlemmens L, Mahier-Aït Oukhatar C, Lokiec F, Fumoleau P. Phase I study of lapatinib plus vinorelbine in patients with locally advanced or metastatic breast cancer overexpressing HER2. Br J Cancer. 2012;106:673–677. doi: 10.1038/bjc.2011.591. [DOI] [PMC free article] [PubMed] [Google Scholar]
  26. Jamei M, Marciniak S, Feng K, Barnett A, Tucker G, Rostami-Hodjegan A. The Simcyp® population-based ADME simulator. Expert Opin Drug Metab Toxicol. 2009;5:211–223. doi: 10.1517/17425250802691074. [DOI] [PubMed] [Google Scholar]
  27. Shapiro LE, Shear NH. Drug interactions: proteins, pumps, and P-450s. J Am Acad Dermatol. 2002;47:467–484. doi: 10.1067/mjd.2002.126823. [DOI] [PubMed] [Google Scholar]
  28. Pal D, Mitra AK. CYP3A4 and MDR mediated interactions in drug therapy. Clin Res Regul Aff. 2006;23:125–163. [Google Scholar]
  29. Purnapatre K, Khattar SK, Saini KS. Cytochrome P450s in the development of target-based anticancer drugs. Cancer Lett. 2008;259:1–15. doi: 10.1016/j.canlet.2007.10.024. [DOI] [PubMed] [Google Scholar]
  30. Pajares B, Torres E, Trigo JM, Sáez MI, Ribelles N, Jiménez B, Alba E. Tyrosine kinase inhibitors and drug interactions: a review with practical recommendations. Clin Transl Oncol. 2012;14:94–101. doi: 10.1007/s12094-012-0767-5. [DOI] [PubMed] [Google Scholar]
  31. Kenny JR, Mukadam S, Zhang C, Tay S, Collins C, Galetin A, Khojasteh SC. Drug-drug interaction potential of marketed oncology drugs: in vitro assessment of time-dependent cytochrome P450 inhibition, reactive metabolite formation and drug−drug interaction prediction. Pharm Res. 2012;29:1960–1976. doi: 10.1007/s11095-012-0724-6. [DOI] [PubMed] [Google Scholar]
  32. Castellino S, O'Mara M, Koch K, Borts DJ, Bowers GD, MacLauchlin C. Human metabolism of lapatinib, a dual kinase inhibitor: implications for hepatotoxicity. Drug Metab Dispos. 2012;40:139–150. doi: 10.1124/dmd.111.040949. [DOI] [PubMed] [Google Scholar]
  33. Lammert C, Einarsson S, Saha C, Niklasson A, Bjornsson E, Chalasani N. Relationship between daily dose of oral medications and idiosyncratic drug-induced liver injury: search for signals. Hepatology. 2008;47:2003–2009. doi: 10.1002/hep.22272. [DOI] [PubMed] [Google Scholar]
  34. Uetrecht J. Idiosyncratic drug reactions: current understanding. Annu Rev Pharmacol Toxicol. 2007;47:513–539. doi: 10.1146/annurev.pharmtox.47.120505.105150. [DOI] [PubMed] [Google Scholar]
  35. Yu K, Geng X, Chen M, Zhang J, Wang B, Ilic K, Tong W. High daily dose and being a substrate of cytochrome P450 enzymes are two important predictors of drug-induced liver injury. Drug Metab Dispos. 2014;42:744–750. doi: 10.1124/dmd.113.056267. [DOI] [PubMed] [Google Scholar]
  36. Gambillara E, Laffitte E, Widmer N, Decosterd LA, Duchosal MA, Kovacsovics T, Panizzon RG. Severe pustular eruption associated with imatinib and voriconazole in a patient with chronic myeloid leukemia. Dermatology. 2005;211:363–365. doi: 10.1159/000088510. [DOI] [PubMed] [Google Scholar]
  37. Xu CF, Xue Z, Bing N, King KS, McCann LA, De PL, Goodman VL, Spraggs CF, Mooser VE, Pandite LN. Concomitant use of pazopanib and simvastatin increases the risk of transaminase elevations in patients with cancer. Ann Oncol. 2012;23:2470–2471. doi: 10.1093/annonc/mds249. [DOI] [PubMed] [Google Scholar]
  38. Seminerio MJ, Ratain MJ. Are drug labels static or dynamic? Clin Pharmacol Ther. 2013;94:302–304. doi: 10.1038/clpt.2013.109. [DOI] [PubMed] [Google Scholar]
  39. Seminerio MJ, Ratain MJ. Preventing adverse drug-drug interactions: a need for improved data and logistics. Mayo Clin Proc. 2013;88:126–128. doi: 10.1016/j.mayocp.2012.12.005. [DOI] [PubMed] [Google Scholar]
  40. Wu K, House L, Ramírez J, Seminerio MJ, Ratain MJ. Evaluation of utility of pharmacokinetic studies in phase I trials of two oncology drugs. Clin Cancer Res. 2013;19:6039–6043. doi: 10.1158/1078-0432.CCR-13-0597. [DOI] [PMC free article] [PubMed] [Google Scholar]
  41. Duckett DR, Cameron MD. Metabolism considerations for kinase inhibitors in cancer treatment. Expert Opin Drug Metab Toxicol. 2010;6:1175–1193. doi: 10.1517/17425255.2010.506873. [DOI] [PMC free article] [PubMed] [Google Scholar]
  42. Hanigan MH, Dela Cruz BL, Shord SS, Medina PJ, Fazili J, Thompson DM. Optimizing chemotherapy: concomitant medication lists. Clin Pharmacol Ther. 2011;89:114–119. doi: 10.1038/clpt.2010.253. [DOI] [PMC free article] [PubMed] [Google Scholar]
  43. Banna GL, Collovà E, Gebbia V, Lipari H, Giuffrida P, Cavallaro S, Condorelli R, Buscarino C, Tralongo P, Ferraù F. Anticancer oral therapy: emerging related issues. Cancer Treat Rev. 2010;36:595–605. doi: 10.1016/j.ctrv.2010.04.005. [DOI] [PubMed] [Google Scholar]
  44. Riechelmann RP, Tannock IF, Wang L, Saad ED, Taback NA, Krzyzanowska MK. Potential drug interactions and duplicate prescriptions among cancer patients. J Natl Cancer Inst. 2007;99:592–600. doi: 10.1093/jnci/djk130. [DOI] [PubMed] [Google Scholar]
  45. Riechelmann RP, Zimmermann C, Chin SN, Wang L, O'Carroll A, Zarinehbaf S, Krzyzanowska MK. Potential drug interactions in cancer patients receiving supportive care exclusively. J Pain Symptom Manage. 2008;35:535–543. doi: 10.1016/j.jpainsymman.2007.06.009. [DOI] [PubMed] [Google Scholar]
  46. Blower P, De Wit R, Goodin S, Aapro M. Drug−drug interactions in oncology: why are they important and can they be minimized? Crit Rev Oncol Hematol. 2005;55:117–142. doi: 10.1016/j.critrevonc.2005.03.007. [DOI] [PubMed] [Google Scholar]
  47. Szałek E, Karbownik A, Połom W, Matuszewski M, Sobańska K, Urjasz H, Grabowski T, Wolc A, Grześkowiak E. Sunitinib in combination with clarithromycin or azithromycin – Is there a risk of interaction or not? Pharmacol Rep. 2012;64:1554–1559. doi: 10.1016/s1734-1140(12)70953-2. [DOI] [PubMed] [Google Scholar]
  48. Gao B, Yeap S, Clements A, Balakrishnar B, Wong M, Gurney H. Evidence for therapeutic drug monitoring of targeted anticancer therapies. J Clin Oncol. 2012;30:4017–4025. doi: 10.1200/JCO.2012.43.5362. [DOI] [PubMed] [Google Scholar]
  49. Josephs DH, Fisher DS, Spicer J, Flanagan RJ. Clinical pharmacokinetics of tyrosine kinase inhibitors: implications for therapeutic drug monitoring. Ther Drug Monit. 2013;35:562–587. doi: 10.1097/FTD.0b013e318292b931. [DOI] [PubMed] [Google Scholar]
  50. Yu H, Steeghs N, Nijenhuis CM, Schellens JHM, Beijnen JH, Huitema ADR. Practical guidelines for therapeutic drug monitoring of anticancer tyrosine kinase inhibitors: focus on the pharmacokinetic targets. Clin Pharmacokinet. 2014;53:305–325. doi: 10.1007/s40262-014-0137-2. [DOI] [PubMed] [Google Scholar]
  51. Widmer N, Bardin C, Chatelut E, Paci A, Beijnen J, Levêque D, Veal G, Astier A. Review of therapeutic drug monitoring of anticancer drugs part two – targeted therapies. Eur J Cancer. 2014;50:2020–2036. doi: 10.1016/j.ejca.2014.04.015. [DOI] [PubMed] [Google Scholar]
  52. 2013. Gilotrif (afatinib) prescribing information. Boehringer Ingelheim;
  53. 2013. Inlyta (axitinib) prescribing information. Pfizer;
  54. 2012. Cometriq (cabozantinib) prescribing information. Exelixis;
  55. 2014. Xalkori (crizotinib) prescribing information. Pfizer;
  56. 2014. Sprycel (dasatinib) prescribing information. Bristol-Myers Squibb;
  57. 2014. Tarceva (erlotinib) prescribing information. OSI Pharmaceuticals;
  58. 2005. Iressa (gefitinib) prescribing information. AstraZeneca;
  59. 2014. Gleevec (imatinib) prescribing information. Novartis;
  60. 2013. Tykerb (lapatinib) prescribing information. GlaxoSmithKline;
  61. 2014. Tasigna (nilotinib) prescribing information. Novartis;
  62. 2014. Votrient (pazopanib) prescribing information. GlaxoSmithKline;
  63. 2014. Iclusig (ponatinib) prescribing information. Ariad Pharmaceuticals;
  64. 2013. Stivarga (regorafenib) prescribing information. Bayer;
  65. 2013. Nexavar (sorafenib) prescribing information. Bayer;
  66. 2013. Sutent (sunitinib) prescribing information. Pfizer;
  67. 2014. Caprelsa (vandetanib) prescribing information. AstraZeneca;
  68. Food and Drug Administration. 2014. Product label information. Available at http://www.accessdata.fda.gov/scripts/cder/drugsatfda/index.cfm (last accessed July )
  69. European Medicines Agency. 2014. European public assessment reports. Available at http://www.ema.europa.eu/ema/index.jsp?curl=pages/includes/medicines/medicines_landing_page.jsp&mid= (last accessed August )
  70. Pithavala YK, Tong W, Mount J, Rahavendran SV, Garrett M, Hee B, Selaru P, Sarapa N, Klamerus KJ. Effect of ketoconazole on the pharmacokinetics of axitinib in healthy volunteers. Invest New Drugs. 2012;30:273–281. doi: 10.1007/s10637-010-9511-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  71. Pithavala YK, Tortorici M, Toh M, Garrett M, Hee B, Kuruganti U, Ni G, Klamerus KJ. Effect of rifampin on the pharmacokinetics of axitinib (AG-013736) in Japanese and Caucasian healthy volunteers. Cancer Chemother Pharmacol. 2010;65:563–570. doi: 10.1007/s00280-009-1065-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  72. Johnson FM, Agrawal S, Burris H, Rosen L, Dhillon N, Hong D, Blackwood-Chirchir A, Luo FR, Sy O, Kaul S, Chiappori AA. Phase 1 pharmacokinetic and drug-interaction study of dasatinib in patients with advanced solid tumors. Cancer. 2010;116:1582–1591. doi: 10.1002/cncr.24927. [DOI] [PubMed] [Google Scholar]
  73. Chhun S, Verstuyft C, Rizzo-Padoin N, Simoneau G, Becquemont L, Peretti I, Swaisland A, Wortelboer R, Bergmann JF, Mouly S. Gefitinib−phenytoin interaction is not correlated with the 14C-erythromycin breath test in healthy male volunteers. Br J Clin Pharmacol. 2009;68:226–237. doi: 10.1111/j.1365-2125.2009.03438.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  74. Filppula AM, Tornio A, Niemi M, Neuvonen PJ, Backman JT. Gemfibrozil impairs imatinib absorption and inhibits the CYP2C8-mediated formation of its main metabolite. Clin Pharmacol Ther. 2013;94:383–393. doi: 10.1038/clpt.2013.92. [DOI] [PubMed] [Google Scholar]
  75. Bolton AE, Peng B, Hubert M, Krebs-Brown A, Capdeville R, Keller U, Seiberling M. Effect of rifampicin on the pharmacokinetics of imatinib mesylate (Gleevec, STI571) in healthy subjects. Cancer Chemother Pharmacol. 2004;53:102–106. doi: 10.1007/s00280-003-0722-9. [DOI] [PubMed] [Google Scholar]
  76. Pursche S, Schleyer E, von Bonin M, Ehninger G, Said SM, Prondzinsky R, Illmer T, Wang Y, Hosius C, Nikolova Z, Bornhäuser M, Dresemann G. Influence of enzyme-inducing antiepileptic drugs on trough level of imatinib in glioblastoma patients. Curr Clin Pharmacol. 2008;3:198–203. doi: 10.2174/157488408785747656. [DOI] [PMC free article] [PubMed] [Google Scholar]
  77. Smith DA, Koch KM, Arya N, Bowen CJ, Herendeen JM, Beelen A. Effects of ketoconazole and carbamazepine on lapatinib pharmacokinetics in healthy subjects. Br J Clin Pharmacol. 2009;67:421–426. doi: 10.1111/j.1365-2125.2009.03370.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  78. Tanaka C, Yin OQP, Smith T, Sethuraman V, Grouss K, Galitz L, Harrell R, Schran H. Effects of rifampin and ketoconazole on the pharmacokinetics of nilotinib in healthy participants. J Clin Pharmacol. 2011;51:75–83. doi: 10.1177/0091270010367428. [DOI] [PubMed] [Google Scholar]
  79. Tan AR, Gibbon DG, Stein MN, Lindquist D, Edenfield JW, Martin JC, Gregory C, Suttle AB, Tada H, Botbyl J, Stephenson JJ. Effects of ketoconazole and esomeprazole on the pharmacokinetics of pazopanib in patients with solid tumors. Cancer Chemother Pharmacol. 2013;71:1635–1643. doi: 10.1007/s00280-013-2164-3. [DOI] [PubMed] [Google Scholar]
  80. Narasimhan NI, Dorer DJ, Niland K, Haluska F, Sonnichsen D. Effects of ketoconazole on the pharmacokinetics of ponatinib in healthy subjects. J Clin Pharmacol. 2013;53:974–981. doi: 10.1002/jcph.109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  81. Rudek MA, Moore PC, Mitsuyasu RT, Dezube BJ, Aboulafia D, Gerecitano J, Sullivan R, Cianfrocca ME, Henry DH, Ratner L, Haigentz M, Dowlati A, Little RF, Ivy SP, Deeken JF. A phase 1/pharmacokinetic study of sunitinib in combination with highly active antiretroviral therapy in human immunodeficiency virus-positive patients with cancer: AIDS Malignancy Consortium trial AMC 061. Cancer. 2014;120:1194–1202. doi: 10.1002/cncr.28554. [DOI] [PMC free article] [PubMed] [Google Scholar]
  82. Carlini P, Papaldo P, Fabi A, Felici A, Ruggeri EM, Milella M, Ciccarese M, Nuzzo C, Cognetti F, Ferretti G. Liver toxicity after treatment with gefitinib and anastrozole: drug−drug interactions through cytochrome p450? J Clin Oncol. 2006;24:e60–61. doi: 10.1200/JCO.2006.07.8261. [DOI] [PubMed] [Google Scholar]
  83. Arai S, Mitsufuji H, Nishii Y, Onoda S, Ryuge S, Wada M, Katono K, Iwasaki M, Takakura A, Otani S, Yamamoto M, Yanaihara T, Yokoba M, Kubota M, Katagiri M, Fukui T, Kobayashi H, Yanase N, Hataishi R, Masuda N. Effect of gefitinib on warfarin antithrombotic activity. Int J Clin Oncol. 2009;14:332–336. doi: 10.1007/s10147-008-0871-2. [DOI] [PubMed] [Google Scholar]
  84. Ross DM. Peripheral neuropathy on imatinib treatment for chronic myeloid leukaemia: suspected adverse drug interaction with amlodipine. Intern Med J. 2009;39:708. doi: 10.1111/j.1445-5994.2009.02028.x. [DOI] [PubMed] [Google Scholar]
  85. Di Gion P, Kanefendt F, Lindauer A, Scheffler M, Doroshyenko O, Fuhr U, Wolf J, Jaehde U. Clinical pharmacokinetics of tyrosine kinase inhibitors: focus on pyrimidines, pyridines and pyrroles. Clin Pharmacokinet. 2011;50:551–603. doi: 10.2165/11593320-000000000-00000. [DOI] [PubMed] [Google Scholar]
  86. de Groot JWB, Links TP, van der Graaf WTA. Tyrosine kinase inhibitors causing hypothyroidism in a patient on levothyroxine. Ann Oncol. 2006;17:1719–1720. doi: 10.1093/annonc/mdl112. [DOI] [PubMed] [Google Scholar]
  87. Noda S, Shioya M, Hira D, Fujiyama Y, Morita SY, Terada T. Pharmacokinetic interaction between sorafenib and prednisolone in a patient with hepatocellular carcinoma. Cancer Chemother Pharmacol. 2013;72:269–272. doi: 10.1007/s00280-013-2187-9. [DOI] [PubMed] [Google Scholar]

Articles from British Journal of Clinical Pharmacology are provided here courtesy of British Pharmacological Society

RESOURCES