U.S. flag An official website of the United States government
  1. Home
  2. Drugs
  3. Development & Approval Process | Drugs
  4. Development Resources
  5. Drug Interactions & Labeling
  6. Drug Development and Drug Interactions | Table of Substrates, Inhibitors and Inducers
  1. Drug Interactions & Labeling

Drug Development and Drug Interactions | Table of Substrates, Inhibitors and Inducers


Table 1-1: Examples of in vitro marker reactions for CYP-mediated metabolism

Enzyme

Marker reaction

CYP1A2

7-ethoxyresorufin-O-deethylation, phenacetin O-deethylation

CYP2B6

bupropion hydroxylation, efavirenz hydroxylation

CYP2C8

amodiaquine N-deethylation, paclitaxel 6α-hydroxylation

CYP2C9

diclofenac 4'-hydroxylation, S-warfarin 7-hydroxylation

CYP2C19

S-mephenytoin 4'-hydroxylation

CYP2D6

bufuralol 1'-hydroxylation, dextromethorphan O-demethylation

CYP3A4/5(a)

midazolam 1'-hydroxylation, testosterone 6β-hydroxylation

Recommend the use of two structurally unrelated CYP3A4/5 substrates to evaluate in vitro CYP3A4/5 inhibition.

Abbreviations:
CYP: cytochrome P450


Table 1-2: Examples of in vitro selective inhibitors for CYP-mediated metabolism

Enzyme

Inhibitor

CYP1A2

α-naphthoflavone, furafylline(a)

CYP2B6

clopidogrel(a), sertraline, thiotepa(a), ticlopidine(a)

CYP2C8

gemfibrozil glucuronide(a), montelukast, phenelzine(a)

CYP2C9

sulfaphenazole, tienilic acid(a)

CYP2C19

N-3-benzyl-nirvanol, loratadine, nootkatone, ticlopidine(a)

CYP2D6

paroxetine(a), quinidine

CYP3A4/5

azamulin(a), itraconazole, ketoconazole, troleandomycin(a), verapamil(a)

Note: Many of these chemical inhibitors are not specific for an individual CYP enzyme. The selectivity and potency of inhibitors should be verified using the same experimental conditions with probe substrates for each CYP enzyme.

Time-dependent inhibitors.

Abbreviations:
CYP: cytochrome P450


Table 1-3. Examples of in vitro inducers for CYP-mediated metabolism

Enzyme

Inducer

CYP1A2

omeprazole

CYP2B6

phenobarbital

CYP2C8

rifampicin

CYP2C9

rifampicin

CYP2C19

rifampicin

CYP3A4/5

rifampicin

Abbreviations:
CYP: cytochrome P450.


Table 2-1: Examples of clinical index substrates for CYP-mediated metabolism (for use in index clinical DDI studies)

 Enzyme

Sensitive index substrates unless otherwise noted

CYP1A2

caffeine, tizanidine

CYP2B6(a)

-

CYP2C8

repaglinide(b)

CYP2C9

tolbutamide(c), S-warfarin(c)

CYP2C19

lansoprazole(c,d), omeprazole

CYP2D6

desipramine, dextromethorphan, nebivolol

CYP3A

midazolam, triazolam

Note: Index substrates predictably exhibit exposure increases due to inhibition of a given metabolic pathway and are commonly used in prospective clinical DDI studies. See section IV.A.2 of the FDA guidance for industry entitled Clinical Drug Interaction Studies —Cytochrome P450 Enzyme- and Transporter-Mediated Drug Interactions (January 2020) for details. Sensitive index substrates are index drugs that demonstrate an increase in AUC of ≥5-fold with strong index inhibitors of a given metabolic pathway in clinical DDI studies. Moderately sensitive substrates are drugs that demonstrate an increase in AUC of ≥2- to <5-fold with strong index inhibitors of a given metabolic pathway in clinical DDI studies.

This table provides examples of clinical sensitive or moderately sensitive index substrates and is not intended to be an exhaustive list. Index substrates listed in this table were selected considering their sensitivity, specificity, safety profiles, and adequate number of reported clinical DDI studies with different in vivo inhibitors (≥ 3 for CYP3A or ≥ 2 for CYP1A2, 2C8, 2C9, 2C19, and 2D6). DDI data were collected based on a search of the University of Washington Metabolism and Transport Drug Interaction Database [Hachad et al. (2010), Hum Genomics, 5(1):61], and the list of references is available here.

We currently do not have sensitive index substrates for CYP2B6.
Also OATP1B1 substrate.
Moderately sensitive substrates.
S-lansoprazole is a sensitive substrate in CYP2C19 EM subjects.

Abbreviations:
AUC: area under the concentration-time curve; CYP: cytochrome P450; DDI: drug-drug interaction; EM: extensive metabolizer; OATP1B1: organic anion transporting polypeptide 1B1.


Table 2-2: Examples of clinical index inhibitors for CYP enzymes for use in index clinical DDI studies)

 Enzyme

Strong index inhibitors

Moderate index inhibitors

CYP1A2

fluvoxamine(a)

-

CYP2B6(b)

-

-

CYP2C8

gemfibrozil(c)

clopidogrel(d)

CYP2C9

-

fluconazole(e)

CYP2C19

fluvoxamine(a)

-

CYP2D6

fluoxetine(f), paroxetine

Mirabegron

CYP3A

clarithromycin(g), itraconazole(g)

erythromycin(g), fluconazole(e), verapamil(g)

Note: Index inhibitors predictably inhibit metabolism via a given pathway and are commonly used in prospective clinical DDI studies. See section IV.A.2 of the FDA guidance for industry entitled Clinical Drug Interaction Studies —Cytochrome P450 Enzyme- and Transporter-Mediated Drug Interactions (January 2020) for details. Strong and moderate inhibitors are drugs that increase the AUC of sensitive index substrates of a given metabolic pathway ≥5-fold and ≥2- to <5-fold, respectively.

This table provides examples of clinical index inhibitors and is not intended to be an exhaustive list. Index inhibitors listed in this table were selected based on potency and selectivity of inhibition, safety profiles, and an adequate number of reported clinical DDI studies with different in vivo substrates [≥ 3 for CYP3A, ≥ 2 for CYP1A2, 2C9, 2C19, and 2D6, or ≥ 1 for CYP2C8 (strong inhibitors)]. DDI data were collected based on a search of the University of Washington Metabolism and Transport Drug Interaction Database [Hachad et al. (2010), Hum Genomics, 5(1):61)], and the list of references is available here.

a Strong inhibitor of CYP1A2 and CYP2C19, moderate inhibitor of CYP3A, and weak inhibitor of CYP2D6.

b We currently do not have index inhibitors for CYP2B6.

c Strong inhibitor of CYP2C8 and inhibitor of OATP1B1 and OAT3.

d Moderate inhibitor of CYP2C8 at the 75 mg dose of clopidogrel and a weak inhibitor of CYP2B6.

e Strong inhibitor of CYP2C19 and moderate inhibitor of CYP2C9 and CYP3A.

f Strong inhibitor of CYP2C19 and CYP2D6.

g Inhibitor of P-gp (defined as those increasing the AUC of digoxin to ≥1.25-fold).

Abbreviations:
AUC: area under the concentration-time curve; CYP: cytochrome P450; DDI: drug-drug interaction; OATP1B1: organic anion transporting polypeptide 1B1; OAT3: organic anion transporter 3; P-gp: P-glycoprotein.


Table 2-3: Examples of clinical index inducers for CYP-mediated metabolism (for use in index clinical DDI studies)

 

Strong inducers

Moderate inducers

CYP1A2

-

-

CYP2B6

-

rifampin(a)

CYP2C8

-

rifampin(a)

CYP2C9

-

rifampin(a)

CYP2C19

rifampin(a)

-

CYP3A

carbamazepine(b), phenytoin(c), rifampin(a)

-

Note: Index inducers predictably induce metabolism via a given pathway and are commonly used in prospective clinical DDI studies. See section IV.A.2 of the FDA guidance for industry entitled Clinical Drug Interaction Studies — Cytochrome P450 Enzyme- and Transporter-Mediated Drug Interactions (January 2020) for more details. Strong and moderate index inducers are drugs that decrease the AUC of sensitive substrates of a given metabolic pathway by ≥80 percent and ≥50 to <80 percent, respectively.

This table provides examples of clinical index inducers and is not intended to be an exhaustive list. Index inducers listed in this table were selected based on potency of induction, safety profiles, and number of reported clinical DDI studies with different in vivo substrates (≥ two substrates). DDI data were collected based on a search of the University of Washington Metabolism and Transport Drug Interaction Database [Hachad et al. (2010), Hum Genomics, 5(1):61], and the list of references is available here.

a Strong inducer of CYP2C19 and CYP3A, and moderate inducer of CYP1A2, CYP2B6, CYP2C8, CYP2C9.
b Strong inducer of CYP2B6 and CYP3A and a weak inducer of CYP2C9.
c Strong inducer of CYP3A and a moderate inducer of CYP1A2 and CYP2C19.

Abbreviations:
AUC: area under the concentration-time curve; CYP: cytochrome P450; DDI: drug-drug interaction.


Table 3-1: Examples of clinical substrates for CYP-mediated metabolism (for concomitant use in clinical DDI studies and/or drug labeling)

 

Sensitive substrates

Moderate sensitive substrates

CYP1A2

alosetron, caffeine, duloxetine, melatonin, ramelteon, tasimelteon, tizanidine

clozapine, pirfenidone, ramosetron, theophylline

CYP2B6

bupropion(a)

efavirenz

CYP2C8

repaglinide(b)

montelukast, pioglitazone, rosiglitazone

CYP2C9

celecoxib(c)

glimepiride, phenytoin, tolbutamide, S-warfarin

CYP2C19

S-mephenytoin, omeprazole

diazepam, lansoprazole(d), rabeprazole, voriconazole

CYP2D6

atomoxetine, desipramine, dextromethorphan, eliglustat(e), nebivolol, nortriptyline, perphenazine, tolterodine, R-venlafaxine

encainide, imipramine, metoprolol, propafenone, propranolol, tramadol, trimipramine, S-venlafaxine

CYP3A

Substrates with ≥10-fold increase in AUC by co-administration of strong inhibitors:

alfentanil, avanafil, buspirone, conivaptan, darifenacin, darunavir(f), ebastine, everolimus, ibrutinib, lomitapide, lovastatin(b), midazolam, naloxegol, nisoldipine, saquinavir(f), simvastatin(b), sirolimus, tacrolimus, tipranavir(f), triazolam, vardenafil

alprazolam, aprepitant, atorvastatin(b), colchicine, eliglustat(e), pimozide, rilpivirine, rivaroxaban, tadalafil

 

Substrates with 5- to 10-fold increase in AUC by co-administration of strong inhibitors:

budesonide, dasatinib, dronedarone, eletriptan, eplerenone, felodipine, indinavir(f), isavuconazole, ivabradine, lemborexant, lurasidone, maraviroc, mobocertinib, quetiapine, sildenafil, ticagrelor, tolvaptan, venetoclax

Note: Sensitive substrates are drugs that demonstrate an increase in AUC of ≥5-fold with strong index inhibitors of a given metabolic pathway in clinical DDI studies. Moderate sensitive substrates are drugs that demonstrate an increase in AUC of ≥2- to <5-fold with strong index inhibitors of a given metabolic pathway in clinical DDI studies. Other elimination pathways can also contribute to the elimination of the substrates listed in the table above and should be considered when assessing the drug interaction potential.

This table provides examples of clinical substrates and is not intended to be an exhaustive list. DDI data were collected based on a search of the University of Washington Metabolism and Transport Drug Interaction Database [Hachad et al. (2010), Hum Genomics, 5(1):61].

a Bupropion itself is not a sensitive substrate. It is metabolized by multiple enzymes including CYP2B6 that is primarily responsible for the formation of hydroxybupropion. When used in a clinical DDI study, both bupropion and its metabolite hydroxybupropion should be measured and reported.
b OATP1B1 substrate.
c Listed based on pharmacogenetic studies.
d S-lansoprazole is a sensitive substrate in CYP2C19 EM subjects.
e Sensitive substrate of CYP2D6 and moderate sensitive substrate of CYP3A.
f Usually administered to patients in combination with ritonavir, a strong CYP3A inhibitor.

Abbreviations:
AUC: area under the concentration-time curve; CYP: cytochrome P450; DDI: drug-drug interaction; EM: extensive metabolizer; OATP1B1: organic anion transporting polypeptide 1B1.


Table 3-2: Examples of clinical inhibitors for CYP-mediated metabolism (for concomitant use clinical DDI studies and/or drug labeling)

 

Strong inhibitors

Moderate inhibitors

Weak inhibitors

CYP1A2

ciprofloxacin, enoxacin, fluvoxamine(a)

methoxsalen, mexiletine, oral contraceptives, vemurafenib

acyclovir, allopurinol, cimetidine, peginterferon alpha-2a, piperine, zileuton

CYP2B6

-

-

clopidogrel(b), tenofovir, ticlopidine(c), voriconazole(d)

CYP2C8

gemfibrozil(e)

clopidogrel(b), deferasirox, teriflunomide

trimethoprim

CYP2C9

-

amiodarone(h), fluconazole(f), miconazole, piperine

ceritinib, diosmin, disulfiram, fluvastatin, fluvoxamine(a), voriconazole(d)

CYP2C19

fluconazole(f), fluoxetine(g), fluvoxamine(a), ticlopidine(c)

cenobamate, felbamate, voriconazole(d)

omeprazole

CYP2D6

bupropion, fluoxetine(g), paroxetine, quinidine(h), terbinafine

abiraterone, cinacalcet, duloxetine, lorcaserin, mirabegron, rolapitant

amiodarone(h), celecoxib, cimetidine, clobazam, cobicistat, escitalopram, fluvoxamine(a), labetalol, sertraline, vemurafenib

CYP3A4

The inhibitors below cause a ≥10-fold increase in AUC of sensitive substrate(s):

cobicistat(h), danoprevir and ritonavir(j), elvitegravir and ritonavir(j), grapefruit juice(k), indinavir and ritonavir(j), itraconazole(h), ketoconazole(h), lopinavir and ritonavir(h,j), paritaprevir and ritonavir and ombitasvir (and/or dasabuvir)(j), posaconazole, ritonavir(h,i,j), saquinavir and ritonavir(h,j), tipranavir and ritonavir(j), telithromycin, troleandomycin, voriconazole(d)

aprepitant, ciprofloxacin, conivaptan(l), crizotinib, cyclosporine, diltiazem(m), dronedarone(h), erythromycin(h), fluconazole(f), fluvoxamine(a), grapefruit juice(k), imatinib, isavuconazole, tofisopam, verapamil(h)

chlorzoxazone, cilostazol, cimetidine, clotrimazole, fosaprepitant, istradefylline, ivacaftor, lomitapide, ranitidine, ranolazine(h), ticagrelor(h)

The inhibitors below cause a 5- to 10-fold increase in the AUC of sensitive substrate(s):

ceritinib, clarithromycin(h), idelalisib, nefazodone, nelfinavir

 -

 -

Note: Strong, moderate, and weak inhibitors are drugs that increase the AUC of sensitive index substrates of a given metabolic pathway ≥5-fold, ≥2 to <5-fold, and ≥1.25 to <2-fold, respectively.

This table provides examples of clinical inhibitors and is not intended to be an exhaustive list. DDI data were collected based on a search of the University of Washington Metabolism and Transport Drug Interaction Database [Hachad et al. (2010), Hum Genomics, 5(1):61].

a Strong inhibitor of CYP1A2 and CYP2C19, moderate inhibitor of CYP3A, and weak inhibitor of CYP2D6.
b Moderate inhibitor of CYP2C8 and a weak inhibitor of CYP2B6.
c Strong inhibitor of CYP2C19 and a weak inhibitor of CYP2B6. The classification as a CYP2B6 inhibitor is based on the AUC change of bupropion. The effect of ticlopidine on hydroxybupropion, which is primarily metabolized by CYP2B6, is larger.
d Strong inhibitor of CYP3A, moderate inhibitor of CYP2C19, and weak inhibitor of CYP2B6 and CYP2C9.
e Strong inhibitor of CYP2C8 and an inhibitor of OATP1B1 and OAT3.
f Strong inhibitor of CYP2C19 and a moderate inhibitor of CYP2C9 and CYP3A.
g Strong inhibitors of CYP2C19 and CYP2D6.
h Inhibitor of P-gp (, defined as those increasing AUC or Cmax of digoxin, dabigatran, or edoxaban ≥1.5-fold).
i Strong inhibitor of CYP3A4 and weak inducer of CYP2B6, CYP2C9, and CYP2C19.
j Ritonavir is usually given in combination with other anti-HIV or anti-HCV drugs in clinical practice. Caution should be used when extrapolating the observed effect of ritonavir alone to the effect of combination regimens on CYP3A activities.
k The effect of grapefruit juice varies widely among brands and is concentration-, dose-, and preparation-dependent. Studies have shown that it can be classified as a ‘strong CYP3A inhibitor’ when a certain preparation is used (e.g., high dose, double strength) or as a ‘moderate CYP3A inhibitor’ when another preparation is used (e.g., low dose, single strength).
l The classification is based on studies conducted with intravenously administered conivaptan.
m Diltiazem increased the AUC of certain sensitive CYP3A substrates (e.g., buspirone) more than 5-fold.

Abbreviations:
AUC: area under the concentration-time curve; CYP: cytochrome P450; DDI: drug-drug interaction; HIV: human immunodeficiency virus; HCV: hepatitis C virus; OATP1B1: organic anion transporting polypeptide 1B1; OAT3: organic anion transporter 3; P-gp: P-glycoprotein.


Table 3-3: Examples of clinical inducers for CYP-mediated metabolism (for concomitant use clinical DDI studies and/or drug labeling)

 

Strong inducers

Moderate inducers

Weak inducers

CYP1A2

 -

phenytoin(a), rifampin(b), smoking, teriflunomide

-

CYP2B6

carbamazepine(c)

efavirenz(d), rifampin(b)

isavuconazole, lemborexant, lorlatinib, nevirapine, ritonavir(e,f)

CYP2C8

-

rifampin(b)

-

CYP2C9

-

enzalutamide(g), rifampin(b)

apalutamide(h), aprepitant, carbamazepine(c), dabrafenib, lorlatinib, ritonavir(e,f)

CYP2C19

rifampin(b)

apalutamide(h), efavirenz(d), enzalutamide(g), phenytoin(a)

ritonavir(e,f)

CYP3A

apalutamide(h), carbamazepine(c), enzalutamide(g), ivosidenib(i), lumacaftor, mitotane, phenytoin(a), rifampin(b), St. John’s wort(j)

bosentan, cenobamate(k), dabrafenib, efavirenz(d), etravirine, lorlatinib, pexidartinib, phenobarbital, primidone, sotorasib

armodafinil, elagolix, mobocertinib, modafinil(l), rufinamide, vemurafenib, zanubrutinib

Note: Strong, moderate, and weak inducers are drugs that decreases the AUC of sensitive index substrates of a given metabolic pathway by ≥80%, ≥50% to <80%, and ≥20% to <50%, respectively.

This table provides examples of clinical index inducers and is not intended to be an exhaustive list. DDI data were collected based on a search of the University of Washington Metabolism and Transport Drug Interaction Database [Hachad et al. (2010), Hum Genomics, 5(1):61].

a Strong inducer of CYP3A and a moderate inducer of CYP1A2 and CYP2C19.
b Strong inducer of CYP2C19 and CYP3A and a moderate inducer of CYP1A2, CYP2B6, CYP2C8, and CYP2C9.
c Strong inducer of CYP2B6 and CYP3A and weak inducer of CYP2C9.
d Moderate inducer of CYP2B6, CYP2C19, and CYP3A.
e Weak inducer of CYP2B6, CYP2C9, and CYP2C19. Classification is based on studies conducted with ritonavir itself (not with other anti-HIV drugs) at doses of 100-200 mg/day, although larger effects have been reported in literature for high doses of ritonavir.
f Moderate inducer of CYP1A2 with a dose of 800 mg/day ritonavir (not with other anti-HIV drugs). The effect on CYP1A2 at lower doses of ritonavir is unknown.
g Strong inducer of CYP3A and moderate inducer of CYP2C9 and CYP2C19.
h Strong inducer of CYP3A, moderate inducer of CYP2C19, and weak inducer of CYP2C9.
i The effect was based on prediction using physiologically based pharmacokinetic (PBPK) modeling.
j The effect of St. John’s wort varies widely and is preparation dependent.
k The classification is based on a 200 mg daily dose of cenobamate. Its effect potentially could be stronger at 400 mg/day.
l The classification is based on effect of 200 mg/day modafinil. A higher dose (400 mg/day) modafinil had a larger induction effect on CYP3A.

Abbreviations:
AUC: area under the concentration-time curve; CYP: cytochrome P450; DDI: drug-drug interaction.


Table 4-1: Examples of in vitro substrates for transporters

Transporter

Gene

Substrate

P-gp

ABCB1

digoxin, fexofenadine(a,b,c,d), loperamide, N-methylquinidine (NMQ)(h), quinidine, talinolol, vinblastine(c)

BCRP

ABCG2

2-amino-1-methyl-6-phenylimidazo[4,5-b]pyridine (PhIP), estradiol-17-beta-glucuronide(a,c,e,h), estrone-3-sulfate(a,b,d,f), methotrexate(a,b,c,j), rosuvastatin(a,b,f), prazosin(e), sulfasalazine

OATP1B1,
OATP1B3

SLCO1B1,
SLCO1B3

cholecystokinin octapeptide(CCK-8)(g), estradiol-17β-glucuronide(a,c,e,i), pitavastatin(e,f,i), pravastatin(b,c,f,i), rosuvastatin(b,f,i), telmisartan(g)

OAT1

SLC22A6

adefovir, p-aminohippurate (PAH), cidofovir, tenofovir

OAT3

SLC22A8

benzylpenicillin, estrone-3-sulfate (a,d,f,i), methotrexate(a,c,i,j), pravastatin(a,c,f,i)

MATE1,

MATE-2K

SLC47A1, SLC47A2

creatinine(j), metformin(j), 1-methyl-4-phenylpyridinium (MPP+)(j), tetraethylammonium (TEA)(j)

OCT2

SLC22A2

creatinine(j), metformin(j), tetraethylammonium (TEA)(j)

Note: This table provides examples of in vitro substrates for various transporters and is not intended to be an exhaustive list.

Note: The IC50 values of several OATP1B inhibitors measured using estrone-3-sulfate as a substrate were larger than those measured using estradiol-17-beta-glucuronide or pitavastatin as substrates. Thus, using estrone-3-sulfate as a substrate may underpredict the potential of a drug as an inhibitor of OATP1B.

Note: The IC50 values of several OCT2 inhibitors measured using 1-methyl-4-phenylpyridinium (MPP+) as a substrate were larger than those measured using metformin or creatinine as substrate. Thus, using MPP+ as a substrate may underpredict the potential of a drug as an inhibitor of OCT2.
a Also a substrate of OATPs.
b Also a substrate of OAT3.
c Also a substrate of MRP2.
d Also a substrate of MATEs.
e Also a substrate of P-gp.
f Also a substrate of NTCP.
g Selective substrate of OATP1B3 (vs. OATP1B1).
h Used in vesicle experiments.
i Also a substrate of BCRP.
j Substrate of OCTs and MATEs.

Abbreviations:
BCRP: breast cancer resistance protein; MATE: multidrug and toxin extrusion protein; MRP2: multidrug resistance-associated protein 2; NTCP: Na+-taurocholate co-transporting polypeptide; OAT: organic anion transporter; OATP: organic anion transporting polypeptide; OCT: organic cation transporter; P-gp: P-glycoprotein, also called as multidrug resistance protein 1 (MDR1).


Table 4-2: Examples of in vitro inhibitors for transporters

Transporter

Gene

Inhibitor

P-gp

ABCB1

cyclosporine(a,b,c,d), elacridar (GF120918)(a), ketoconazole(a,c,e,f,g), quinidine(c,f,g), valspodar (PSC833), verapamil(c,f,g), zosuquidar (LY335979)

BCRP

ABCG2

elacridar (GF120918)(h), fumitremorgin C (FTC), ko143, novobiocin(c,e)

OATP1B1,
OATP1B3

SLCO1B1,
SLCO1B3

bromosulfophthalein (BSP) (b,d), cyclosporine(a,b,d,,h,i), estrone-3-sulfate(a,b,e), rifampicin(a,d,e,h), rifamycin SV

OAT1,

OAT3

SLC22A6, SLC22A8

benzylpenicillin(j), probenecid(c,d)

MATE1,

MATE-2K

SLC47A1, SLC47A2

cimetidine(e,f), pyrimethamine(f)

OCT2

SLC22A2

cimetidine(e,g), clonidine(g)

Note: This table provides examples of in vitro inhibitors for various transporters and is not intended to be an exhaustive list.
a Also an inhibitor of BCRP.
b Also an inhibitor of NTCP.
c Also an inhibitor of OATPs.
d Also an inhibitor of MRP2.
e Also an inhibitor of OAT3.
f Also an inhibitor of OCT2.
g Also an inhibitor of MATEs.
h Also an inhibitor of P-gp.
i Preincubation with inhibitors prior to inhibition studies causes a decrease of the Ki value.
j Selectively inhibit OAT3 at lower concentrations. Note at the concentration inhibiting OAT3, benzylpenicillin also inhibits OATP1B3.

Abbreviations:
BCRP: breast cancer resistance protein; MATE: multidrug and toxin extrusion protein; MRP2: multidrug resistance-associated protein 2; NTCP: Na+-taurocholate co-transporting polypeptide; OAT: organic anion transporter; OATP: organic anion transporting polypeptide; OCT: organic cation transporter; P-gp: P-glycoprotein, also called as multidrug resistance protein 1 (MDR1).


Table 5-1: Examples of clinical substrates for transporters (for use in clinical DDI studies and/or drug labeling)

Transporter

Gene

Substrate

 

P-gp

ABCB1

dabigatran etexilate(a), digoxin, edoxaban, fexofenadine(b,c,d)

 

BCRP

ABCG2

rosuvastatin(b,c), sulfasalazine(e)

 

OATP1B1,
OATP1B3

SLCO1B1,
SLCO1B3

atorvastatin(f,g,h), bosentan(g), docetaxel(d,g,i), elagolix(g,h), fexofenadine(c,d,g), glecaprevir(f,g,h), glyburide(j), grazoprevir(g,h), letermovir, paclitaxel(d,g,k), pitavastatin, pravastatin(c,d), repaglinide(k), rosuvastatin(c,f), simvastatin acid(h)

 

OAT1,
OAT3

SLC22A6,
SLC22A8

adefovir(l,m), baricitinib(n), bumetanide(n), cefaclor(n), ceftizoxime(n), ciprofloxacin, famotidine(n), furosemide, methotrexate(n), oseltamivir carboxylate(m,n), benzylpenicillin (penicillin G)(n), tenofovir(l,m)

 
 

 

MATE1,
MATE-2K,

OCT2

SLC47A1,

SLC47A2,

SLC22A2

metformin

 

 

Note: A clinical substrate should meet the following criteria:

  • P-gp: (1) AUC fold-increase is ≥1.5 with itraconazole, quinidine, or verapamil co-administration; (2) not extensively metabolized in humans; and (3) in vitro transported by P-gp expression systems.
  • BCRP: (1) AUC fold-increase is ≥1.5 with pharmacogenetic alteration of ABCG2 (421C>A) and (2) in vitro transported by BCRP expression systems.
  • OATP1B1/OATP1B3: (1) AUC fold-increase is ≥2 with rifampin (single dose) or cyclosporine A co-administration or pharmacogenetic alteration of SLCO1B1 (521T>C); and (2) in vitro transported by OATP1B1 and/or OATP1B3 expression systems.
  • OAT1/OAT3: (1) AUC fold-increase is ≥1.5 with probenecid co-administration; (2) fraction excreted unchanged into urine as an unchanged drug is ≥0.5; and (3) in vitro transported by OAT1 and/or OAT3 expression systems.
  • OCT2/MATE: (1) AUC fold-increase is ≥1.5 with dolutegravir or pyrimethamine co-administration; (2) fraction excreted unchanged into urine as an unchanged drug is ≥0.5; and (3) in vitro transported by OCT2 and/or MATEs expression systems.

This table provides examples of clinical substrates for various transporters and is not intended to be an exhaustive list. DDI data were collected based on a search of the University of Washington Metabolism and Transport Drug Interaction Database [Hachad et al. (2010), Hum Genomics, 5(1):61].

a Only affected by intestinal P-gp. Dabigatran etexilate is a pro-drug and converted by carboxylesterase (CES) to dabigatran which is the measured moiety (dabigatran is not a substrate of P-gp). Thus, for correct interpretation of clinical DDI results, pre-assessment of the inhibitory effects of an investigational drug on CES activity should be considered.

b Also a substrate of OATPs.
c Also a substrate of OAT3.
d Also a substrate of MRP2. .
e Only affected by intestinal BCRP.
f Also a substrate of BCRP.
g Also a substrate of P-gp.
h Also a substrate of CYP3A.
i In vitro data suggest a higher contribution of OATP1B3 than OATP1B1.
j Also a substrate of CYP2C9.
k Also a substrate of CYP2C8.
l In vitro data suggest a higher contribution of OAT1 than OAT3.
m These drugs are active moieties of their corresponding pro-drugs, adefovir dipivoxil, oseltamivir, tenofovir alafenamide fumarate (TAF), and tenofovir disoproxil fumarate (TDF). Those pro-drugs are substrates of P-gp.
n In vitro data suggest a higher contribution of OAT3 than OAT1.

Abbreviations:
AUC: area under the plasma concentration-time curve. BCRP: breast cancer resistance protein; MATE: multidrug and toxin extrusion protein; MRP2: multidrug resistance-associated protein 2; OAT: organic anion transporter; OATP: organic anion transporting polypeptide; OCT: organic cation transporter; P-gp: P-glycoprotein, also called as multidrug resistance protein 1 (MDR1).


Table 5-2: Examples of clinical inhibitors for transporters (for use in clinical DDI studies and drug labeling)  

Transporter

Gene

Inhibitor

 

P-gp(a)

ABCB1

amiodarone, clarithromycin(b), cobicistat, cyclosporine(b,c), dronedarone, erythromycin, itraconazole, ketoconazole, lapatinib(c), lopinavir and ritonavir, quinidine, ranolazine, saquinavir and ritonavir, verapamil

 

BCRP

ABCG2

curcumin, cyclosporine A(b,d), darolutamide(b,e), eltrombopag(b), febuxostat(e), fostamatinib(d), rolapitant(d,f), teriflunomide(b,e)

 

OATP1B1,

OATP1B3

SLCO1B1, SLCO1B3

atazanavir and ritonavir, clarithromycin(d), cyclosporine(c,d), gemfibrozil(e), lopinavir and ritonavir, rifampin (single dose)(d)

 

OAT1,

OAT3

SLC22A6, SLC22A8

probenecid, teriflunomide(b,c)

 
 

 

MATE1,

MATE2-K,

OCT2

SLC47A1, SLC47A2,
SLC22A2

cimetidine, dolutegravir, isavuconazole, pyrimethamine, ranolazine, trilaciclib, vandetanib

   

Note: Criteria for selecting in vivo inhibitors are as follows:

  • P-gp: (1) AUC fold-increase of dabigatran, digoxin, or edoxaban is ≥1.5 with co-administration; and (2) in vitro inhibitor of P-gp.
  • BCRP: (1) AUC fold-increase of rosuvastatin or sulfasalazine is ≥1.5 with co-administration and (2) in vitro inhibitor of BCRP.
  • OATP1B1/OATP1B3: (1) AUC fold-increase is ≥2 for at least one of clinical substrates in Table 5-1 with co-administration; and (2) in vitro inhibitor of OATPB1 and/or OATP1B3.
  • OAT1/OAT3: (1) AUC fold-increase ≥1.5 for at least one of clinical substrates in Table 5-1 with co-administration and (2) in vitro inhibitor of OAT1 and/or OAT3.
  • OCT2/MATE: (1) AUC fold-increase of metformin is ≥ 1.5 with co-administration: and (2) in vitro inhibitor of OCT2 and/or MATEs.

This table provides examples of clinical inhibitors for various transporters and is not intended to be an exhaustive list. DDI data were collected based on a search of the University of Washington Metabolism and Transport Drug Interaction Database [Hachad et al. (2010), Hum Genomics, 5(1):61].

a A number of P-gp inhibitors also inhibit CYP3A.
b Also an inhibitor of OATP1B1 and/or OATP1B3.
c Also an inhibitor of BCRP.
d Also an inhibitor of P-gp.
e Also an inhibitor of OAT3.
f Intravenously administered rolapitant does not inhibit BCRP.

Abbreviations:
AUC: area under the plasma concentration-time curve. BCRP: breast cancer resistance protein; MATE: multidrug and toxin extrusion protein; OAT: organic anion transporter; OATP: organic anion transporting polypeptide; OCT: organic cation transporter; P-gp: P-glycoprotein, also called as multidrug resistance protein 1 (MDR1).

 

Back to Top