Abstract
The role of P-glycoprotein (P-gp) and breast cancer resistance protein (BCRP) in drug-drug interactions (DDIs) and limiting drug absorption as well as restricting the brain penetration of drugs with certain physicochemical properties is well known. P-gp/BCRP inhibition by drugs in the gut has been reported to increase the systemic exposure to substrate drugs. A previous International Transporter Consortium (ITC) perspective discussed the feasibility of P-gp/BCRP inhibition at the blood-brain barrier and its implications. This ITC perspective elaborates and discusses specifically the hepatic and renal P-gp/BCRP (referred as systemic) inhibition of drugs and whether there is any consequence for substrate drug disposition. This perspective summarizes the clinical evidence-based recommendations regarding systemic P-gp and BCRP inhibition of drugs with a focus on biliary and active renal excretion pathways. Approaches to assess the clinical relevance of systemic P-gp and BCRP inhibition in liver and kidney included (1) curation of DDIs involving intravenously administered substrates or inhibitors; (2) in vitro-to-in vivo extrapolation of P-gp- mediated DDIs at the systemic level; and (3) curation of drugs with information available about the contribution of biliary excretion and related DDIs. Based on the totality of evidence reported to date, this perspective supports limited clinical DDI risk upon P-gp or BCRP inhibition in liver or kidney.
Keywords: P-gp, BCRP, biliary, renal, systemic, drug interaction, in vitro-to-in vivo extrapolation
Introduction
P-glycoprotein (P-gp, ABCB1, MDR1) and breast cancer resistance protein (BCRP, ABCG2) are efflux transporters located in many organs relevant to drug disposition, including the intestine, brain, liver, and kidney. These generally apically located transporters can limit intestinal absorption of substrate drugs, presenting potential drug-drug interactions (DDIs) when intestinal P-gp and/or BCRP is inhibited by co-administered drugs. P-gp and BCRP also play a crucial role in limiting brain penetration of substrate drugs, and CNS drug discovery programs often aim to prevent or limit P-gp/BCRP efflux to facilitate drug distribution into the brain. In contrast, many drugs are P-gp/BCRP-inhibitors with potencies that are more relevant to the gastrointestinal tract. Use of P-gp-inhibitors in particular to enhance systemic or tissue exposure has been the focus of investigations for many years with some success. For instance, enhanced absorption of P-gp substrates from the gastrointestinal tract is achievable because sufficient concentrations of P-gp-inhibitors, which allow relevant concentrations at the binding site of intestinal P-gp for inhibition, can be attained following oral administration. Besides the use of such P-gp-inhibitors in fixed formulations such as D-α-Tocopherol polyethylene glycol succinate (TPGS),1 a recent clinical example is the use of encequidar to boost oral paclitaxel systemic exposure in cancer patients, allowing treatment from home instead of in an outpatient/hospital setting.2 Except for the intestine, drugs that inhibit P-gp/BCRP typically may not achieve sufficient unbound systemic concentrations at tolerated clinical doses to inhibit efflux at tissue barriers such as the blood-brain barrier capillary endothelium. Extensive investigations have revealed that P-gp inhibition by a drug is not a clinically viable method to enhance CNS drug penetration.3 Because of this lack of brain P-gp inhibition by almost all approved/marketed drugs to date that can inhibit P-gp, luminal efflux can be optimized to effectively limit unwanted CNS drug exposure in humans as a strategy during drug development. Examples of P-gp drug substrates with limited brain exposure include loperamide, ivermectin, and second generation non-sedating antihistamines and antimuscarinics; if not for efflux at the blood-brain barrier, the success of these therapeutic agents likely would be more limited. Similar logic may be applicable to BCRP inhibitors, but prototypical BCRP specific inhibitors at the systemic level have not been thoroughly tested nor identified in the clinic as compared to P-gp-inhibitors.
P-gp/BCRP may also play a role in the biliary (e.g., rosuvastatin for BCRP and digoxin for P-gp) and renal (e.g., digoxin for P-gp) clearance of drugs. Examples of extensive clearance by P-gp-mediated biliary excretion or active tubular secretion are limited because P-gp substrates (generally large molecular weight, lipophilic cations) usually are cleared by metabolism instead of renal excretion (predominant clearance route for more hydrophilic drugs), and direct biliary excretion of parent drug as a major elimination pathway is not common in humans (<5% of marketed drugs).4 Similarly, examples of BCRP-mediated renal clearance of drugs are scarce; furthermore, BCRP protein expression in the human kidney cortex is reported to be low.5 However, there are cases where biliary excretion and renal efflux are (or appear to be) a major elimination pathway of a drug that is not driven by P-gp/BCRP. Thus, estimating when biliary or renal active secretion via P-gp/BCRP is involved and dictate overall drug disposition remains a challenge. Furthermore, there is potential overlap between P-gp/BCRP substrates and inhibitors. These compounds also may be substrates or inhibitors of other transporters and enzymes, which may further complicate evaluation of the hepatic/renal P-gp/BCRP contribution to elimination. The potential for inhibition of both P-gp and CYP3A by substrates and inhibitor drugs must be considered.6
Understanding the potential clinical relevance of systemic P-gp/BCRP inhibition may be important for other scenarios. For example, hepatic and renal P-gp/BCRP inhibition may be relevant in the context of understanding the disposition of antibody-drug conjugate payloads, which often are small molecules delivered by intravenous infusions. The payload is generally cleaved inside cells and may be a substrate for P-gp/BCRP, which may be relevant to understand the altered disposition of these small molecule payloads when P-gp/BCRP are inhibited.7
This perspective summarizes the clinical evidence-based recommendations regarding systemic P-gp and BCRP inhibition of drugs with a focus on biliary and active renal excretion pathways. Throughout this perspective, systemic inhibition of P-gp or BCRP refers to inhibition of either canalicular P-gp or BCRP in the liver or luminal P-gp or BCRP in the kidney, not inhibition of P-gp or BCRP in the intestine, brain or any other organ that expresses these transporters. Diverse approaches were used to assess the clinical relevance of systemic P-gp and BCRP inhibition in liver and kidney, including (1) curation of DDIs involving intravenously administered substrates or inhibitors; (2) in vitro-to-in vivo extrapolation (IVIVE) of P-gp -mediated DDIs at the systemic level; and (3) curation of drugs with information available about the contribution of biliary excretion and related DDIs.
Evaluation of hepatic and renal clinical DDIs that potentially involve systemic P-gp inhibition
P-gp knockout mice or rats provide extreme examples of the impact of P-gp loss-of-function on drug disposition, but results must be interpreted carefully. 3, 8 Although extensive intestinal P-gp inhibition is clinically tenable with an oral inhibitor, P-gp inhibitor drugs at their highest tolerated clinical doses at most achieve maximum unbound systemic concentrations approximating their P-gp inhibitory (IC50) potencies.3 At the systemic disposition level, 100% genetic ablation of mdr1 in rodent knockout models can elicit more drastic changes in substrate disposition than the worst-case scenario of 50% inhibition of systemic P-gp in humans. For example, a 65-fold higher loperamide CNS distribution in knockout mice vs at most around two-fold increase with intravenous infusion of P-gp inhibitors at the highest level tolerated in humans has been reported.9, 10
A review of the University of Washington Drug Interaction Database (UW DIDB, https://didb.druginteractionsolutions.org/) was conducted to evaluate reported systemic P-gp-mediated DDIs (searched in February of 2022). The search criteria were clinical DDIs involving P-gp inhibitors and intravenously administered substrates (victim drugs), which showed a ≥25% increase in plasma area under the concentration-time curve (AUC) of substrates (Table 1). Intravenous (IV) administration of P-gp substrates was selected to avoid DDIs driven by intestinal P-gp inhibition and to specifically focus on the implications of systemic P-gp inhibition. As shown by the data, 83% of the AUC changes (44 of 53 DDI records) for the identified drugs were within <2-fold (i.e., <100% increase) after co-administration with small molecules or drugs known to inhibit P-gp. The 15 P-gp inhibitors identified were amiodarone, biricodar (VX-710), cyclosporine, dofequidar (MS209), elacridar (GF120918), OC144-093 (ONT-093), paclitaxel, propafenone, quinidine, quinine, ritonavir, spironolactone, verapamil, valspodar (PSC833), and zosuquidar (LY335979). Some P-gp-inhibitors were excluded, such as itraconazole and ketoconazole, because the studies involved substrates that are also metabolized by cytochrome P450 (CYP) 3A, rendering difficulty in determining whether and how much P-gp inhibition contributes to the observed DDI.
Table 1:
Object | Object dosing route |
Perpetrator | Perpetrator dosing route |
% Change of Object AUC |
% Change of CL |
% Change of CLrenal/ CLnon- renal |
Precipitant Dose |
Precipitant Interval |
Reference |
---|---|---|---|---|---|---|---|---|---|
Digoxin | IV | amiodarone | Oral | 33a | −25 | −16 (CLr) −31 (CLnr) | 400 mg (21-25 days) | not provided | 58 |
Digoxin | IV | amiodarone | Oral | 47a | −32 | −30 (CLr) −34 (CLnr) | 1600 mg (14 days) | qd on Days 5-18 | 59 |
Digoxin | IV | propafenone | Oral | 28 | −22 | −9 (CLr) −44 (CLnr) | 300 mg (12 days) | tid | 35 |
Digoxin | IV | quinidine | Oral | 53a | −35 | −27 (CLr) −46 (CLnr) | Not Provided | (chronic therapy) | 46 |
Digoxin | IV | quinidine | Oral | 57a | −36 | −32 (CLr) | 200 mg (4 days) | qid | 60 |
Digoxin | IV | quinidine | Oral | 57a | −36 | −54 (CLr) −22 (CLnr) | 400 mg (4 days) | qid starting 3 days before digoxin administration | 61 |
43a | −30 | −32 (CLr) −29 (CLnr) | 200 mg (4 days) | ||||||
65a | −39 | 200 mg (11 days) | qid | 62 | |||||
Digoxin | IV | quinidine | Oral | 79a | −44 | 202 mg (26 days) | bid | 63 | |
Digoxin | IV | quinidine | Oral | 83a | −45 | −33 (CLr) −60 (CLnr) | 200 mg (8 days) | qid | 64 |
Digoxin | IV | quinidine | Oral | 178a | −64 | −61 (CLr) | 200 mg (6 days) | qid | 65 |
Digoxin | IV | quinidine | Oral | 128a | −56 | −52 (CLr) | 0.6 g (11 days) | bid (after a loading dose of 0.1 g/10 kg) | 66 |
Digoxin | IV | quinine | Oral | 38a | −27 | −15 (CLr) −69 (CLnr) | 200 mg (9 days) | tid, 4 days before and after the second digoxin administration | 67 |
Digoxin | IV | ritonavir | Oral | 86 | −42 | −35 (CLr) −48 (CLnr) | 300 mg (11 days) | bid on Days 1-11 | 68 |
Digoxin | IV | Spironolactone | Oral | 47 | −32 | −26 (CLr) −42 (CLnr) | 100 mg (5 days) | bid | 69 |
Digoxin | IV | verapamil | Oral | 57a | −36 | −23 (CLr) −67 (CLnr) | 80 mg (10 days) | tid | 70 |
Digoxin | IV | verapamil | Oral | 41a | −29 | -- | 120 mg (12 days) | tid | 71 |
Docetaxel | IV | MS209 | Oral | 27, 33 | -- | -- | 300 mg (1 cycle) | administered 30 min before docetaxel infusion at cycle 2 | 23 |
38.5 | -- | -- | 600 mg (1 cycle) | ||||||
96 | -- | -- | 900 mg (1 cycle) | ||||||
54 | -- | -- | 1200 mg (1 cycle) | ||||||
Doxorubicin | IV | Verapamil | Oral | 71 | −40 | -- | 240-480 mg/day [7 days | 80 mg tid for 3 days followed by 120 mg qid for 4 days; one hour before doxorubicin on study day. | 72 |
Doxorubicin | IV | cyclosporine | IV | 55 | −47 | A subset of patients −35(CLtotal) −32 (CLr) −35 (CLnr) | 18 mg/Kg | Co-administered with doxorubicin as a 48-hour continuous infusion after a loading dose of 6 mg/Kg over 2 h | 73 |
Doxorubicin | IV | cyclosporine | IV | 48 | −36.6 | -- | 16 mg/kg/24h (2 days) | continuous infusion, preceded by a 6 mg/kg bolus infused over 2 hours | 74 |
Doxorubicin | IV | Elacridar (GF120918) | Oral | 47 | -- | 400 mg (5d) | bid | 20 | |
Doxorubicin | IV | paclitaxel | IV | 30.4 | −11.8 | -- | 200 mg/m2 | infusion over 3 hours | 75 |
Doxorubicin (polyethylene glycol-coated liposomal formulation | IV | paclitaxel | IV | 97.3 | −38.9 | -- | 175 mg/m2 | Single, 3-hr infusion | 76 |
58.6 | −22.8 | -- | 70 mg/m2 | 1-hr infusion, weekly | |||||
65.5 | −39.5 | -- | |||||||
Doxorubicin | IV | Valspodar (PSC833) | IV | 119.4 | −55.7 | -- | 1-10 mg/kg | single dose as an infusion over 24 hours after a loading dose of 1-2 mg/kg or 2 mg/kg administered as an infusion over 2 hours | 77 |
169.5 | −61.5 | -- | |||||||
49.3 | −36.5 | -- | |||||||
Doxorubicin | IV | Valspodar (PSC833) | Oral | 54.3 | −30.3 | -- | 1.25-12.5 mg/kg (5 days) | bid (7 am) | 78 |
Doxorubicin | IV | Valspodar (PSC833) | Oral | 84.6 | −45.2 | -- | 5 mg/kg (3 days) | qid | 79 |
Doxorubicin | IV | Valspodar (PSC833) | Oral | 93.8 | −41.6 | -- | 2.5 mg/kg (7 days) | bid | 80 |
Doxorubicin | IV | verapamil | Oral | 71 | −40 | -- | 240-480 mg/day (7 days) | 80 mg tid for 3 days followed by 120 mg qid for 4 days | 72 |
Doxorubicin | IV | Zosuquidar (LY335979) | IV | 25 | −17 | -- | 80 - 640 mg/m2/day (2 days/cycle) | continuous i.v. infusion over 48 hours starting on day 1 of cycle 2 | 21 |
Edoxaban | IV | quinidine | Oral | 34.8 | −25.5 | −25 (CLr) | 300 mg (4 days) | three times daily | 81 |
Etoposide | IV | valspodar (PSC833) | IV | 59a | −37 | -- | 1.5 mg/kg then 10 mg/kg (5 days) | 2-hour infusion as 1.5 mg/kg loading dose on Day 2, followed by 10 mg/kg/day continuous infusion on Days 2-6 | 82 |
Etoposide | IV | valspodar (PSC833) | IV | 133a | −57 | -- | 10 mg/kg/day (5 days) | continuous 120 h infusion with concomitant loading dose of 2 mg/kg over 4 h | 83 |
Etoposide | IV | valspodar (PSC833) | IV | 76 | −41 | -- | 1-15 mg/kg (5 days) | continuous infusion increasing from 1 or 6.6 mg/kg to 15 mg/kg from day 2 to day 6, after a loading dose of 1 to 2 mg/kg over 2 hours | 52 |
90 | −46 | −28 (CLr) −55 (CLnr) | 6.6-15 mg/kg (5 days) | ||||||
Etoposide | IV | valspodar (PSC833) | Oral | 113a | −53 | -- | 4 mg/kg (n = 5), 5 mg/kg (n = 11) or 6 mg/kg (n = 7) (one 28-day cycle) | qid on days 0-4, 7-11 and 14-18 | 84 |
Idarubicin | IV | cyclosporine | IV | 66.5 | - | -- | 16 mg/kg/day (6 days) | continuous infusion on days 1-6 after a loading dose of 6 mg/kg over 2 hrs on day 1 | 85 |
Idarubicin | IV | cyclosporine | IV | 78 | −40.2 | 10 mg/kg/day (3.5 days) | injection starting 12 h before the 1st idarubicin administration and stopping 24 h after the last idarubicin dose | 86 | |
Paclitaxel | IV | biricodar (VX-710) | IV | 100a | −50 | 120 mg/m2/hr (24 hours) | 24-hr continuous infusion | 18 | |
Paclitaxel | IV | OC144–093 (ONT-093) | Oral | 44.5 | −29 | 500 mg (1-3 cycles) | 14h before, 2 h before and 10 h after paclitaxel administration | 87 | |
25 | −23 | 300 mg (1-3 cycles) | |||||||
Paclitaxel | IV | valspodar (PSC833) | Oral | 108a | −52 | 5 mg/kg (7 days) | qid; starting 72 hours before the initiation of paclitaxel infusion | 88 | |
Paclitaxel | IV | valspodar (PSC833) | Oral | 150a | −60.1 | 5 mg/Kg (7 days) | qid; starting 72 hours before paclitaxel infusion | 88 | |
Vinorelbine | IV | Zosuquidar (LY335979) | Oral | 30 | −24 | 200 - 300 mg/m2 (9 days/28 day-cycle) | TID for 7 doses on days 7-9 and 14-16 of the first 28-day cycle | 22 |
The AUC change was derived from the change of CL of substrate, since AUC ratio is the reciprocal of CL ratio.
Similarly, we examined the potential involvement of other transporters (e.g., BCRP, OATP1B1, OATP1B3, and the efflux transporter multidrug resistance-associated protein (MRP) 2) for studies involving cyclosporine, which inhibits all of these transporters. Only studies conducted with doxorubicin and idarubicin are included in the dataset (Table 1). Doxorubicin is not a substrate for OATP1B1 or OATP1B3.11, 12 Although there are conflicting data regarding whether doxorubicin is transported by BCRP, studies suggested that doxorubicin was transported by a genetic variant of BCRP but not wild-type BCRP.12, 13, 14 Studies demonstrated that BCRP did not confer drug resistance to idarubicin.15, 16 There were no reports about idarubicin being a substrate for OATP1B. As such, we assumed that idarubicin is not an OATP1B substrate. Although the interference of CYP3A or OATP1B inhibition was minimized for our analysis purpose, CYP3A and hepatic OATPs are often rate-determining in systemic drug clearance and should be considered as important parallel mechanisms of DDIs when evaluating the DDI potential between a P-gp substrate and an inhibitor.17
The largest AUC increase in the compiled dataset was 178% for digoxin in the presence of quinidine. However, numerous DDI studies involved coadministration of digoxin and quinidine (n=9). On average, the AUC of digoxin was increased by 83% (Table 1). The remaining DDIs with a substrate AUC that increased >2-fold commonly involved the inhibitor valspodar (PSC833) (a P-gp inhibitor used only as a clinical research tool). When all the DDIs were compiled, valspodar on average increased the AUC of doxorubicin by 95% (n=4), etoposide by 94% (n=4), and paclitaxel by 129% (n=2). Another study involved biricodar (VX-710), which doubled the AUC of paclitaxel.18 Valspodar (PSC833) and biricodar (VX-710) are designer P-gp-inhibitors, as are elacridar (GF120918), dofequidar, and zosuquidar (LY335979), which were developed to overcome multiple drug resistance of tumor cells to some chemotherapeutic agents that are P-gp-substrates. However, elacridar (GF120918) had only a minor effect on doxorubicin exposure, except at the highest combined doses (i.e., 75 mg/m2 doxorubicin and 400 mg twice daily elacridar (GF120918)).19, 20 Zosuquidar (LY335979) modestly increased the AUC of doxorubicin and vinorelbine, by up to 25% and 30%, respectively.21, 22 Dofequidar increased docetaxel AUC by approximately 50% or less except for one group of treatments showing a 96% increase.23 A study evaluating blood-brain barrier P-gp inhibition using PET imaging showed that tariquidar inhibited P-gp in a dose-dependent manner by up to 75% (N-des-loperamide as substrate) or 58% (verapamil as substrate); however, at the dose (2 mg/kg) evaluated in efficacy trials, tariquidar inhibited P-gp by 17%-23%.3 In addition, some P-gp-inhibitors known to increase the AUC of orally administered digoxin by ≥25% (e.g., carvedilol, clarithromycin, conivaptan, diltiazem, disopyramide, erythromycin) did not or at best modestly increased the AUC (<20%) of IV administered digoxin, suggesting that these drugs primarily inhibit intestinal P-gp at the doses studied. Collectively, based on the totality of evidence, although a few marketed drugs, such as cyclosporine, quinidine and verapamil elicit systemic P-gp inhibition at therapeutic doses, the DDI risk for canalicular and renal efflux based on systemic exposure to substrates is generally limited in magnitude (<100% increase in substrate AUC), which includes in vitro potent investigational P-gp-inhibitors.
Besides reporting alterations in substrate AUC or systemic clearance, some studies measured urine concentrations of substrates (most for digoxin, with some studies for doxorubicin, edoxaban, and etoposide) and reported renal clearance (CLr) and non-renal clearance (CLnr) of those drugs. In general, the reductions in substrate CLr (range of 9-61% with an average of 32%) in the presence of inhibitors were comparable to the reductions in systemic clearance (range of 22-64% with an average of 37%) but were less than the reductions in CLnr (range of 22-69% with an average of 45%) as shown in Figure 1. This observation may be due to CLr consisting of both P-gp-mediated active renal secretion and glomerular filtration, the latter of which does not involve transporters and thus is not affected by P-gp- inhibitors. CLnr may consist of P-gp-mediated biliary excretion and potentially intestinal secretion, as well as limited metabolism.
In vitro-in vivo extrapolation of systemic P-gp inhibition
The criteria used to predict in vivo DDIs mediated by P-gp inhibition from in vitro data have been evaluated extensively for orally administered substrates.24, 25, 26, 27, 28, 29 However, such analyses have not been conducted for non-orally administered drugs. While the magnitude of DDIs mediated by inhibition of systemic P-gp tends to be limited as described above, the effect can still be clinically significant considering some P-gp-substrates have a narrow therapeutic window (e.g., digoxin, doxorubicin). Thus, DDI risk cannot be ruled out for an investigational drug that inhibits P-gp in vitro and is administered intravenously or via other non-oral routes with systemic drug delivery. Another applicable scenario is for a drug that is administered orally where the metabolite formed post-absorption (i.e., after the parent drug reaches the systemic circulation) inhibits P-gp. Accordingly, we compiled a dataset of IV administered substrates and/or inhibitors by searching the UW DIDB for clinical DDIs involving well-known P-gp-substrates that are minimally metabolized, i.e., dabigatran etexilate (it is a pro-drug of dabigatran which is the measured moiety), digoxin, edoxaban, fexofenadine, and talinolol to avoid possible confounding by inhibition of enzymes (e.g., CYP3A). In the case of doxorubicin which undergoes metabolism by CYP3A, majority of the studies were conducted with designer P-gp inhibitors (i.e., elacridar (GF120918), valspodar (PSC833), zosuquidar (LY335979)) which inhibited CYP3A in vitro at concentrations much higher than their Cmax,u (Cmax,u/Ki <0.02) and thus were determined not able to inhibit hepatic CYP3A in vivo. Two studies were conducted with cyclosporine which had limited inhibitory effects on CYP3A. Only one study was performed with verapamil that is a moderate inhibitor of CYP3A. Thereafter, we evaluated the DDI predictability of two criteria: (1) ratio of total plasma Cmax of a P-gp inhibitor to in vitro P-gp inhibition potency (Cmax/IC50 or Ki ≥0.1) as recommended in the current FDA in vitro DDI guidance,30 and (2) ratio of unbound Cmax to in vitro P-gp inhibition potency (Cmax,u/IC50 or Ki ≥0.02) as recommended in the EMA DDI guideline.31 If the ratio is less than the corresponding threshold, then the risk of an inhibitor causing an in vivo DDI of importance can be considered low, which is usually defined as an increase of 25% or more in the AUC of a substrate in the presence of a P-gp-inhibitor (i.e, ratio of the AUC of the substrate in the presence to absence of inhibitor is ≥1.25), a threshold that was used in some earlier IVIVE analyses.24, 25, 26, 27, 28, 29
The in vitro inhibition potency of a P-gp-inhibitor (most values were reported as IC50) varies depending on experimental systems and among different laboratories.32 To minimize potential bias from a single IC50 or Ki value, we searched the literature reported values for a P-gp-inhibitor via the UW DIDB and calculated the geometric mean of all the available IC50 or Ki values determined with the same substrate as that was used in the clinical DDI studies. For the few cases where no IC50 or Ki was determined using the same substrate as that studied in vivo (e.g., doxorubicin, talinolol), the geometric mean was calculated from all the IC50 or Ki values reported for a perpetrator (e.g., digoxin, elacridar (GF120918), fluvoxamine, simvastatin, sorafenib, verapamil, zosuquidar (LY335979)) with various substrates. As details for the apparent Ki or IC50 determination were not always available, a systematic determination of intrinsic Ki values for the inhibitors was not possible, hence this pragmatic approach was used. Although IC50 values depend on assay conditions and comparing these values among different labs can be challenging, combining all available IC50 values from public databases for meta-analyses is common practice.33 This practice may reduce bias from individual values, with the average value more likely reflecting the true inhibition potency.
The Cmax of an inhibitor was obtained from each clinical DDI study conducted with the P-gp substrate when reported. If Cmax values were not available, the average concentration (Cavg) or trough concentration (Ctrough) of the inhibitor was used because in most cases, the ratio of Cavg or Ctrough to IC50 or Ki exceeded the corresponding threshold; thus, the predictions remained the same as those predicted using Cmax. For studies where inhibitor concentrations were not reported, we searched the primary literature for other pharmacokinetic studies where the same or a similar dosing regimen of the P-gp inhibitor was used and a Cmax was reported. Human plasma protein binding (PPB) information for an inhibitor was obtained from the drug product labelling approved by the US FDA. If this information was not available or the P-gp inhibitor was not approved, PPB values were curated from the literature. Because some drugs are >99% bound to human plasma proteins, PPB in the current analysis was capped at 99%; that is, the lower limit of the unbound fraction in plasma (fu,p) was set at 0.01 to be conservative and consistent with the recommendation in DDI guidance documents from regulatory agencies (FDA, EMA, and PMDA).30, 31, 34
As shown in Table 2 and Figure 2, 57 DDI records (some studies have multiple DDI evaluations) with 26 inhibitors were identified. The majority (n=32) of the studies involved digoxin as the substrate, followed by 22 DDIs involving doxorubicin, 2 involving talinolol, and one involving edoxaban. Twenty-five of these records were considered positive in vivo DDIs in which the substrate AUC increased from 25% to 178%, with a median increase of 55%, in the presence of P-gp inhibitors. When total Cmax/IC50 or Ki ≥0.1 was applied, 23 true positive (TP), 15 true negative (TN), 17 false positive (FP), and 2 false negative (FN) predictions resulted, leading to a high sensitivity (92%) with moderate specificity (47%) (Table 3). The positive predictive error (PPE) and negative predictive error (NPE) were 43% and 12%, respectively. When unbound Cmax,u/IC50 or Ki ≥0.02 was applied, FPs reduced to 7 at the cost of increased FNs to 6 (Table 3). This analysis generated a more balanced sensitivity (76%) and specificity (78%). Accordingly, the PPE was 27% (i.e., a proportion of studies predicted a DDI risk that actually showed no risk), and NPE was 19% (i.e., a proportion of studies predicted as no risk actually demonstrated a DDI risk). Among the 6 FNs, the AUC increase (28%) of the substrate (digoxin) in one study marginally exceeded the threshold to be considered a positive DDI (i.e., 25%) in the presence of propafenone.35 In addition, in that study, the CLr of digoxin decreased by only 9%, indicating that propafenone has a small effect on systemic P-gp. Other FNs involved the inhibitors amiodarone and spironolactone, increasing digoxin AUC by 33% and 47%, respectively. The remaining three FNs involved the inhibitor verapamil, increasing digoxin AUC by 41% or 57%, and increasing doxorubicin AUC by 71%. In addition, we also explored applying a cut-off of 0.1 to Cmax,u/IC50 or Ki, this led to 9 more FNs, i.e., valspodar (PSC833), sorafenib, elacridar (GF120918), and cyclosporine on doxorubicin (AUC increased by 41%-94%, 47%, 47%, and 48%-55%, respectively), and ritonavir and amiodarone on digoxin (AUC increased by 86% and 47%, respectively). The sensitivity and specificity of this higher cut-off were 44% and 90%, respectively, with PPE of 20% and NPE of 35%. Thus, it is considered that the prediction performance becomes worse with this cut-off.
Table 2:
Inhibitor | Plasma protein binding |
Inhibitor concentration (μM) | IC50 or Ki (μM) for P-gp | Cmax/IC50 or Ki |
Prediction category |
Cmax,u/IC50 or Ki |
Prediction category |
AUC ratio of P-gp substrate |
Reference for inhibitor concentration |
|||
---|---|---|---|---|---|---|---|---|---|---|---|---|
Dosing regimen | concentration | geomean | range | substrate | ||||||||
Amiodarone | 96% | 200 mg x 15 d | 1.8 (Cmax)a | 10.6 | 4 - 45.6 | digoxin | 0.17 | FP | 0.007 | TN | 1.13 (digoxin) | 89 |
400 mg x 21-25 d | 2.6 (Cavg)a | > 0.25 | TP | < 0.02 | FN | 1.33 (digoxin) | 90 | |||||
1600 mg qd x14 d | 5.6 (Ctrough) | > 0.53 | TP | > 0.021 | TP | 1.47 (digoxin) | 59 | |||||
Carvedilol | > 98% | 25 mg, single | 0.054 (Cmax)a | 2.53 | 0.15-32 | digoxin | 0.021 | TN | 0.0004 | TN | 0.99 (digoxin) | 91 |
Cimetidine | 18-26% b | 1000 mg/d qid x 4d | 3.96 (C) | > 1000 | digoxin | 0.004 | TN | 0.003 | TN | 1.07 (digoxin) | 92 | |
Clarithromycin | 72% c | 250 mg bid | 1.54 (Cmax) | 21 | 4 - 66 | digoxin | 0.07 | TN | 0.020 | FP | 1.2 (digoxin) | 93 |
200 mg bid x 5 d | 1.34 (Cmax)a | 0.06 | TN | 0.018 | TN | 1 (digoxin) | 94 | |||||
200 mg bid x 8 d | 1.47 (Cmax)a | 0.07 | TN | 0.019 | TN | 1.09 (digoxin) | 94 | |||||
Conivaptan | 99% | 20 mg, single | 0.13 (Cmax) | 25.3 | 16.6-39 | digoxin | 0.005 | TN | 0.00005 | TN | 1.08 (digoxin) | 95 |
Diltiazem | 70-80% | 30 mg qid x 2 wk | 0.17 (Cmax)a | 28.6 | 4.6 - 88.1 | digoxin | 0.006 | TN | 0.0015 | TN | 0.91 (digoxin) | 96 |
30 mg qid x 8 d | 0.17 (Cmax)a | 0.006 | TN | 0.0015 | TN | 1.06 (digoxin) | ||||||
Disopyramide | 50-65% | 200 mg tid x 21 d | 8.54 (C) | 40 | digoxin | ≥ 0.21 | FP | ≥ 0.09 | FP | 0.87 (digoxin) | 97 | |
Erythromycin | 65-90% d | 200 mg qid x 5 d | 1.64 (Cmax)a | 23.2 | 10 - 54.8 | digoxin | 0.07 | TN | 0.016 | TN | 0.96 (digoxin) | 98, 99, 100, 101 |
Fluvoxamine | 80% | 50 mg bid x 17 d | 0.17 (C) | 146.7 | talinolol | 0.001 | TN | 0.0002 | TN | 0.8 (digoxin) | 102 | |
Isradipine | 95% | 2.5 mg bid x 2d, 5 mg bid x 2d, then 5 mg tid x 10 d | 0.014 (Cmax)a | 16.1 | 6 - 36 | digoxin | 0.0008 | TN | 0.00004 | TN | 0.96 (digoxin) | 103 |
Losartan | 98.7% | 50 mg qd x 15 d | 0.53 (Cmax) | 96.7 | 65, 144 | digoxin | 0.005 | TN | 0.00007 | TN | 1.03 (digoxin) | 104 |
Nifedipine | 92-98% | 10 mg tid x 10 d | 0.48 (Cmax) | 71.2 | 22 - 472 | digoxin | 0.007 | TN | 0.0003 | TN | 1.02 (digoxin) | 105 |
Propafenone | >95% | 150 mg tid x 12 d | 1.27 (Cavg) | 11.7 | 6.8, 20.3 | digoxin | > 0.11 | FP | > 0.0054 | TN | 1.14 (digoxin) | 35 |
300 mg tid x 12 d | 3.51 (Cmax)a | 0.30 | TP | 0.015 | FN | 1.28 (digoxin) | 106 | |||||
Quinidine | 80-89% | 200 mg qid x 4 d | 7.71 (Cavg) | 4.45 (2.29 Ki values) | 0.2 - 56 | digoxin | > 1.73 | TP | > 0.27 | TP | 1.57 (digoxin) | 60 |
200 mg qid x 6 d | 7.4 (Cavg) | >1.66 | TP | > 0.26 | TP | 2.78 (digoxin) | 65 | |||||
200 mg qid x 8 d | 6.16 (Ctrough) | > 1.39 | TP | > 0.21 | TP | 1.83 (digoxin) | 64 | |||||
200 mg qid x11 d | 9.86 (Ctrough) | > 2.22 | TP | > 0.34 | TP | 1.65 (digoxin) | 62 | |||||
200 mg bid x26 d | 4.87 (Ctrough) | > 1.09 | TP | > 0.17 | TP | 1.79 (digoxin) | 63 | |||||
600 mg bid x11 d | 10.3 (Cpre-dose) | > 2.31 | TP | > 0.36 | TP | 2.28 (digoxin) | 66 | |||||
Quinine | 80.5% | 200 mg tid x 9d | 7.55 (Cavg) | 7.83 | 3.3, 18.6 | digoxin | > 0.96 | TP | > 0.19 | TP | 1.38 (digoxin) | 67 |
Ritonavir | 98.5% | 300 mg bid x11 d | 19.6 (Cmax) | 7.65 | 5 - 28 | digoxin | 2.56 | TP | 0.038 | TP | 1.86 (digoxin) | 68 |
Rolapitant (IV) | 99.8% | 185 mg,single dose | 3.97 (Cmax) | 7.36 | digoxin | 0.54 | FP | 0.005 g | TN | 1.07 (digoxin, PO) | 107 | |
Spironolactone | > 90% | 100 mg bid x 5 d | 0.18 (Cmax)a | 7.68 | 6.7, 8.8 | digoxin | 0.024 | FN | < 0.0024 | FN | 1.47 (digoxin) | 108 |
Verapamil | 90% | 80 mg bid x 4d, then tid x 10d | 0.51 (Cmax)a | 4.53 (4.76, Ki values) | 0.06 - 224 | digoxin | 0.11 | FP | 0.011 | TN | 1.24 (digoxin) | 36 |
80 mg tid x 10 d | 0.51 (Cmax)a | 0.11 | TP | 0.011 | FN | 1.57 (digoxin) | 36 | |||||
120 mg tid x12 d | 0.63 (Cmax)a | 0.14 | TP | 0.014 | FN | 1.41 (digoxin) | 109 | |||||
Cyclosporine (IV) | 90% | 18 mg/kg/d | 3.15 (Cavg) | 3.66 | doxorubicin | > 0.86 | TP | > 0.086 | TP | 1.55 (doxorubicin) | 73 | |
38.9 mg/kg over 50 hr | 1.42 (C24hr) | > 0.39 | TP | > 0.039 | TP | 1.48 (doxorubicin) | 74 | |||||
Elacridar (GF120918) | >99.9% e | 50 mg daily | 0.06 (Cmax) | 0.077 (0.307, Ki values) | 0.0156 - 0.8 | various substrates | 0.79 | FP | 0.008 g | TN | 0.86 (doxorubicin) | 20 |
50 mg bid | 0.10 (Cmax) | 1.24 | FP | 0.012 g | TN | 1.03 (doxorubicin) | ||||||
100 mg bid | 0.07 (Cmax) | 0.91 | FP | 0.009 g | TN | 0.88 (doxorubicin) | ||||||
200 mg bid | 0.14 (Cmax) | 1.83 | FP | 0.018 g | TN | 1.23 (doxorubicin) | ||||||
400 mg bid | 0.24 (Cmax) | 3.16 | FP | 0.032g | FP | 1.20 (doxorubicin) | ||||||
400 mg bid | 0.35 (Cmax) | 4.5 | FP | 0.045 g | FP | 1.04 (doxorubicin) | ||||||
400 mg bid | 0.31 (Cmax) | 4.1 | TP | 0.041g | TP | 1.47 (doxorubicin) | ||||||
Sorafenib | 99.5% | 100 mg bid x19 d | 3.14 (Cmax) | 4.56 | 0.84 - 25 | various substrates | 0.69 | FP | 0.007 g | TN | 1.1 (doxorubicin) | 110 |
200 mg bid x19 d | 8.95 (Cmax) | 1.96 | FP | 0.0196 g | TN | 0.85 (doxorubicin) | ||||||
400 mg bid x19 d | 7.94 (Cmax) | 1.74 | FP | 0.017 g | TN | 1.01 (doxorubicin) | ||||||
400 mg bid x19 d | 16.97 (Cmax) | 3.72 | TP | 0.037 g | TP | 1.47 (doxorubicin) | ||||||
Valspodar (PSC833) (IV or PO) | 97.8% f | 1-10 mg/kg, single dose | 1.42 - 2.05 (Cmax) | 0.291 | doxorubicin | 4.88 - 7.06 | TP | 0.107 - 0.155 | TP | 1.49 - 2.69 (doxorubicin) | 77 | |
1.25 - 12.5 mg/kg bid for 5d | ≥0.65 (Cmax) | ≥ 2.24 | TP | ≥ 0.049 | TP | 1.54 (doxorubicin) | 80 | |||||
2.5 mg/kg bid for 7d | 0.93 (Cmax) | 3.21 | TP | 0.07 | TP | 1.94 (doxorubicin) | 78 | |||||
0.42 mg/kg/hr x 3 d | 0.82 (Css) | > 2.82 | TP | > 0.062 | TP | 1.41 (doxorubicin) | 111 | |||||
Verapamil | 90% | 80 mg tid x 3d then 120 mg qid x 4d | 0.35 (Cmax) | 8.27 (6.38, Ki values) | 0.06 - 446.5 | various substrates | 0.043 | FN | 0.0043 | FN | 1.71 (doxorubicin) | 72 |
Zosuquidar (LY335979) (IV or PO) | 99.95% d | 80 - 640 mg/m2/d x2d | 0.49 (Cmax) | 0.025 | 0.0002 - 0.38 | various substrates | 19.5 | TP | 0.19 g | TP | 1.25 (doxorubicin) | 21 |
480-640 mg/m2/d x2d | 0.96 (Cmax) | 38.7 | FP | 0.39 g | FP | 1.15 (doxorubicin) | ||||||
160-320 mg/m2 bid | 0.33 (Cmax) | 13.4 | FP | 0.134 g | FP | 1 (doxorubicin) | 112 | |||||
300-400 mg/m2 tid | 0.41 (Cmax) | 16.5 | FP | 0.165 g | FP | 0.97 (doxorubicin) | ||||||
Quinidine | 84.5% | 300 mg tid x 4 d | 3.54 (Ctrough) | 0.898 | edoxaban | > 3.95 | TP | > 0.61 | TP | 1.35 (edoxaban) | 113 | |
Digoxin | 25% | 0.5 mg, single dose | 0.003 (Cmax) | 94 | 7 - 393 | various substrates | 0.00003 | TN | 0.00002 | TN | 0.95 (talinolol) | 114 |
Simvastatin | 95% | 40 mg qd x 21 d | 0.013 (Cmax) | 25.4 | 4.6 - 209 | various substrates | 0.0005 | TN | 0.00003 | TN | 1 (talinolol) | 115 |
Substrates were administered intravenously, and inhibitors were given orally, unless otherwise indicated. Plasma protein binding is obtained from drug product prescriber information (U.S. labeling), unless otherwise indicated.
Inhibitor concentration was not reported in the DDI study conducted with P-gp-substrates. Thus, inhibitor concentration was obtained from other PK studies with same or similar dosing regimen(s) as the that used in the DDI study with P-gp-substrates. The sources of PK studies are provided in references.
Plasma protein binding of cimetidine. 116
Plasma protein binding of valspodar (PSC833).124
Free fraction of drug in plasma (fu,p) was capped as 0.01 to calculate Cmax,u.
Table 3:
Total records | TN | TP | FN | FP | Sensitivity | Specificity | PPE | NPE | |
---|---|---|---|---|---|---|---|---|---|
total Cmax/IC50 (0.1 as cut-off) | 57 | 15 | 23 | 2 | 17 | 92% | 47% | 43% | 12% |
unbound Cmax/IC50 (0.02 as cut-off) | 57 | 25 | 19 | 6 | 7 | 76% | 78% | 27% | 19% |
TN: true negative; TP: true positive; FN: false negative; FP: false positive; Sensitivity: TP/(TP + FN), proportion of actual positive DDIs that were predicted as such; Specificity: TN/(TN+ FP), proportion of actual negative or no DDI risks that were predicted as such; PPE: positive predictive error, FP/(FP + TP), proportion of studies that were conducted that actually did not need to be (no DDI risk); NPE: negative predictive error, FN/(FN+ TN), proportion of studies that were predicted as no risk but actually demonstrated DDI risk.
This analysis only considered P-gp inhibition by parent drugs. However, norverapamil, a metabolite with comparable in vivo plasma concentrations as verapamil,36 also inhibits P-gp and appears to be more potent than the parent drug. One study examining the effect of verapamil and its metabolites on digoxin transport reported an IC50 for norverapamil and verapamil of 0.3 μM and 1.1 μM, respectively.37 The IC50 of verapamil is lower than the geometric mean value (4.5 μM) used in the current analysis. Thus, when using 1.1 μM alone, Cmax,u/IC50 exceeded 0.02 and corrected the two FNs with digoxin to TPs. However, adding norverapamil can resolve the FNs if using 4.5 μM as the inhibition potency for verapamil. Norverapamil was also incorporated in a physiologically based pharmacokinetic (PBPK) model used to predict the effect of verapamil on several substrates including digoxin. 38
There is large variability in inhibition potency of P-gp inhibitors. Attempts to reduce this variability by deriving an experimental system-independent Ki for P-gp-mediated digoxin transport from system-dependent IC50 data have been reported. The derived Ki values represent the dissociation constant of an inhibitor relative to the aqueous concentration of the inhibitor in the cytosol of cell monolayers. The derivation required fitting a kinetic model with in vitro transport data to estimate kinetic rate constants of digoxin, which were further used to derive Ki. Using this approach, more consistent Ki values (0.3 or 0.6 μM) were estimated for verapamil from digoxin transport data from several laboratories.39 In contrast, the IC50 values of verapamil calculated using the same experimental data were 1.8, 8.6, 11.8, and 32.3 μM. In that study, system-independent Ki values (0.2-0.3 μM) were also estimated for quinidine and carvedilol. As a comparison, the corresponding IC50 values for quinidine were 2.3, 8.4, and 15.7 μM, and those for carvedilol were 0.7, 1.4, 6.8, and 8.1 μM.39 When this system-independent Ki for verapamil (0.5 μM on average) was used to calculate Cmax,u/Ki, the ratios previously resulting in FN predictions were increased from 0.011, 0.014, 0.0043 to 0.1, 0.126, 0.07, respectively, converting the FN to TP predictions. Using the system-independent Ki, the predictions remained the same for quinidine (TPs) and for carvedilol (TN). Although using the model-derived Ki appeared to mitigate some FNs associated with the criterion of Cmax,u/Ki ≥0.02, this observation is based on limited data of only one inhibitor (verapamil). It should be noted that derivation of system-independent Ki values requires more research and model fitting. During drug discovery and development, such analyses rarely have been conducted. However, if derivations of such Ki values for other inhibitors result in fewer FN predictions with similar FP predictions, then the expenditure of resources to update Ki values to make predictions may prove to be a cost-effective way to minimize risk to patients and avoid unnecessary clinical studies.
A related question is whether such an evaluation is also needed for orally administered drugs. Currently, the regulatory guidances from EMA, FDA, and PMDA recommend calculating [I]/IC50 or Ki, where [I] is dose/250 mL as a rough estimation of intestinal drug concentration and compare this ratio with a cut-off of 10. If the ratio is 10 or above, an inhibitor may have the potential to inhibit P-gp in vivo and a further clinical DDI study may be needed. Previously, two criteria were recommended, i.e., [I1]/IC50 ≥0.1, where [I1] is total Cmax of an inhibitor at its therapeutic dose, and [I2]/IC50 ≥10, where [I2] is dose/250 mL. The former reflects systemic inhibition of P-gp, while the latter represents inhibition of intestinal P-gp. A recent analysis of 53 orally administered drugs (as perpetrators) demonstrated that the predictions (TNs, TPs, FNs, FPs) are identical between the criterion only considering [I2]/IC50 ≥10 and the combined criteria [I1]/IC50 ≥0.1 or [I2]/IC50 ≥10, and also are identical using the criterion [I1,u]/IC50 ≥0.02 or [I2]/IC50 ≥10, where [I1,u] is unbound Cmax of an inhibitor at its therapeutic dose.29 This indicates that P-gp inhibition-mediated DDIs for orally administered drugs may be driven mainly by intestinal P-gp inhibition, which is not surprising because orally administered drugs tend to have high intracellular concentrations in intestinal cells that are more likely to reach the level sufficient to inhibit P-gp than systemic drug concentrations. Thus, the current regulatory guidances recommend only [I2]/IC50 ≥10 for orally administered drugs. We also examined the datasets from other publications of IVIVE analyses for orally administered P-gp inhibitors.25, 26, 27 There are rare cases where an inhibitor has an [I2]/IC50 <10 but [I1,u]/IC50 ≥ 0.02, which are conivaptan, montelukast, omeprazole, and pantoprazole only when the higher IC50 values of these drugs towards P-gp are used for calculations. Among these, only conivaptan increased AUC of digoxin more than 25% (by 43%).26 The prediction changes depending on the IC50. If a geometric mean of IC50 values is used, the [I2]/IC50 of conivaptan exceeds 10. Overall, the vast majority of clinical P-gp inhibitors orally administered will be captured with the criterion just considering [I2] (dose/250 mL).
In summary, the analysis based on the dataset we compiled suggests that for inhibition of systemic P-gp, using total Cmax/IC50 or Ki ≥0.1 has reasonable control on FN prediction but also leads to substantial FP prediction. In contrast, using unbound Cmax,u/IC50 or Ki ≥0.02 reduces FPs but has a higher risk of FN predictions. Considering the overall limited magnitude of DDIs mediated by inhibition of systemic P-gp, using Cmax,u may provide an improved balance between capturing significant DDIs and reducing the cost of conducting unnecessary clinical studies. The decision whether to conduct a clinical study with a P-gp substrate and an investigational drug that is administered by a non-oral route with systemic drug exposure should take into consideration the likelihood of concomitant use of the drug with a P-gp substrate and the safety margin of that substrate. In addition, when there is a major metabolite (i.e., its radioactivity accounts for >10% of total drug-related material) with exposure comparable to parent drug, the potential of the metabolite inhibiting P-gp should be considered; if the metabolite is indeed a P-gp-inhibitor, the metabolite should be considered when predicting DDI risk. Lastly, further research is warranted to explore approaches to reduce the variability of inhibitory potency determinations from in vitro experiments. One potential approach is to derive a system-independent Ki for an inhibitor and assess prediction performance.
Evaluation of hepatic and renal clinical DDIs that potentially involve systemic BCRP inhibition
To collect clinical data for hepatic/renal BCRP inhibitors, we evaluated clinical DDI reports from the UW DIDB that met the following criteria: (1) BCRP inhibition is listed as a potential mechanism of a DDI, (2) either victim or perpetrator (or both) were administered intravenously, (3) prospectively designed DDI study, and (4) >25% increase in victim systemic exposure in the presence of the inhibitor (Table 4). Furthermore, DDI studies involving the victim drug rosuvastatin and perpetrators rifampicin or cyclosporine A (CsA) were excluded, as these DDIs were likely due primarily to inhibition of hepatic OATP1B and intestinal BCRP. As a result, only two studies showed potential involvement of hepatic BCRP inhibition between a victim and perpetrator, respectively: 1) copanlisib and itraconazole 40 and 2) irinotecan and regorafenib.41 However, both DDIs were relatively small in magnitude (AUC increase of 58% and 28%, respectively), and the involvement of other elimination pathways cannot be excluded, e.g., copanlisib is metabolized by CYP3A. Similarly, drugs that are known BCRP inhibitors based on in vitro studies were reported to increase methotrexate exposure by >20%, although the actual increase in methotrexate AUC was not reported (Table 4). Methotrexate is a substrate of additional transporters, including MRP2, OAT1 and OAT3. In vitro inhibitors of BCRP such as vindesine also inhibit MRP2, OAT1 and OAT3.42 Therefore, based on currently available data, there do not appear to be any drugs in clinical use with evidence of DDIs involving systemic inhibition of hepatic/renal BCRP. There is lack of clinical evidence for the involvement of BCRP in the renal clearance of substrate drugs, consistent with the low protein expression of BCRP in the human kidney cortex.5 It is important to highlight that the lack of clinical evidence does not prove that there is not a DDI, particularly considering that changes in hepatic or renal cell concentrations may not be reflected in systemic concentrations. Furthermore, there may be simultaneous inhibition of uptake transporters, as discussed with the PET imaging study involving an intravenous infusion of CsA43 and oral administration of rifampicin.44
Table 4:
Overall Effect | Object | Object dosing route |
Precipitant | Precipitant dosing route |
Percent Change AUC |
Percent Change Plasma Concentration |
Concentration Type |
Comments | Reference |
---|---|---|---|---|---|---|---|---|---|
In Vivo Inhibition > 20% Effect | Copanlisiba | IV | itraconazole | Oral | 57.7 | 3.9 | Cmax | In vitro studies suggest that copanlisib is a substrate of P-gp and BCRP. According to the sponsor, the contribution of P-gp and BCRP inhibition to the increase of copanlisib exposure observed when co-administered with itraconazole is likely to be small as copanlisib was administered by IV infusion. | 40 |
In Vivo Inhibition > 20% Effect | irinotecan | IV | regorafenib | Oral | 28 | 22 | Cmax | BCRP likely contributes to the transport of irinotecan. Regorafenib inhibits BCRP in vitro. | 41 |
In Vivo Inhibition > 20% Effect | methotrexate | IV | proton pump inhibitors | Oral | -- | 103.2 | Cavg | The proposed mechanisms for the interaction may involve inhibition of breast cancer resistance protein, which is also involved in mediating MTX transport. | 125 |
In Vivo Inhibition > 20% Effect | methotrexate | IV | vindesine | IV | -- | 133.3 | -- | Membrane transport of methotrexate involves multiple transporters including the renal uptake transporters OAT1 and OAT3 in tubular cells, and a series of efflux transporters including MRP2 and BCRP. Vindesine is an inhibitor of BCRP, MRP2, OAT1, and OAT3. | 42 |
IV – Intravenous
Copanlisib is metabolized by CYP3A and transported by P-gp. Itraconazole inhibits CYP3A and P-gp.
P-gp- and BCRP-mediated inhibition of biliary clearance
Biliary excretion of drugs and their metabolites is a critical, yet often overlooked, route of hepatic elimination. Drugs or metabolites excreted from bile into the intestine may undergo further metabolism, exhibit pharmacologic or toxicologic effects, undergo fecal excretion, and/or be absorbed/reabsorbed into the systemic circulation. Canalicular (apical) transport proteins, including P-gp, BCRP, MRP2, BSEP, and MATE1 are primarily responsible for the biliary excretion of drugs/metabolites. Impaired biliary clearance of endogenous substrates or drugs due to alterations in canalicular transporters by genetic polymorphisms,45 DDIs,46, 47 or disease48 may increase hepatic and/or systemic exposure to substrates. Increased hepatocellular concentrations may impact drug efficacy and/or toxicity if the target site resides within hepatocytes. Importantly, systemic concentrations may not accurately reflect hepatocyte concentrations if hepatic transporters govern substrate disposition. Multiple canalicular transporters can excrete a substrate into bile (e.g., glucuronide conjugates). Loss of function of one transporter may result in biliary excretion or basolateral efflux of the substrate by an alternate transporter, which can shift the route of hepatic excretion from bile (e.g., via MRP2) to blood (e.g., via MRP3), or result in unexpected DDIs due to interactions with the alternate transporter(s). Evaluation of this interplay in humans would be technically difficult and requires extensive sampling from sites where catheters are not routinely placed (i.e., hepatic vein and bile duct). Such complex interactions could be simulated using mechanistic or victim PBPK models to evaluate the potential impact. However, fundamental information is required for accurate predictions of drug disposition, including a reasonable estimate of biliary clearance (CLbile). Direct measurement of CLbile in humans is challenging due to the anatomical location of the gallbladder and common bile duct. Numerous methods have been used to evaluate human CLbile, including quantitative and semi-quantitative in vivo and in vitro approaches.49 Qualitative characterization of biliary disposition using the Entero-Test,50 or fecal recovery after IV administration, also could be used to estimate CLbile assuming that the substrate does not undergo enterohepatic recycling or intestinal secretion into the lumen.
An extensive literature search was conducted to identify drugs that are P-gp and/or BCRP substrates that undergo biliary excretion in humans. Data were reported (Table 5) if the drug was administered IV and the biliary excretion of drug/metabolite(s) was measured directly (via a bile duct catheter), estimated from intestinal sampling via an oroenteric tube, or based on fecal recovery. Data based on fecal recovery were reported only if there was no evidence for intestinal secretion from the systemic circulation into the gut lumen via enterocytes and no significant enterocyte or luminal metabolism, which could reduce the fecal recovery of parent drug. Data obtained for drugs administered orally were excluded due to potential confounding of intestinal P-gp and/or BCRP on overall drug disposition and biliary excretion. When total systemic clearance after IV administration (CLiv) was reported, CLbile was estimated as the product of CLiv and the fraction of the dose recovered in bile (fbile). Drugs with a reported CLbile after IV administration were evaluated further using the UW DIDB for P-gp- and/or BCRP-mediated DDIs.
Table 5:
Transporter | Compound | CLiv data | CLiv [L/h] |
Human CLbile |
CLr [L/h] | Other | Comments | Reference | |||
---|---|---|---|---|---|---|---|---|---|---|---|
[Yes/No] | [Yes/No] | CLbile [L/h] | % of dose excreted in bile or feces |
CLmetabolic [L/h] |
CLnon-renal [L/h] |
||||||
BCRP | Azlocillin | Yes | 8.4-22.5 | Yes | 5.03-9.01 | 0.08-1.55 | 1.1-6.9 (unchanged parent in bile) | 1.00-18.9 | -- | Duodenal perfusion (n=5 healthy volunteers) | 126 |
BCRP, P-gp, MRP2 | Ceftriaxone | Yes | 0.54-1.2 | Yes | 0.24-0.78 | 0.06-0.78 | 11-65 total | -- | -- | Duodenal perfusion for 6-8 hr (n=5 healthy volunteers) | 127 |
BCRP, P-gp | Copanlisib | Yes | 14.2-40.8 | No | 3.72 (active renal secretion) | 30 (unchanged parent in feces) | -- | -- | 41% of dose metabolized with 15% excreted unchanged in urine | 128 | |
P-gp | Digoxin | Yes | 10.5-24.3 | Yes | 6.12-11.9 | 3.48-20.6 | -- | -- | Intestinal perfusion (n=6 healthy volunteers) | 129, 130 | |
Elsamitrucin | Yes | 17.0-55.0 | Yes | 3.74-12.1 | ~22 (unchanged parent in bile) | -- | -- | Indwelling biliary catheter (n=1 cancer patient) | 131 | ||
P-gp, MRP2 | Etoposide | Yes | 2.05-2.96 | Yes | 0.70-0.95 | 0.019 | <2 (unchanged parent in bile) | -- | 1.30-1.52 L/h | Biliary drainage (n=2 pediatric cancer patients); 16 or 35 cancer patients | 51, 132 |
BCRP, P-gp, MRP2 | Fluvastatin | Yes | 67.9 | No | 1.8 (unchanged parent in feces) | -- | -- | -- | 133 | ||
BCRP, P-gp, MRP2 | Irinotecan | Yes | 24.8 | Yes | 6.18 | 5.25 | 18.5 (unchanged parent in bile) | -- | -- | Bile duct exteriorized (n=1 patient) | 134 |
Mezlocillin | Yes | 11.2-28.6 | Yes | 5.58-17.1 | 2.75-7.37 | 23.9-50.5 (unchanged parent in bile) | 0.36-8.58 | -- | Duodenal perfusion (n=5 healthy volunteers) | 126 | |
BCRP, P-gp | Nemiralisib | Yes | 10.8 | Yes | 0.93 | ND | 52.4 (total radioactivity in bile as unchanged parent) | -- | -- | Entero-Test string device (n=6 healthy men) | 135 |
Piperacillin | Yes | 12.2-16.6 | Yes | 5.79-8.78 | 0.10-0.13 | 0.7-1.1 (unchanged parent in bile) | -- | -- | T-tube (n=5 cholecystectomized patients); duodenal aspiration (n=3 healthy volunteers) | 136, 137 | |
BSEP, MRP2, (BCRP) | Pravastatin | Yes | 56.7 | No | 26.5 | 23 (unchanged parent in feces) | -- | 30.2 | Clrenal is much higher than GFR, indicating active tubular secretion | 138 | |
Quinupristin/Dalfopristin | Yes | ~60/~60 | No | <15 (unchanged in feces) | -- | -- | -- | 139 | |||
P-gp, BCRP | Revefenacin | Yes | ? | No | 54 (radiolabeled dose in feces) | -- | -- | 27% of total radioactivity recovered in urine; 19% of radioactive dose recovered in feces as active metabolite | 140 | ||
BCRP, P-gp | Tacrolimus | Yes | 2.25 | No | <0.001 | 0.29 (unchanged parent in feces) | -- | -- | -- | 141, 142 | |
MPR2 | Tc-99m Mebrofenin | Yes | 72.7 | Yes | <1.5 | 67.6 | 84.2 (unchanged parent in bile) | -- | -- | Duodenal aspiration (n=4 healthy volunteers) | 143 |
P-gp | Tc-99m Sestamibi | Yes | 108 | Yes | 25.2 | 21.5 (unchanged parent in bile) | -- | -- | Duodenal aspiration (n=7 healthy volunteers) | 144 | |
Temocillin | Yes | ND | Yes | ND | ND | 2.2 (unchanged parent in bile) | -- | -- | Duodenal perfusion (n=6 healthy volunteers); 38% of dose excreted as parent in urine | 145 | |
MRP2 | Valsartan | Yes | 2.18 | Yes | 0.48 | 1.93 | ~89 (unchanged parent in bile) | -- | -- | Biliary-enteric diversion (n=1 patient) | 146 |
P-gp | Vincristine | Yes | 19.8 | Yes | ND | ND | 28.4 (total radioactivity in bile) | -- | -- | T-tube (n=1 patient with jejunostomy); 26.2% of radioactivity excreted in urine | 147, 148 |
ND, not determined
A total of 86 drugs identified in the initial literature search were reported to undergo a measurable extent of biliary excretion in humans, but in most cases, oral administration confounded data interpretation. Several reports attributed fecal recovery of drug to biliary excretion even when the drug was administered orally and fecal excretion due to unabsorbed drug could not be excluded. Fecal recovery of radioactivity often was attributed to biliary excretion without consideration of metabolic stability of the parent drug. CLIV data were reported for only 20 of the 86 drugs. Of these 20 drugs, CLbile was reported, or could be determined, for 11 (Table 5). CLnr was calculated for some drugs (e.g., etoposide), but insufficient data were available to estimate the fraction of CLnr that could be attributed to CLbile. For example, CsA 51 and valspodar (PSC833) 52 decreased CLnr of IV administered etoposide by 52% and 48%, respectively; however, the contribution of metabolism- vs. transporter-mediated inhibition to the overall decrease in CLnr could not be ascertained from the data provided. Among the nine drugs with a CLbile to CLiv ratio of at least 0.20, relevant DDIs attributed to P-gp- and/or BCRP-mediated inhibition of biliary excretion were identified for only digoxin and etoposide. A CLbile to CLiv ratio of at least 0.2 was selected as a more conservative, arbitrary cut-off based on the 2020 FDA Guidance that defines an elimination pathway (e.g., renal clearance, biliary secretion) as significant when that clearance pathway contributes ≥25% to total drug clearance.
A major knowledge gap exists in understanding the contribution of P-gp- and/or BCRP-mediated CLbile to the overall (systemic) clearance of drugs. Thus, the ability to accurately predict the clinical impact of co-administered P-gp and/or BCRP inhibitors on the systemic disposition of victim drugs is limited. Drugs with a CLbile to CLiv ratio of at least 0.2 may be susceptible to canalicular transporter mediated DDIs; such an observation may be assessed in clinical studies if feasible. Alternatively, the magnitude of the potential impact of impaired CLbile on the hepatic and systemic exposure to the victim drug during co-administration of P-gp and/or BCRP inhibitors could be simulated using PBPK modelling. Innovative approaches to estimate CLbile of drugs after IV administration in clinical studies can be implemented.49, 53 When possible, metabolite(s) should be differentiated from parent drug when fecal recovery of radioactivity is reported. BCRP-mediated transport of glucuronide conjugates has been documented in the literature.54 In the case of asciminib,55 the human mass balance study estimated that 28-58% of total drug clearance can be attributed to the glucuronidation pathway. In vitro studies with hepatocyte co-cultures have provided some evidence to support the biliary excretion of both asciminib and its glucuronide conjugate. However, analysis of human fecal samples did not show the presence of glucuronide metabolites likely due to hydrolysis by gut flora. The inability to directly attribute a specific transporter as the contributor to the active biliary secretion in humans is challenging as is the interpretation for the inhibition of such pathways by co-medications in vivo. Finally, fecal recovery of drug/metabolite(s) after oral administration should not be attributed necessarily to biliary excretion unless complete absorption of the drug has been confirmed.
Conclusion and Future Perspective
This perspective supports limited clinical DDI risk from P-gp or BCRP inhibition in liver or kidneys based on systemic exposure changes for P-gp substrates and limited examples of BCRP substrate drugs. These observations are consistent with the ITC’s previous review of PET- and/or CNS activity-focused clinical studies of blood-brain barrier P-gp inhibition that consistently showed no to negligible enhancement in CNS exposure to P-gp substrates.3 As shown by the dataset we compiled, the increases in systemic concentrations of P-gp substrates due to inhibition of liver and kidney P-gp are usually less than 2-fold. However, because some P-gp substrates have narrow therapeutic windows (e.g., digoxin, doxorubicin), the DDI risk of a P-gp-inhibitor that is administered by a non-oral route with systemic drug delivery cannot be ruled out. Such a potential DDI can be evaluated by comparing the ratio of unbound Cmax divided by IC50 or Ki to a cut-off of 0.02. If the ratio is less than the cut-off, then the risk can be considered as low. If not, further evaluation is warranted and the decision on whether to follow up with a clinical DDI study will depend on the likelihood of concomitant use of the inhibitor with P-gp-substrates, whether there are other interacting mechanisms besides P-gp inhibition, and the safety margin of P-gp-substrates. Such an evaluation seems unnecessary for orally administered drugs because intestinal P-gp inhibition is the main driving force in those situations. This is supported by our previous analysis showing that considering only estimated intestinal concentrations of an orally administered P-gp-inhibitor is sufficient to provide a reasonable prediction of an in vivo DDI. It should be noted that inhibition of P-gp or BCRP in liver and/or kidney may not always be reflected by the systemic exposure to substrates. Modelling and simulation approaches involving IVIVE-linked PBPK models may be employed to predict the hepatic/renal cell concentration of the P-gp/BCRP substrate/inhibitor drugs and establish correlations with their systemic, urinary, and/or pharmacodynamic effects if suitable.56, 57
Supplementary Material
Acknowledgements
The authors are grateful to Drs A David Rodrigues, Manthena V S Varma, Mitchell E Taub, Donna A. Volpe and Elimika Pfuma Fletcher for their valuable comments.
Funding.
This work was supported by the National Institutes of Health National Institute of General Medical Sciences (Grant No. R35 GM122576) and the National Institutes of Health National Center for Complementary and Integrative Health (Grant No. U54 AT008909) to K.L.R.B and M.F.P., respectively.
Abbreviations
- AUC
area under the concentration vs. time curve
- AUCR
ratio of the AUC of the substrate in the presence to absence of perpetrator
- BCRP
breast cancer resistance protein
- DDI
drug-drug interaction
- FN
false negative
- FP
false positive
- IVIVE
in vitro-to-in vivo extrapolation
- MDR
multidrug resistance
- MRP
multidrug resistance-associated protein
- NPE
negative predictive error
- OATP
organic anion transporting polypeptide
- P-gp
P-glycoprotein
- PBPK
physiologically based pharmacokinetic
- PPB
plasma protein binding
- PPE
positive predictive error
- TN
true negative
- TP
true positive
- UW DIDB
University of Washington Drug Interaction Database
Footnotes
Conflict of Interest Statement: The authors declared no competing interests for this work. The views in this paper are those of the authors and should not be construed to represent FDA’s views or policies.
SUPPORTING INFORMATION
Supplementary information accompanies this paper on the Clinical Pharmacology & Therapeutics website (www.cpt-journal.com).
References
- 1.Yan H, Du X, Wang R, Zhai G. Progress in the study of D-alpha-tocopherol polyethylene glycol 1000 succinate (TPGS) reversing multidrug resistance. Colloids Surf B Biointerfaces 205 111914. (2021) [DOI] [PubMed] [Google Scholar]
- 2.Smolinski MP, et al. Discovery of Encequidar, First-in-Class Intestine Specific P-glycoprotein Inhibitor. J Med Chem 64 3677–3693. (2021) [DOI] [PubMed] [Google Scholar]
- 3.Kalvass JC, et al. Why clinical modulation of efflux transport at the human blood-brain barrier is unlikely: the ITC evidence-based position. Clin Pharmacol Ther 94 80–94. (2013) [DOI] [PubMed] [Google Scholar]
- 4.Williams JA, et al. Drug-drug interactions for UDP-glucuronosyltransferase substrates: a pharmacokinetic explanation for typically observed low exposure (AUCi/AUC) ratios. Drug Metab Dispos 32 1201–1208. (2004) [DOI] [PubMed] [Google Scholar]
- 5.Prasad B, et al. Abundance of Drug Transporters in the Human Kidney Cortex as Quantified by Quantitative Targeted Proteomics. Drug Metab Dispos 44 1920–1924. (2016) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Kim RB, et al. Interrelationship between substrates and inhibitors of human CYP3A and P-glycoprotein. Pharm Res 16 408–414. (1999) [DOI] [PubMed] [Google Scholar]
- 7.Liu-Kreyche P, et al. Lysosomal P-gp-MDR1 Confers Drug Resistance of Brentuximab Vedotin and Its Cytotoxic Payload Monomethyl Auristatin E in Tumor Cells. Front Pharmacol 10 749. (2019) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Zamek-Gliszczynski MJ, Hoffmaster KA, Tweedie DJ, Giacomini KM, Hillgren KM. Highlights from the International Transporter Consortium second workshop. Clin Pharmacol Ther 92 553–556. (2012) [DOI] [PubMed] [Google Scholar]
- 9.Kalvass JC, Graff CL, Pollack GM. Use of loperamide as a phenotypic probe of mdr1a status in CF-1 mice. Pharm Res 21 1867–1870. (2004) [DOI] [PubMed] [Google Scholar]
- 10.Zamek-Gliszczynski MJ, Bedwell DW, Bao JQ, Higgins JW. Characterization of SAGE Mdr1a (P-gp), Bcrp, and Mrp2 knockout rats using loperamide, paclitaxel, sulfasalazine, and carboxydichlorofluorescein pharmacokinetics. Drug Metab Dispos 40 1825–1833. (2012) [DOI] [PubMed] [Google Scholar]
- 11.Durmus S, et al. In vivo disposition of doxorubicin is affected by mouse Oatp1a/1b and human OATP1A/1B transporters. Int J Cancer 135 1700–1710. (2014) [DOI] [PubMed] [Google Scholar]
- 12.Lee HH, Leake BF, Kim RB, Ho RH. Contribution of Organic Anion-Transporting Polypeptides 1A/1B to Doxorubicin Uptake and Clearance. Mol Pharmacol 91 14–24. (2017) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Burger H, et al. Imatinib mesylate (STI571) is a substrate for the breast cancer resistance protein (BCRP)/ABCG2 drug pump. Blood 104 2940–2942. (2004) [DOI] [PubMed] [Google Scholar]
- 14.Honjo Y, et al. Acquired mutations in the MXR/BCRP/ABCP gene alter substrate specificity in MXR/BCRP/ABCP-overexpressing cells. Cancer Res 61 6635–6639. (2001) [PubMed] [Google Scholar]
- 15.Benderra Z, et al. Breast cancer resistance protein and P-glycoprotein in 149 adult acute myeloid leukemias. Clin Cancer Res 10 7896–7902. (2004) [DOI] [PubMed] [Google Scholar]
- 16.Abbott BL, et al. Low levels of ABCG2 expression in adult AML blast samples. Blood 100 4594–4601. (2002) [DOI] [PubMed] [Google Scholar]
- 17.Maeda K, Sugiyama Y. Transporter biology in drug approval: regulatory aspects. Mol Aspects Med 34 711–718. (2013) [DOI] [PubMed] [Google Scholar]
- 18.Seiden MV, et al. A phase II study of the MDR inhibitor biricodar (INCEL, VX-710) and paclitaxel in women with advanced ovarian cancer refractory to paclitaxel therapy. Gynecol Oncol 86 302–310. (2002) [DOI] [PubMed] [Google Scholar]
- 19.Sparreboom A, et al. Clinical pharmacokinetics of doxorubicin in combination with GF120918, a potent inhibitor of MDR1 P-glycoprotein. Anticancer Drugs 10 719–728. (1999) [DOI] [PubMed] [Google Scholar]
- 20.Planting AS, et al. A phase I and pharmacologic study of the MDR converter GF120918 in combination with doxorubicin in patients with advanced solid tumors. Cancer Chemother Pharmacol 55 91–99. (2005) [DOI] [PubMed] [Google Scholar]
- 21.Sandler A, et al. A Phase I trial of a potent P-glycoprotein inhibitor, zosuquidar trihydrochloride (LY335979), administered intravenously in combination with doxorubicin in patients with advanced malignancy. Clin Cancer Res 10 3265–3272. (2004) [DOI] [PubMed] [Google Scholar]
- 22.Le LH, et al. Phase I study of the multidrug resistance inhibitor zosuquidar administered in combination with vinorelbine in patients with advanced solid tumours. Cancer Chemother Pharmacol 56 154–160. (2005) [DOI] [PubMed] [Google Scholar]
- 23.Dieras V, et al. Phase I combining a P-glycoprotein inhibitor, MS209, in combination with docetaxel in patients with advanced malignancies. Clin Cancer Res 11 6256–6260. (2005) [DOI] [PubMed] [Google Scholar]
- 24.Cook JA, et al. Refining the in vitro and in vivo critical parameters for P-glycoprotein, [I]/IC50 and [I2]/IC50, that allow for the exclusion of drug candidates from clinical digoxin interaction studies. Mol Pharm 7 398–411. (2010) [DOI] [PubMed] [Google Scholar]
- 25.Fenner KS, et al. Drug-drug interactions mediated through P-glycoprotein: clinical relevance and in vitro-in vivo correlation using digoxin as a probe drug. Clin Pharmacol Ther 85 173–181. (2009) [DOI] [PubMed] [Google Scholar]
- 26.Ellens H, et al. Application of receiver operating characteristic analysis to refine the prediction of potential digoxin drug interactions. Drug Metab Dispos 41 1367–1374. (2013) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Poirier A, et al. Calibration of in vitro multidrug resistance protein 1 substrate and inhibition assays as a basis to support the prediction of clinically relevant interactions in vivo. Drug Metab Dispos 42 1411–1422. (2014) [DOI] [PubMed] [Google Scholar]
- 28.Agarwal S, Arya V, Zhang L. Review of P-gp inhibition data in recently approved new drug applications: utility of the proposed [I(1) ]/IC(50) and [I(2) ]/IC(50) criteria in the P-gp decision tree. J Clin Pharmacol 53 228–233. (2013) [DOI] [PubMed] [Google Scholar]
- 29.Zhou T, Arya V, Zhang L. Comparing Various In Vitro Prediction Methods to Assess the Potential of a Drug to Inhibit P-glycoprotein (P-gp) Transporter In Vivo. J Clin Pharmacol 59 1049–1060. (2019) [DOI] [PubMed] [Google Scholar]
- 30.FDA Guidance for Industry: In Vitro Drug Interaction Studies — Cytochrome P450 Enzyme- and Transporter-Mediated Drug Interactions. 2020. https://www.fda.gov/media/134582/download.
- 31.EMA guideline on the investigation of drug interactions (2013) https://www.ema.europa.eu/en/documents/scientific-guideline/guideline-investigation-drug-interactions-revision-1_en.pdf.
- 32.Bentz J, et al. Variability in P-glycoprotein inhibitory potency (IC(5)(0)) using various in vitro experimental systems: implications for universal digoxin drug-drug interaction risk assessment decision criteria. Drug Metab Dispos 41 1347–1366. (2013) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Kalliokoski T, Kramer C, Vulpetti A, Gedeck P. Comparability of mixed IC(5)(0) data - a statistical analysis. PLoS One 8 e61007. (2013) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Japanese MHLW (PMDA) Guidance on Drug Interaction for Drug Development and Appropriate Provision of Information. 2018. https://www.pmda.go.jp/files/000228122.pdf.
- 35.Nolan PE Jr., et al. Effects of coadministration of propafenone on the pharmacokinetics of digoxin in healthy volunteer subjects. J Clin Pharmacol 29 46–52. (1989) [DOI] [PubMed] [Google Scholar]
- 36.Johnson BF, Cheng SL, Venitz J. Transient kinetic and dynamic interactions between verapamil and dofetilide, a class III antiarrhythmic. J Clin Pharmacol 41 1248–1256. (2001) [DOI] [PubMed] [Google Scholar]
- 37.Pauli-Magnus C, et al. Characterization of the major metabolites of verapamil as substrates and inhibitors of P-glycoprotein. J Pharmacol Exp Ther 293 376–382. (2000) [PubMed] [Google Scholar]
- 38.Hanke N, et al. A Mechanistic, Enantioselective, Physiologically Based Pharmacokinetic Model of Verapamil and Norverapamil, Built and Evaluated for Drug-Drug Interaction Studies. Pharmaceutics 12. (2020) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Chaudhry A, et al. Derivation of a System-Independent Ki for P-glycoprotein Mediated Digoxin Transport from System-Dependent IC50 Data. Drug Metab Dispos 46 279–290. (2018) [DOI] [PubMed] [Google Scholar]
- 40.NDA 209936 Aliqopa (Copanlisib dihydrochloride) Bayer Healthcare<https://www.accessdata.fda.gov/scripts/cder/daf/index.cfm?event=overview.process&varApplNo=209936>
- 41.NDA 203085 Stivarga (Regorafenib) Bayer Healthcare <https://www.accessdata.fda.gov/scripts/cder/daf/index.cfm?event=overview.process&varApplNo=203085>
- 42.Huang C, et al. Coadministration of vindesine with high-dose methotrexate therapy increases acute kidney injury via BCRP, MRP2, and OAT1/OAT3. Cancer Chemother Pharmacol 85 433–441. (2020) [DOI] [PubMed] [Google Scholar]
- 43.Billington S, et al. Positron Emission Tomography Imaging of [(11) C]Rosuvastatin Hepatic Concentrations and Hepatobiliary Transport in Humans in the Absence and Presence of Cyclosporin A. Clin Pharmacol Ther 106 1056–1066. (2019) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Kaneko K, et al. A Clinical Quantitative Evaluation of Hepatobiliary Transport of [(11)C]Dehydropravastatin in Humans Using Positron Emission Tomography. Drug Metab Dispos 46 719–728. (2018) [DOI] [PubMed] [Google Scholar]
- 45.Keitel V, et al. A common Dubin-Johnson syndrome mutation impairs protein maturation and transport activity of MRP2 (ABCC2). Am J Physiol Gastrointest Liver Physiol 284 G165–174. (2003) [DOI] [PubMed] [Google Scholar]
- 46.Pedersen KE, Christiansen BD, Klitgaard NA, Nielsen-Kudsk F. Changes in steady state digoxin pharmacokinetics during quinidine therapy in cardiac patients: influence of plasma quinidine concentration. Acta Pharmacol Toxicol (Copenh) 52 357–363. (1983) [DOI] [PubMed] [Google Scholar]
- 47.Angelin B, Arvidsson A, Dahlqvist R, Hedman A, Schenck-Gustafsson K. Quinidine reduces biliary clearance of digoxin in man. Eur J Clin Invest 17 262–265. (1987) [DOI] [PubMed] [Google Scholar]
- 48.Ali I, et al. Transporter-Mediated Alterations in Patients With NASH Increase Systemic and Hepatic Exposure to an OATP and MRP2 Substrate. Clin Pharmacol Ther. (2017) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Ghibellini G, Leslie EM, Brouwer KL. Methods to evaluate biliary excretion of drugs in humans: an updated review. Mol Pharm 3 198–211. (2006) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Guiney WJ, et al. Use of Entero-Test, a simple approach for non-invasive clinical evaluation of the biliary disposition of drugs. Br J Clin Pharmacol 72 133–142. (2011) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Lum BL, et al. Alteration of etoposide pharmacokinetics and pharmacodynamics by cyclosporine in a phase I trial to modulate multidrug resistance. J Clin Oncol 10 1635–1642. (1992) [DOI] [PubMed] [Google Scholar]
- 52.Boote DJ, et al. Phase I study of etoposide with SDZ PSC 833 as a modulator of multidrug resistance in patients with cancer. J Clin Oncol 14 610–618. (1996) [DOI] [PubMed] [Google Scholar]
- 53.Hershey N Assessing peer review in the quest for improved medical services: Part III. Qual Assur Util Rev 5 63–68. (1990) [DOI] [PubMed] [Google Scholar]
- 54.Jarvinen E, Deng F, Kidron H, Finel M. Efflux transport of estrogen glucuronides by human MRP2, MRP3, MRP4 and BCRP. J Steroid Biochem Mol Biol 178 99–107. (2018) [DOI] [PubMed] [Google Scholar]
- 55.Tran P, et al. Disposition of asciminib, a potent BCR-ABL1 tyrosine kinase inhibitor, in healthy male subjects. Xenobiotica 50 150–169. (2020) [DOI] [PubMed] [Google Scholar]
- 56.Taskar KS, et al. Physiologically-Based Pharmacokinetic Models for Evaluating Membrane Transporter Mediated Drug-Drug Interactions: Current Capabilities, Case Studies, Future Opportunities, and Recommendations. Clin Pharmacol Ther 107 1082–1115. (2020) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Zamek-Gliszczynski MJ, et al. ITC recommendations for transporter kinetic parameter estimation and translational modeling of transport-mediated PK and DDIs in humans. Clin Pharmacol Ther 94 64–79. (2013) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Fenster PE, White NW Jr., Hanson CD. Pharmacokinetic evaluation of the digoxin-amiodarone interaction. J Am Coll Cardiol 5 108–112. (1985) [DOI] [PubMed] [Google Scholar]
- 59.Nademanee K, et al. Amiodarone-digoxin interaction: clinical significance, time course of development, potential pharmacokinetic mechanisms and therapeutic implications. J Am Coll Cardiol 4 111–116. (1984) [DOI] [PubMed] [Google Scholar]
- 60.Hager WD, et al. Digoxin-quinidine interaction Pharmacokinetic evaluation. N Engl J Med 300 1238–1241. (1979) [DOI] [PubMed] [Google Scholar]
- 61.Leahey EB Jr., et al. Quinidine-digoxin interaction: time course and pharmacokinetics. Am J Cardiol 48 1141–1146. (1981) [DOI] [PubMed] [Google Scholar]
- 62.Hager WD, Mayersohn M, Graves PE. Digoxin bioavailability during quinidine administration. Clin Pharmacol Ther 30 594–599. (1981) [DOI] [PubMed] [Google Scholar]
- 63.Fenster PE, et al. Digoxin-quinidine interaction in patients with chronic renal failure. Circulation 66 1277–1280. (1982) [DOI] [PubMed] [Google Scholar]
- 64.Fenster PE, Hager WD, Goodman MM. Digoxin-quinidine-spironolactone interaction. Clin Pharmacol Ther 36 70–73. (1984) [DOI] [PubMed] [Google Scholar]
- 65.Ochs HR, Bodem G, Greenblatt DJ. Impairment of digoxin clearance by coadministration of quinidine. J Clin Pharmacol 21 396–400. (1981) [DOI] [PubMed] [Google Scholar]
- 66.Schenck-Gustafsson K, Dahlqvist R. Pharmacokinetics of digoxin in patients subjected to the quinidine-digoxin interaction. Br J Clin Pharmacol 11 181–186. (1981) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Wandell M, et al. Effect of quinine on digoxin kinetics. Clin Pharmacol Ther 28 425–430. (1980) [DOI] [PubMed] [Google Scholar]
- 68.Ding R, et al. Substantial pharmacokinetic interaction between digoxin and ritonavir in healthy volunteers. Clin Pharmacol Ther 76 73–84. (2004) [DOI] [PubMed] [Google Scholar]
- 69.Waldorff S, et al. Spironolactone-induced changes in digoxin kinetics. Clin Pharmacol Ther 24 162–167. (1978) [DOI] [PubMed] [Google Scholar]
- 70.Pedersen KE, Dorph-Pedersen A, Hvidt S, Klitgaard NA, Nielsen-Kudsk F. Digoxin-verapamil interaction. Clin Pharmacol Ther 30 311–316. (1981) [DOI] [PubMed] [Google Scholar]
- 71.Pedersen KE, Christiansen BD, Kjaer K, Klitgaard NA, Nielsen-Kudsk F. Verapamil-induced changes in digoxin kinetics and intraerythrocytic sodium concentration. Clin Pharmacol Ther 34 8–13. (1983) [DOI] [PubMed] [Google Scholar]
- 72.Kerr DJ, et al. The effect of verapamil on the pharmacokinetics of adriamycin. Cancer Chemother Pharmacol 18 239–242. (1986) [DOI] [PubMed] [Google Scholar]
- 73.Bartlett NL, et al. Phase I trial of doxorubicin with cyclosporine as a modulator of multidrug resistance. J Clin Oncol 12 835–842. (1994) [DOI] [PubMed] [Google Scholar]
- 74.Rushing DA, et al. The effects of cyclosporine on the pharmacokinetics of doxorubicin in patients with small cell lung cancer. Cancer 74 834–841. (1994) [DOI] [PubMed] [Google Scholar]
- 75.Moreira A, et al. Influence of the interval between the administration of doxorubicin and paclitaxel on the pharmacokinetics of these drugs in patients with locally advanced breast cancer. Cancer Chemother Pharmacol 48 333–337. (2001) [DOI] [PubMed] [Google Scholar]
- 76.Briasoulis E, et al. Interaction pharmacokinetics of pegylated liposomal doxorubicin (Caelyx) on coadministration with paclitaxel or docetaxel. Cancer Chemother Pharmacol 53 452–457. (2004) [DOI] [PubMed] [Google Scholar]
- 77.Minami H, et al. Phase I study of intravenous PSC-833 and doxorubicin: reversal of multidrug resistance. Jpn J Cancer Res 92 220–230. (2001) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Giaccone G, et al. A dose-finding and pharmacokinetic study of reversal of multidrug resistance with SDZ PSC 833 in combination with doxorubicin in patients with solid tumors. Clin Cancer Res 3 2005–2015. (1997) [PubMed] [Google Scholar]
- 79.Advani R, et al. A phase I trial of doxorubicin, paclitaxel, and valspodar (PSC 833), a modulator of multidrug resistance. Clin Cancer Res 7 1221–1229. (2001) [PubMed] [Google Scholar]
- 80.Sonneveld P, et al. Reversal of multidrug resistance by SDZ PSC 833, combined with VAD (vincristine, doxorubicin, dexamethasone) in refractory multiple myeloma. A phase I study. Leukemia 10 1741–1750. (1996) [PubMed] [Google Scholar]
- 81.Matsushima N, Lee F, Sato T, Weiss D, Mendell J. Bioavailability and Safety of the Factor Xa Inhibitor Edoxaban and the Effects of Quinidine in Healthy Subjects. Clin Pharmacol Drug Dev 2 358–366. (2013) [DOI] [PubMed] [Google Scholar]
- 82.Kornblau SM, et al. Phase I study of mitoxantrone plus etoposide with multidrug blockade by SDZ PSC-833 in relapsed or refractory acute myelogenous leukemia. J Clin Oncol 15 1796–1802. (1997) [DOI] [PubMed] [Google Scholar]
- 83.Advani R, et al. Treatment of refractory and relapsed acute myelogenous leukemia with combination chemotherapy plus the multidrug resistance modulator PSC 833 (Valspodar). Blood 93 787–795. (1999) [PubMed] [Google Scholar]
- 84.Pein F, et al. Dose finding study of oral PSC 833 combined with weekly intravenous etoposide in children with relapsed or refractory solid tumours. Eur J Cancer 43 2074–2081. (2007) [DOI] [PubMed] [Google Scholar]
- 85.Smeets M, et al. Cyclosporin increases cellular idarubicin and idarubicinol concentrations in relapsed or refractory AML mainly due to reduced systemic clearance. Leukemia 15 80–88. (2001) [DOI] [PubMed] [Google Scholar]
- 86.Pea F, et al. Multidrug resistance modulation in vivo: the effect of cyclosporin A alone or with dexverapamil on idarubicin pharmacokinetics in acute leukemia. Eur J Clin Pharmacol 55 361–368. (1999) [DOI] [PubMed] [Google Scholar]
- 87.Chi KN, et al. A phase I pharmacokinetic study of the P-glycoprotein inhibitor, ONT-093, in combination with paclitaxel in patients with advanced cancer. Invest New Drugs 23 311–315. (2005) [DOI] [PubMed] [Google Scholar]
- 88.Chico I, et al. Phase I study of infusional paclitaxel in combination with the P-glycoprotein antagonist PSC 833. J Clin Oncol 19 832–842. (2001) [DOI] [PubMed] [Google Scholar]
- 89.Shoaf SE, et al. Tolvaptan administration does not affect steady state amiodarone concentrations in patients with cardiac arrhythmias. J Cardiovasc Pharmacol Ther 10 165–171. (2005) [DOI] [PubMed] [Google Scholar]
- 90.Robinson K, et al. The digoxin-amiodarone interaction. Cardiovasc Drugs Ther 3 25–28. (1989) [DOI] [PubMed] [Google Scholar]
- 91.Rasool MF, Khalil F, Laer S. Optimizing the Clinical Use of Carvedilol in Liver Cirrhosis Using a Physiologically Based Pharmacokinetic Modeling Approach. Eur J Drug Metab Pharmacokinet 42 383–396. (2017) [DOI] [PubMed] [Google Scholar]
- 92.Ochs HR, Gugler R, Guthoff T, Greenblatt DJ. Effect of cimetidine on digoxin kinetics and creatinine clearance. Am Heart J 107 170–172. (1984) [DOI] [PubMed] [Google Scholar]
- 93.Rengelshausen J, et al. Contribution of increased oral bioavailability and reduced nonglomerular renal clearance of digoxin to the digoxin-clarithromycin interaction. Br J Clin Pharmacol 56 32–38. (2003) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Ishii K, et al. Comparative study of pharmacokinetic parameters between clarithromycin and erythromycin stearate in relation to their physicochemical properties. Drug Dev Ind Pharm 24 129–137. (1998) [DOI] [PubMed] [Google Scholar]
- 95.NDA 021697 Vaprisol (Conivaptan hydrochloride) Cumberland Pharm <https://www.accessdata.fda.gov/scripts/cder/daf/index.cfm?event=overview.process&ApplNo=21697>.
- 96.Weir SJ, Dimmitt DC, Lanman RC, Morrill MB, Geising DH. Steady-state pharmacokinetics of diltiazem and hydrochlorothiazide administered alone and in combination. Biopharm Drug Dispos 19 365–371. (1998) [DOI] [PubMed] [Google Scholar]
- 97.Risler T, Burk M, Peters U, Grabensee B, Seipel L. On the interaction between digoxin and disopyramide. Clin Pharmacol Ther 34 176–180. (1983) [DOI] [PubMed] [Google Scholar]
- 98.Welling PG, Elliott RL, Pitterle ME, Corrick-West HP, Lyons LL. Plasma levels following single and repeated doses of erythromycin estolate and erythromycin stearate. J Pharm Sci 68 150–155. (1979) [DOI] [PubMed] [Google Scholar]
- 99.DiSanto AR, Chodos DJ. Influence of study design in assessing food effects on absorption of erythromycin base and erythromycin stearate. Antimicrob Agents Chemother 20 190–196. (1981) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Mather LE, Austin KL, Philpot CR, McDonald PJ. Absorption and bioavailability of oral erythromycin. Br J Clin Pharmacol 12 131–140. (1981) [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.