Skip to main content
Clinical and Translational Science logoLink to Clinical and Translational Science
. 2023 Apr 23;16(6):922–936. doi: 10.1111/cts.13507

Assessment of the drug–drug interaction potential for therapeutic proteins with pro‐inflammatory activities

Yanke Yu 1,, Charity Henrich 2, Diane Wang 1
PMCID: PMC10264930  PMID: 36890677

Abstract

It is well‐recognized that therapeutic proteins (TPs) with pro‐inflammatory activities elevate the pro‐inflammatory cytokines and result in cytokine‐drug interactions. In the current review, several pro‐inflammatory cytokines, including IL‐2, IL‐6, IFN‐γ, and TNF‐α, as well as an anti‐inflammatory cytokine IL‐10, were summarized for their respective effect on major cytochrome P450 enzymes and efflux transporter PgP. Pro‐inflammatory cytokines are generally associated with suppression of CYP enzymes across assay systems but have varied effect on Pgp expression levels and activities depending on the individual cytokines and assay systems, whereas IL‐10 had no significant impact on CYP enzymes and P‐gp. A cocktail drug‐drug interaction (DDI) study design could be an ideal approach for simultaneously assess the impact of TPs with pro‐inflammatory activities on multiple CYP enzymes. Clinical DDI studies using the cocktail approach have been conducted for several TPs with pro‐inflammatory activities and for those TPs with pro‐inflammatory activities which had no clinical DDI study conducted, languages for potential DDI risk due to cytokine‐drug interaction were included in the label. Up to date drug cocktails, including clinically validated and unvalidated for DDI assessment, were summarized in this review. Most clinically validated cocktails focused either on CYP enzymes or transporters. Additional effort was needed to validate a cocktail to include both the major CYP enzymes and key transporters. In silico methods for assessment of the DDI for TPs with pro‐inflammatory activities were also discussed.

INTRODUCTION

The importance of drug–drug interactions (DDI) in drug development and clinical practice is clear: DDIs may increase the risk of treatment failures and the incidence and severity of adverse events. The successful evaluation and prediction of DDIs during different phases of drug development and regulatory evaluation is critical to mitigate this risk.

Cytokines are a class of secreted small proteins (~5–25 kDa) that regulates the cell survival, growth, differentiation, and effector function. 1 Some cytokines, especially pro‐inflammatory cytokines, are elevated during host immune responses to inflammation, infection, trauma, or cancer, and play an important role by controlling immune cell maturation, growth, and responsiveness. There is long‐standing evidence for cytokine‐drug interaction with early clinical evidence in the 1970s, which demonstrated that acute inflammatory responses (associated with cytokine release) to infection or tissue injury could lead to altered drug pharmacokinetics. 2 , 3 , 4 Quinine exposure was increased during induced malaria and induced fever, and theophylline had a longer half‐life and therefore higher exposure during acute respiratory viral illness. 3 , 4 With the advancement in the knowledge of cytokines and drug metabolism, direct link of cytokines and drug metabolizing enzymes were identified. For instance, interleukin 6 (IL‐6) was found to be critical in reduction of cytochromes P450 (CYP) enzymes CYP1A2, CYP2A5, and CYP3A11 mRNA levels during turpentine‐induced inflammation but not during LPS‐mediated inflammation in mice. 5 In general, cytokines downregulate CYP enzyme levels, reduce CYP enzyme substrates metabolism, and increase drug exposures.

Therapeutic proteins (TPs) with pro‐inflammatory activities are a class of TPs with the properties either as pro‐inflammatory cytokines, such as peginterferon, or as pro‐inflammatory cytokine modulators, such as blinatumomab. By definition, TPs with pro‐inflammatory activities could result in persistent or transient elevation of cytokine levels in the body, therefore, administration of TPs with pro‐inflammatory activities could lead to DDIs with co‐administered drugs owing to the cytokine‐drug interaction. 6 , 7 Recent US Food and Drug Administration (FDA) draft guidance on “Drug‐drug interaction assessment for therapeutic proteins” laid out recommendations regarding assessing DDI risks for TPs. 8 For TPs as pro‐inflammatory cytokines, a clinical DDI study is recommended, and, for TPs as pro‐inflammatory cytokine modulators, a clinical DDI study may not be conducted but label language to include potential CYP/transporter mediated DDIs.

The objectives of the current review are to provide a summary of key pro‐inflammatory cytokines (IL‐2, IL‐6, TNF‐α, and IFN‐γ) and their effect on major CYP enzymes and efflux transporter P‐glycoprotein (P‐gp); clinically observed DDIs for TPs with pro‐inflammatory activities; possible drug cocktails for clinical DDI study to evaluate such DDIs; and in silico methods for assessment of such DDIs.

Cytokines effect on CYP enzymes and transporters

IL‐2

IL‐2 is a 15.5‐16‐kDa four‐α‐helix‐bundle cytokine, secreted primarily by antigen‐stimulated CD4+ T cells, and also can be secreted by CD8+ T cells, NK cells, and activated dendritic cells. 9 IL‐2 plays an important role in immune homeostasis and activation via stimulation both regulatory T cells and cytotoxic effector T cells.

IL‐2 treatment of cryopreserved human hepatocytes had no significant impact on either mRNA levels or enzyme activities for CYP1A2, 2C9, or 3A4, but modestly decreased CYP2B6 (mRNA only) and CYP2C19 (enzyme activity only) by less than 25% (Table 1). 10 On the contrary, the CYP2D6 mRNA expression increased ~50%, whereas CYP2D6 enzyme activity decreased ~22%. 10 In primary human hepatocyte culture, IL‐2 was shown to have transient suppression of CYP3A activity, but demonstrated a sustained and concentration‐dependent 50% to 70% suppression of CYP3A activity in hepatocyte/Kupffer cell coculture. 11 Indinavir, primarily eliminated by CYP3A4 metabolism, was found to have significant increase in area under the curve (AUC; 88% increase on day 5 vs. day 1) after IL‐2 infusion in patients infected with the human immunodeficiency virus. 12 In addition, high‐dose daily administration (9 or 12*106 units/m2) of IL‐2 in patients with hepatic metastases from colon or rectum carcinomas resulted in significant reduction of total CYP enzyme protein level by 34%, CYP1A2 protein level by 37%, CYP2C protein level by 45%, CYP2E1 protein level by 60%, and CYP3A4 protein level by 39%. 13

TABLE 1.

The impact of cytokines on CYP enzymes.

Cytokines Assay system CYP1A2 CYP2B6 CYP2C8 CYP2C9 CYP2C19 CYP2D6 CYP3A4 References
IL‐2 Cryopreserved human hepatocyte ↔ mRNA and activity ↓ mRNA (<25%) only ↔ mRNA and activity ↓ activity (<25%) only

↑ mRNA (~50%)

↓ activity (<22%)

↔ mRNA and activity 10
Primary human hepatocyte ↓ activity (transient) 11
Primary human hepatocyte/Kupffer coculture ↓ activity (50% ‐ 70%) 11
Human ↓ protein (37%) ↓ protein (45%) ↓ protein (45%) ↓ protein (45%) ↓ protein (39%) 13
hPBMCs ↓ mRNA and protein ↓ mRNA and protein 17
IL‐6 Primary human hepatocyte and hepatocyte/Kupffer coculture ↓ activity (~80% ‐ 90%) 11
Primary human hepatocyte

↓ mRNA (~75%)

↓ protein (~60%)

↓ mRNA

(~50%)

↓ mRNA (~35%)

↓ protein (~80%)

↓ mRNA

(~40%)

↓ mRNA (~90%)

↓ protein (~50%)

20
Primary human hepatocyte

↓ mRNA (>75%)

↓ activity (NS)

↓ mRNA (>60%)

↓ activity (NS)

↓ mRNA (>60%)

↓ activity (NS)

↓ mRNA (>60%)

↓ activity (NS)

↓ mRNA (>30%)

↓ activity (NS)

↓ mRNA (~50%)

↓ mRNA (>80%)

↓ activity (NS)

21
HepaRG cells

↓ mRNA (>80%)

↓ activity (>80%)

↓ mRNA (>60%)

↓ activity (>60%)

↓ mRNA (>60%)

↓ activity (>80%)

↓ mRNA (>75%)

↓ activity (>60%)

↓ mRNA (>75%)

↓ activity (>60%)

↓ mRNA (>80%)

↓ activity (>80%)

21
Cryopreserved human hepatocyte

↓ mRNA (27%)

↓ activity (22%)

↓ mRNA (63%)

↓ activity (30%)

↓ mRNA (63%)

↓ activity (35%)

↓ mRNA (72%)

↓ activity (65%)

↑ mRNA (2.4‐fold)

↓ activity (39%)

↓ mRNA (98%)

↓ activity (76%)

10
Caco‐2 cells ↓ mRNA (~15%) 22
hPBMCs

↔ mRNA

↓ protein (~40%)

↔ mRNA

↓ protein (~20%)

17
IFN‐γ Primary human hepatocytes

↓ mRNA (~75%)

↓ protein (~70%)

↓ mRNA (~50%)

↓ protein (~80%)

↔ mRNA

↓ protein (~60%)

↔ mRNA

↓ protein (~60%)

↓ mRNA (~70%)

↓ protein (~50%)

20
hPBMCs

↓ mRNA (~60%)

↓ protein (~60%)

↓ mRNA (~40%)

↓ protein (~50%)

17
Caco‐2 cells ↔ mRNA 22
TNF‐α Primary human hepatocytes

↔ mRNA

↓ protein (~80%)

↓ mRNA (~60%)

↓ protein (~80%)

↔ mRNA

↔ mRNA

↓ mRNA (~80%)

↓ protein (~60%)

20
Cryopreserved human hepatocytes

↓ mRNA (45%)

↓ activity (~75%)

↔ mRNA

↓ activity (~35%)

↔ mRNA

↓ activity (~20%)

↔ mRNA

↓ activity (~80%)

↓ mRNA (45%)

↓ activity (~45%)

↓ mRNA (87%)

↓ activity (~70%)

10
HepaRG cells

↓ mRNA (>40%)

↓ activity (>80%)

↓ mRNA (>40%)

↓ activity (>80%)

↓ mRNA (>40%)

↓ activity (~30%)

↓ mRNA (>40%)

↓ activity (>80%)

↓ mRNA (>40%)

↓ activity (>80%)

↓ mRNA (>40%)

↓ activity (>80%)

21
hPBMCs

↑ mRNA

↑ protein

↔ mRNA

↔ protein

17
Caco‐2 cells ↔ mRNA 22
IL‐10 hPBMCs

↔ mRNA

↔ protein

↔ mRNA

↔ protein

17
Human ↔ activity ↔ activity ↔ activity ↓ activity (~12%) 35

Abbreviations: hPBMCs, human peripheral blood mononuclear cells; NS, not significant.

IL‐2 significantly reduced P‐gp mRNA expression in human colon carcinoma cell lines Lo VO, HT115, and SW480 cells, but had no effect on P‐gp expression in LS174T cells (Table 2). 14 In another two human colon carcinoma cells, HCT15 and HCT116, a transient and reversible reduction of P‐gp mRNA and protein levels when incubated with IL‐2 was observed. 15 In a mouse model, chronic IL‐2 treatment led to a significant decrease of 57% in P‐gp protein expression but no marked differences in P‐gp mRNA level in the intestines, and, as a result, IL‐2 pretreatment increased orally administered P‐gp substrate digoxin exposure by 2.8‐fold in mice. 16 Incubation of IL‐2 with human peripheral blood mononuclear cells (hPBMCs) resulted in reduced CYP2B6 and CYP3A4 mRNA and protein levels; in contrast, IL‐2 increased the P‐gp mRNA level to ~4.7‐fold and protein level to ~2‐fold, and resulted in reduced cellular accumulation of digoxin by 17% and saquinavir by 28%. 17 In another study using hPBMCs, IL‐2 incubation was shown to significantly increase P‐gp mRNA and protein levels in lymphocytes, and flow cytometry further confirmed that P‐gp expression was significantly augmented on CD4+, CD8+, and CD19+ cells. 18

TABLE 2.

The impact of cytokines on P‐gp.

Assay system IL‐2 IL‐6 IFN‐γ TNF‐α IL‐10 References
Human colon carcinoma cell lines Lo VO, HT115, SW480 ↓ mRNA ↓ mRNA ↓ mRNA 14
Human colon carcinoma cell line LS174T ↔ mRNA ↔ mRNA ↔ mRNA 14
Human colon carcinoma cell line HCT15, HCT116 ↓ mRNA and protein (transient) 15
hPBMCs

↑ mRNA (~4.7‐fold)

↑ activity (~2‐fold)

↔ mRNA

↑ protein (~40%)

↑ mRNA (>50‐fold)

↑ protein (~2‐fold)

↑ mRNA

↑ protein

↔ mRNA

↔ protein

17
hPBMCs ↑ mRNA and activity 18
Primary human hepatocyte ↓ mRNA (~40%) 21
HepaRG cells ↓ mRNA (~20%) ↓ mRNA (~12%) 21
Caco‐2 cells ↑ mRNA (~20%) ↑ mRNA (~20%) ↔ mRNA 22
HuH7 cells

↓ mRNA (~35%)

↓ protein (~20%)

↓ activity (~20%)

↓ mRNA (~25%)

↓ protein (~35%)

↓ activity (~35%)

23
HepaG2 cells ↔ mRNA, protein, and activity

↓ mRNA (~25%)

↓ protein (~35%)

↓ activity (~35%)

23
Primary human hepatocyte

↓ mRNA (~35%)

↔ protein

↔ mRNA

↔ protein

24
hCMEC/D3 cells

↓ mRNA (21%)

↔ protein, and activity

↑ mRNA (49%)

↑ protein

↔ activity

25
hCMEC/D3 cells

↓ mRNA (43%)

↔ activity

26
hPBMC derived macrophages

↑ mRNA

↑ activity

28
Primary lymphocytes and monocytic cell lines ↔ mRNA 28
Caco‐2 cells

↑ mRNA (2.5‐fold)

↑ protein (~50%)

↔ activity

↓ mRNA (~56%)

↓ activity (~20%)

29
Caco‐2 cells ↑ protein (~2‐fold) 30
iHBMEC and pHBMEC

↑ mRNA

↔ protein

↓ activity

↑ mRNA

↔ protein

↓ activity

31

Abbreviations: hPBMCs, human peripheral blood mononuclear cells; iHBMEC, immortalized human brain microvascular endothelial cell lines; NS, not significant; pHBMEC, primary human brain microvascular endothelial cell lines.

IL‐6

IL‐6 is a 21‐26‐kDa four‐α‐helix‐bundle cytokine with an additional short α‐helix in the CD loop, secreted primarily by various cell types, including fibroblasts, endothelial cells, macrophages, T cells, and myocytes. 19 IL‐6 plays an important role in host defense through the stimulation of acute phase responses, hematopoiesis, and immune reactions. 19

IL‐6 was shown to have significant suppression of CYP3A activity (~80%–90%) in both human primary hepatocyte culture and hepatocyte/Kupffer cell coculture (Table 1). 11 In another study with primary human hepatocytes, IL‐6 was shown to significantly reduce the mRNA levels of all the CYP isoforms tested (CYP2C8, 2C9, 2C19, 3A4, and 2B6), with the least at ~35% for CYP2C9 and the greatest reduction at ~90% for CYP3A4. 20 IL‐6 also reduced CYP2B6, 2C9, and 3A4 protein levels by ~50–80%. 20 Similarly, co‐incubation of primary human hepatocytes with IL‐6 downregulated the major CYP enzymes mRNA by at least 40%, with CYP1A2 decreased by greater than 75%, CYP3A4 by greater than 80%, CYP2C9 by greater than 60%, and CYP2D6 by ~50%, and P‐gp mRNA decreased by ~40%; similar findings were observed in HepaRG cells with significant downregulation by at least 60% for almost all P450 isoforms with co‐incubation of IL‐6, but only marginal reduction in the P‐gp mRNA level. 21 Consistently, IL‐6 treatment significantly reduced the enzymes activities of P450s 1A2, 2B6, 2C8/9/19, and 3A4 in HepaRG cells, and markedly reduced the enzymes’ activities in primary human hepatocyte but not statistically significant due to high interindividual variability among the three donors. 21 IL‐6 treatment of cryopreserved human hepatocytes also significantly decreased enzyme activities of all six CYP isoforms examined (CYP1A2, 2B6, 2C9, 2C19, 2D6, and 3A4), ranging from the least at 22% for CYP1A2 to the greatest at 76% for CYP3A4. 10 The CYP mRNA expression reduction was similar to the decrease in enzyme activity, except that CYP2D6 enzyme activity decreased by 39%, but the mRNA level increased by 2.4‐fold. 10

IL‐6 treatment with Caco‐2 cells, a model of the intestinal epithelial barrier, had reduced CYP3A4 mRNA expression by ~15% (Table 1), and increased P‐gp mRNA expression by ~20% (Table 2). 22 Whereas, in the human hepatoma cell line HuH7, IL‐6 treatment significantly reduced P‐gp mRNA level ~35% and transporter activity by ~20%, but did not impact P‐gp mRNA, protein expression or transporter activity in HepG2 cells. 23 IL‐6 treatment with primary human hepatocyte modestly reduced the P‐gp mRNA level by ~35% but had no impact on the protein level. 24 Incubation of IL‐6 with hPBMCs had no significant impact on CYP2B6, CYP3A4, and P‐gp mRNA levels but slightly reduced CYP2B6 and CYP3A4 protein levels, and increased the P‐gp protein level. 17 In the human brain endothelial cell line hCMEC/D3 cell line, in vitro model of the human blood–brain barrier (BBB), IL‐6 treatment slightly decreased the P‐gp mRNA level by 21% and had no significant impact on protein expression, and transporter activity. 25 Similar findings were observed in another study, IL‐6 treatment with hCMEC/D3 cell line reduced the P‐gp mRNA level by 43%, but only had mild modification of P‐gp transporter activity. 26

IFN‐γ

IFN‐γ is a dimerized cytokine, secreted primarily by T helper cell type 1 (Th1) cells, CD8+ T cells, B cells, NK cells, and antigen presenting cells (monocytes, macrophages, and dendritic cells). 27 The monomer has a molecular weight of 20–25 kDa, and it is composed of a core of six‐α‐helix‐bundle and an extended C‐terminal region. IFN‐γ plays an important role in host defense by mediating both innate and adaptive immune responses, also, IFN‐γ can exert both antitumor and pro‐tumor effects.

IFN‐γ significantly reduced CYP2C8, 3A4, and 2B6 by ~50% to 90%, but had no significant effect on 2C9 and 2C19 mRNA in primary human hepatocytes (Table 1). 20 IFN‐γ also reduced CYP2B6, 2C9, and 3A4 protein levels by ~60–80%. 20 Incubation of IFN‐γ with hPBMCs resulted in reduced CYP2B6 and CYP3A4 mRNA and protein levels; in contrast, IFN‐γ dramatically increased P‐gp mRNA and protein levels, and resulted in reduced cellular accumulation of digoxin by 26% and saquinavir by 30%. 17

IFN‐γ significantly reduced P‐gp mRNA expression in Lo VO, HT115, and SW480 cells, but had no effect on P‐gp expression in LS174T cells (Table 2). 14 IFN‐γ upregulated P‐gp expression and increased P‐gp transport activity in a dose‐ and time‐dependent manner in human peripheral blood monocyte‐derived macrophages. The upregulation of P‐gp by IFN‐γ is the specific response of primary macrophages, as it has no impact on P‐gp expression in primary lymphocytes and monocytic cell lines. 28 IFN‐γ treatment with Caco‐2 cells had no significant impact on CYP3A4 mRNA expression, but modestly increased P‐gp mRNA expression by ~20%. 22 In another study with Caco‐2 cells, IFN‐γ treatment increased the P‐gp mRNA to 2.5‐fold, and a time‐dependent increase of protein level with maximum increase ~50% at 24 h, but did not impact transporter activity. 29 Similarly, Dixit et al. 30 reported that IFN‐γ increased P‐gp protein level in a concentration‐ and time‐dependent manner in Caco‐2 cells with maximum increase by approximately twofold. In both immortalized and primary human brain microvascular endothelial cell lines (iHBMEC and pHBMEC), another in vitro BBB model, IFN‐γ treatment increased P‐gp mRNA expression transiently with maximum effect observed at 24 h but had no effect on the P‐gp protein level. In addition, IFN‐γ treatment transiently decreased P‐gp transporter activity with a maximum effect observed at 12 h. 31

TNF‐α

TNF‐α exits as two forms, a 26 kDa transmembrane form and a soluble 17 kDa form, which is the extracellular domain cleaved from the transmembrane form. 32 Both forms are biologically active but require trimerization for the activity. TNF‐α is predominantly produced by macrophages and monocytes. TNF‐α plays important roles in acute and chronic inflammation, immunostimulation, resistance to infection agents, antitumor response, sleep regulation, embryonic development, and inducing necrotic or apoptotic cell death. 33

In primary human hepatocytes, TNF‐α significantly reduced the mRNA levels of CYP2C8 and 3A4 by 60–80%, but had no significant effect on CYP2C9, 2C19, and 2B6 mRNA (Table 1). 20 TNF‐α also reduced CYP2B6, 2C9, and 3A4 protein levels by ~60–80%. 20 In cryopreserved human hepatocytes, TNF‐α treatment also significantly decreased enzyme activities of all six CYP isoforms examined (CYP1A2, 2B6, 2C9, 2C19, 2D6, and 3A4), with greater reduction (>70%) for CYP1A2/2C19/3A4 and relatively smaller reduction (<50%) for CYP2B6/2C9/2D6. 10 The reduction of CYP mRNA expression levels was similar to the decrease in enzyme activities for CYP1A2/2D6/3A4, whereas CYP2B6/2C9/2C19 mRNA levels were relatively unchanged. 10 Similarly, in HepaRG cells, TNF‐α treatment significantly reduced all major P450s isoform mRNA levels by at least 40%, but only slightly reduced P‐gp mRNA by ~12%; the enzymatic activities of P450s 1A2, 2B6, 2C8/9/19, and 3A4 were also reduced with most at greater than 80% reduction except about 30% reduction for CYP2C8. 21 In contrast, incubation of TNF‐α with hPBMCs significantly increased the mRNA and protein levels of CYP2B6 and P‐gp, but had no impact on CYP3A4 mRNA and protein levels. 17

TNF‐α significantly reduced P‐gp mRNA expression in Lo VO, HT115, and SW480 cells, but had no effect on P‐gp expression in LS174T cells (Table 2). 14 Mixed results were reported on the impact of TNF‐α treatment on Caco‐2 cells, Bertilsson et al. 22 indicated that TNF‐α treatment had no significant impact on CYP3A4 and P‐gp mRNA expression; whereas, Belliard demonstrated that TNF‐α treatment resulted in a time‐dependent decrease of the P‐gp mRNA level, with maximum reduction ~56% at 48 h, and also had significant but moderate (~20%) reduction in transporter activity. 29 In the human hepatoma cell line, HuH7 and HepG2 cells, TNF‐α treatment significantly reduced the P‐gp mRNA level ~25% and transporter activity by ~35%. 23 TNF‐α treatment with primary human hepatocyte had no impact on either the P‐gp mRNA level or the protein level. 24 In the human hCMEC/D3 cell line, TNF‐α treatment significantly increased P‐gp mRNA level by 49% and protein expression, but had no effect on transporter activity. 25 In another in vitro human BBB model, iHBMEC and pHBMEC cells, TNF‐α treatment also increased P‐gp mRNA expression transiently with maximum effect observed at 24 h in iHBMEC and consistently increased P‐gp mRNA expression in pHBMEC, but had no effect on the P‐gp protein level in both cell lines. 31 Interestingly, TNF‐α treatment transiently decreased P‐gp transporter activity with maximum effect observed at 12 h in iHBMEC cells, and resulted in more consistent reduction of P‐gp transporter activity in pHBMEC cells. 31

IL‐10

IL‐10 is a dimerized anti‐inflammatory cytokine, secreted primarily by monocytes, type‐II T helper cells (TH2), CD4 + CD25+ T regulatory cells, γδT cells, and activated B cells. 34 The monomer has a molecular weight of ~18 kDa, consisting of a core of six‐α‐helix‐bundle. IL‐10 plays an immunoregulatory role and has inhibitory effects on pro‐inflammatory cytokine production and function.

In general, incubation of IL‐10 with hPBMC had no significant impact on the mRNA and protein levels of CYP2B6, CYP3A4, and P‐gp, except that there is a transient slight increase in the CYP3A4 mRNA level (Tables 1 and 2). 17 In addition, IL‐10 treatment in 12 healthy volunteers did not significantly alter CYP1A2, CYP2C9, and CYP2D6 activities, but slightly reduced CYP3A activity by ~12%. 35

Clinical DDI studies involving TPs with pro‐inflammatory activity

Peginterferon alfa‐2a, a covalent conjugate of recombinant alfa‐2a interferon with a polyethylene glycol (PEG) chain, was approved for chronic hepatitis B and C. Peginterferon alfa‐2a was found to have no significant effect on the pharmacokinetics (PKs) of representative drugs metabolized by CYP2C9 (tolbutamide), CYP2C19 (mephenytoin), CYP2D6 (debrisoquine), or CYP3A4 (dapsone); and was associated with an inhibition of CYP1A2 and increased theophylline AUC 25% in healthy volunteers. 36 , 37

Peginterferon alfa‐2b, a covalent conjugate of recombinant alfa‐2b interferon with a PEG chain, was approved for chronic hepatitis C. Peginterferon alfa‐2b increased caffeine (CYP1A2 substrate) AUC 18–39% in healthy and chronic hepatitis C subjects depending on the dose regimen, increased dextromethorphan (CYP2D6 substrate) AUC 103% in healthy subjects but had no impact in chronic hepatitis C subjects, and increased desipramine (CYP2D6 substrate) AUC 30% in healthy subjects. 38 , 39 Peginterferon alfa‐2b had no significant impact on the PKs of tolbutamide (CYP2C9 substrate), midazolam (CYP3A4 substrate), and dapsone (N‐acetyltransferase substrate). 38 , 39 In another report, high‐dose interferon alfa‐2b was found to reduce CYP1A2 activity by 60% in patients with high‐risk melanoma. 40

No clinical DDI information have been reported for the other approved therapeutic proteins with pro‐inflammatory activity, including interferon alfacon‐1, interferon gamma‐1b, interferon beta‐1b, TNF‐α, aldesleukin, denileukin diftitox, blinatumomab, tebentafusp‐tebn, mosunetuzumab, and teclistamab‐cqyv. 41 , 42 , 43 , 44 , 45 , 46 , 47 , 48 , 49 , 50 , 51 However, interferon gamma‐1b had label language describing that the interferon gamma had demonstrated reduction in hepatic CYP450 concentrations in rodents, which could lead to suppression of the hepatic metabolism of certain drugs. 42 Interferon beta‐1b also had label language describing caution when interferon beta‐1b in combination with medicinal products that have a narrow therapeutic index and largely dependent on the hepatic cytochrome P450 system for clearance, as interferon was reported to be associated with reduced activity of hepatic CYP450 enzymes. 44 Blinatumomab, tebentafusp, mosunetuzumab, and teclistamab had label languages describing transient release of cytokines that may suppress CYP450 enzymes which could result in DDI risk. 48 , 49 , 50 , 51

Cocktail studies

Clinically validated cocktails involving CYP mediated DDIs

Nine clinically validated (drugs within the cocktail have no mutual interactions in clinical study) cocktails with at least five CYP enzymes were shown in Table 3, including the publication time for the final version of the cocktail, drugs in the cocktail and their doses/routes, and the applications of the cocktail with no change or minimal changes.

TABLE 3.

Clinically validated cocktails involving CYP mediated DDIs.

Pittsburgh Cocktail 52 , 53 Zhu Cocktail 54 Karolinska Cocktail 55 Blakey Cocktail 58 Cooperstown 5 + 1 Cocktail 56 , 57 Inje Cocktail 59 , 64 Turpault Cocktail 60 Basel Cocktail 61 Geneva Cocktail 62 , 63
Publication date September 2006 November 2001 June 2003 February 2004 November 2003 November 2007 December 2009 March 2014 September 2016
CYP1A2 Caffeine 100 mg Caffeine 100 mg Caffeine 100 mg Caffeine 100 mg Caffeine 2 mg/kg Caffeine 93 mg Caffeine 100 mg Caffeine 100 mg Caffeine 50 mg
CYP2B6 Efavirenz 50 mg Bupropion 20 mg
CYP2C9 Flurbiprofen 50 mg Losartan 25 mg Tolbutamide 250 mg Warfarin 10 mg Losartan 30 mg Warfarin 10 mg Losartan 12.5 mg Flurbiprofen 10 mg
CYP2C19 Mephenytoin 100 mg Mephenytoin 100 mg Omeprazole 20 mg Omeprazole 40 mg Omeprazole 20 mg Omeprazole 20 mg Omeprazole 10 mg Omeprazole 10 mg
CYP2D6 Debrisoquine 10 mg Metoprolol 100 mg Debrisoquine 10 mg Debrisoquine 5 mg Dextromethorphan 30 mg Dextromethorphan 30 mg Metoprolol 100 mg Metoprolol 12.5 mg Dextromethorphan 10 mg
CYP3A4 Dapsone 100 mg Midazolam 7.5 mg Quinine 250 mg Midazolam 0.025 mg/kg (i.v.) Midazolam 0.025 mg/kg (i.v.) Midazolam 2 mg Midazolam 0.03 mg/kg Midazolam 2 mg Midazolam 1 mg
CYP2E1 Chlorzoxazone 250 mg Chlorzoxazone 200 mg Chlorzoxazone 250 mg
Application 85, 86 76, 80, 81, 87, 88, 89, 90, 91, 92, 93 94, 95, 96, 97, 98 82

Note: The dosing route is oral, if not specified.

Abbreviations: DDI, drug‐drug interaction; IV, intravenous.

The major CYP enzymes assessed in these cocktails included CYP1A2, 2B6, 2C9, 2C19, 2D6, and 3A4. 52 , 53 , 54 , 55 , 56 , 57 , 58 , 59 , 60 , 61 , 62 , 63 Caffeine was selected as the CYP1A2 probe substrate in all these cocktails. Four CYP2C9 probe substrate drugs were used in these cocktails, including flurbiprofen, losartan, tolbutamide, and warfarin. Mephenytoin and omeprazole were used as the CYP2C19 probe substrates with omeprazole predominantly used. Debrisoquine, metoprolol, and dextromethorphan were used as the CYP2D6 probe substrates. Dapsone, quinine, and midazolam were used as the CYP3A4 probe substrates with midazolam predominantly used. CYP2B6 was only assessed in the Basel cocktail and Geneva cocktail with efavirenz and bupropion as the respective probe substrate. 61 , 63 In addition, the Pittsburgh cocktail, the Zhu cocktail, and the Blakey cocktail also included CYP2E1 with chlorzoxazone as the probe substrate. 52 , 54 , 58 Despite that some cocktails shared the same probe substrate, the dose amount and dosing route could be different. For example, midazolam was dosed intravenously in the Blakey cocktail and the Cooperstown 5 + 1 cocktail and was dosed orally in the other cocktails. All the individual tested cocktails showed that there were no mutual metabolic interactions among the included drugs at the respective doses, except that whereas caffeine, losartan, omeprazole, and quinine had no significant change when administered together compared with administered alone, an inhibition of debrisoquine metabolism was observed when concurrent administration of the five drugs in the Karolinska cocktail. 55 The Inje cocktail was further validated by the typical CYP inducer rifampicin or inhibitors cimetidine and fluvoxamine, and the degrees of interaction are consistent with the single agent studies. 64

Clinically validated cocktails involving transporter mediated DDIs

Three clinically validated cocktails involving transporters were shown in Table S1, including the publication time for the final version of the cocktail, drugs in the cocktail and their doses/routes, and the applications of the cocktail with no change or minimal changes.

A five‐drug microdose probe drug cocktail (here named as the Microdose cocktail) consisting of 10 μg midazolam (CYP3A), 375 μg dabigatran etexilate (P‐gp), 10 μg pitavastatin (OATP1B1), 50 μg rosuvastatin (BCRP and OATP1B1/1B3), 100 μg atorvastatin (CYP3A, BCRP, OATP1B1/1B3, and P‐gp) was qualified by clinical DDI studies with inducer/inhibitors of rifampin, itraconazole, and clarithromycin. 65 The observed DDIs were consistent with historical observed ones and/or in agreement with theoretical expectations, with the exception that the observed DDI for dabigatran was about twofold higher for microdose compared to that of conventional dosing, which could be due to P‐gp mediated nonlinearity in the PKs of dabigatran. 65

Stopfer et al. 66 evaluated a four‐drug cocktails (here named as the Stopfer transporter cocktail) consisting of 0.25 mg digoxin (P‐gp), 5 mg furosemide (OAT1 and OAT3), 500 mg metformin (OCT2, MATE1, and MATE2‐K), and 10 mg rosuvastatin (OATP1B1, OATP1B3, and BCRP) for evaluation of transporter mediated DDI. Approximately 40% increase in rosuvastatin exposure was observed which could be due to the metformin and furosemide had an effect on rosuvastatin. 66 As a result, reduced doses of furosemide and metformin in this four‐drug cocktail was further tested, with 0.25 mg digoxin, 1 mg furosemide, 10 mg metformin, and 10 mg rosuvastatin, and no mutual interaction was observed. 67 A follow‐up study further evaluated this cocktail with four common transporter inhibitors, rifampin (rosuvastatin inhibitor), probenecid (furosemide inhibitor), cimetidine (metformin inhibitor), and verapamil (digoxin inhibitor). Generally, the DDIs between the transporter inhibitors and substrates were consistent with the historical data, except that the effect of single dose verapamil on digoxin was less than the effect observed from multiple verapamil dose studies. 68

Trueck et al. 69 evaluated a five‐drug transporter cocktails (here named as the Trueck transporter cocktail) consisting of 10 mg adefovir dipivoxil (OAT1), 100 mg sitagliptin (OAT3), 500 mg metformin (OCT2, MATE1, and MATE2‐K), 2 mg pitavastatin (OATP1B1), and 0.5 mg digoxin (P‐gp), and no significant mutual interactions were observed.

Clinically used cocktails without validation source

In addition to the validated cocktails, other cocktails were also used for evaluation of complicated DDIs, although no clinical data can be identified to illustrate whether there is mutual interaction among the cocktail drugs. The unvalidated cocktails can be found in Table S2.

In silico methods

The cocktail DDI studies could be helpful for evaluating the potential DDI risks for TPs with pro‐inflammatory activities with the advantage of simultaneous evaluation of multiple CYP enzymes. In silico methods might also be a viable approach to assess such DDIs.

Machavaram et al. 70 developed a physiologically‐based pharmacokinetic (PBPK) model for IL‐6 incorporating in vitro IL6 CYP suppression kinetics from hepatocytes and evaluated the impact of IL‐6 on CYP3A4 substrates (simvastatin and cyclosporine). A steady‐state concentration of 100 pg/mL IL‐6 was associated with increased exposure of simvastatin, and the predicted increase in AUC is comparable with the observed data (59% vs. 58%, respectively) in patients with rheumatoid arthritis (RA; patients with RA had elevated IL‐6 around 100 ng/mL). 70 , 71 Whereas, a steady‐state concentration of 500 pg/mL IL‐6 resulted in cyclosporine exposure increase and the predicted increase in AUC was consistent with the observed (45% vs. 39%, respectively) in patients with bone marrow transplant (BMT; patients with BMT had higher IL‐6 around 500 pg/mL). 70 , 72 However, in another set of patients with BMT, the predicted cyclosporine increase by IL‐6 was less than clinically observed (1.6–1.7‐fold vs. 3–5‐fold). Machavaram et al. further used this model with modification to predict the change of the exposure of several CYP probe substrates, CYP3A4 (simvastatin and midazolam), CYP1A2 (caffeine), CYP2C9 (S‐warfarin), CYP2C19 (omeprazole), and CYP2D6 (dextromethorphan) in subjects with RA (steady‐state IL6 concentrations of 50 or 100 pg/mL were used) and compared with healthy subjects and found reasonable agreement. 73 , 74

Xu et al. 75 developed a PBPK model to predict the DDI potential of blinatumomab, a TP with pro‐inflammatory activity which can transiently elevate multiple cytokines, including IL‐6, IL‐10, and IFN‐gamma. The model focused on IL‐6 mediated DDI and time‐concentration profile of IL‐6 from patients with non‐Hodgkin's lymphoma receiving blinatumomab was simulated and in vitro IL‐6 CYP suppression kinetics were incorporated into the model. The predicted exposure increases for sensitive substrates of CYP3A4 (simvastatin and midazolam), CYP1A2 (theophylline and caffeine), and CYP2C9 (S‐warfarin) ranged from 1.2‐ to 1.9‐fold, and lasted less than 1 week, indicating transient and weak DDI potential for blinatumomab. 75

The exposure of several CYP probe substrate drugs were altered in patients with RA at pre‐ and post‐sirukumab (an anti‐IL‐6 monoclonal antibody) treatment. 76 Midazolam, omeprazole, and S‐warfarin exposure reduced about 33%, 40%, and 18%, respectively; whereas caffeine exposure increased ~25%. 76 A PBPK model was used to predict the impact of elevated IL‐6 and sirukumab on CYP enzymes in patients with RA. The predicted exposure of several CYP probe substrates, caffeine (CYP1A2), S‐warfarin (CYP2C9), omeprazole (CYP2C19), and midazolam (CYP3A4) in patients with RA pre‐ and post‐sirukumab treatment (IL‐6 assumed to be 50 and 0 pg/mL for pre‐ and post‐sirukumab treatment, respectively) were consistent with the observed from the sirukumab clinical TP‐DDI study. 74

The PBPK model was also used to simulate IL‐6 induced P‐gp activity reduction in BBB and examine its impact on P‐gp substrate digoxin exposure in the brain. 26 The simulation showed that the digoxin brain exposure is marginally affected with IL‐6 treatment (a nonsignificant 4% increase of AUCinf).

DISCUSSION

There is an increasing number of TPs, including TPs with pro‐inflammatory activities in the different stages of drug development. In this review, we examined the DDI potential for TPs with pro‐inflammatory activities via cytokine‐drug interaction. Specifically, several key cytokines and their impact on major CYP enzymes and P‐gp, the clinical DDI information of TPs with pro‐inflammatory activities, drug cocktails for clinical DDI studies, and in silico methods for evaluating clinical DDI potential were summarized.

Among the pro‐inflammatory cytokines examined, IL‐6 exhibited consistent and profound effect in reducing the major CYP enzymes mRNA and protein levels as well as enzymatic activities across different assay systems, except that CYP2D6 mRNA in cryopreserved human hepatocyte was increased and CYP2B6 and CYP3A4 mRNA in hPBMCs were not changed. However, CYP2D6 activity in cryopreserved human hepatocytes and CYP2B6 and CYP3A4 protein levels in hPBMCs were reduced with IL‐6 treatment. Compared to IL‐6, IFN‐γ and TNF‐α showed slightly less effect but also significantly reduced the mRNA, protein levels, and enzymatic activities of the major CYP enzymes in most assay systems. IL‐2 had the least activity in suppression of CYP enzymes, and the mechanism in suppression of CYP enzymes for IL‐2 might be different from IL‐6, IFN‐γ, and TNF‐α, because IL‐2 had minor or only transient effect on CYP enzymes in primary or cryopreserved human hepatocytes but significantly reduced CYP3A4 activity in primary human hepatocyte/Kupffer coculture; in contrast, IL‐6, IFN‐γ, and TNF‐α had similar effect on CYP enzymes in primary or cryopreserved human hepatocytes with/without Kupffer coculture. Therefore, IL‐2 might suppress CYP enzymes in hepatocytes indirectly through inducing IL‐6 and other pro‐inflammatory cytokines in Kupffer cells. 11 In addition to the in vitro assay systems, patients with hepatic metastases treated with IL‐2 resulted in CYP enzyme reduction in a clinical study. 13 For comparison purpose, we also examined the effect of an anti‐inflammatory cytokine, IL‐10, on CYP enzymes. Not surprisingly, IL‐10 had negligible effect on CYP enzymes in hPBMCs and in humans from a clinical study.

In addition to the CYP enzymes, the impact of cytokines on transporter P‐gp was also examined. The impact of cytokines on transporter P‐gp is varied and dependent on assay systems. In general, the pro‐inflammatory cytokines IL‐2, IL‐6, IFN‐γ, and TNF‐α increased P‐gp mRNA and protein levels in hPBMC; whereas the impact of different cytokines on the other assay systems, including human colon carcinoma cell lines, Caco‐2 cells, hepatocyte, HepaG2 cells, and in vitro BBB model cells were varied and could be a reduction, no change, or increase. In addition, IL‐10 had no impact on P+gp. In this review, we only focused on P‐gp as a prototypical transporter. However, other transporters can also be affected by the pro‐inflammatory cytokines. For instance, IL‐6 and TNF‐α significantly reduced BCRP mRNA level and activity in the human hCMEC/D3 cell line. 25 TNF‐α and IL‐2 downregulated NTCP, OATP1B1, OATP1B3, OATB2B1, OCT1, and OAT2 mRNA levels, and reduced NTCP, OATP1B1 protein levels and NTCP, OATP, and OCT1 transporter activities; IL‐6 decreased mRNA level of MDR1, MRP2, and BCRP, and also MRP2 and BCRP protein expression in primary human hepatocyte. 24 TNF‐α but not IL‐6 reduced BSEP mRNA level and increased BCRP protein expression, and TNF‐α and IL‐6 increased MRP3 protein level. 24

The current review focused on several key pro‐inflammatory cytokines (IL‐2, IL‐6, TNF‐α, and IFN‐γ) and anti‐inflammatory cytokine IL‐10 for comparison. Other pro‐inflammatory cytokines could also have impact on the CYP enzymes and transporters. For example, theophylline (a CYP1A2 probe substrate) was shown to have a reduced clearance by ~26% in patients with chronic hepatitis C treated with interferon beta 77 ; whereas, interferon beta had no significant effect on exposure of mephenytoin (CYP2C19 probe substrate) and debrisoquine (CYP2D6 probe substrate) in patients with multiple sclerosis. 78 In addition, incubation of Hepa‐RG cells with IL‐1β resulted in significant reduction of the mRNA levels (>80%) and enzyme activities (>60%) for key CYP enzymes 1A2, 2B6, 2C19, 2C8, 2C9, and 3A4 as well as P‐gp mRNA level ~55%.

Despite numerous reports indicating the impact of pro‐inflammatory cytokines on CYP enzymes and transporters, limited clinical DDI studies were conducted for the approved TPs with pro‐inflammatory activities, with the exception for Peginterferon alfa‐2a and Peginterferon alfa‐2b. However, most of the approved TPs with pro‐inflammatory activities had label languages regarding the potential DDI due to suppression of CYP enzymes. The clinical DDI studies for Peginterferon alfa‐2a and Peginterferon alfa‐2b adopted cocktail approach, which enables assessment of DDIs for multiple CYP enzymes simultaneously to increase study efficiency and cost saving. 39 , 79 In addition, due to the cytokine effect on the CYP enzymes, the CYP enzymes are suppressed in certain diseases with elevated pro‐inflammatory cytokines, such as plaque psoriasis and RA. Therefore, drugs used to treat these diseases could often result in disease‐drug interaction as the treatment normalized the cytokines and the CYP enzymes levels and thus altered the exposure of CYP enzymes substrate drugs post‐treatment compared to pretreatment. Clinical DDI studies have been conducted to evaluate such disease‐drug interactions, some using the cocktail approach. 71 , 76 , 80 , 81 , 82

In this review, we summarized the most up‐to‐date validated cocktails for CYP enzymes and validated cocktails involving transporters, as well as some other cocktails used in the clinical DDI studies, although no validation source can be identified. Almost all the validated cocktails were either for CYP enzymes or for transporters, except that a microdose cocktail included CYP3A4 probe substrate and several transporters probe substrates. 65 Efforts have been made to combine the CYP cocktails with transporter cocktails. For example, Kwon et al. 83 developed and validated a 10‐probe drug cocktail, including five major CYP probe substrates and five transporters probe substrates in rat. Bosilkovska et al. 84 explored the possibility to combine the Geneva cocktail (6 CYP enzymes) with fexofenadine (P‐gp) and found that the Geneva cocktail co‐administration with 25 mg fexofenadine resulted in 49% reduction in maximum concentration and 43% reduction in AUC0‐8 for fexofenadine; hence, fexofenadine cannot be combined with the Geneva cocktail. Currently, there is no validated cocktail in the clinic involving major CYP enzymes and transporters, although several unvalidated cocktails consisting of several major CYP enzymes and transporters were used in the clinical studies to assess the clinical DDIs (Table S2).

In silico methods have also been used to evaluate the complicated DDIs, whereas the population PK approach still needs clinical data to assess the DDIs, the bottom‐up approach PBPK can directly extrapolate the clinical DDI with in vitro data. Current PBPK models for assessing cytokine‐drug DDIs all focus on IL‐6, whereas most of the models can reasonably predict the clinical DDIs based on in vitro determined CYP suppression kinetics retrospectively, Machavaram et al. 70 showed that the PBPK model underpredicted exposure of cyclosporine in certain patients with BMT, which might be due to possible contribution of other cytokines (e.g., TNF‐α) in the overall suppression of CYP3A4 in these patients, and the PBPK model incorporating IL‐6 CYP3A4 suppression alone may underestimate the suppression effect. Therefore, additional efforts may be needed to further evaluate the PBPK models for the other pro‐inflammatory cytokines and/or a PBPK model incorporating multiple pro‐inflammatory cytokines. Moreover, limited PBPK modeling is available to evaluate the DDI potential of TPs with pro‐inflammatory activities with drugs via transporters, and such PBPK models need further development.

Considering the effect of pro‐inflammatory cytokines on the major CYP enzymes and P‐gp, for TPs with pro‐inflammatory activity, a clinical DDI study is recommended for those TPs that can persistently elevate the cytokines and, in this case, the cocktail approach may be used owing to its efficient study design and cost saving; whereas for those TPs transiently elevating cytokines, a clinical DDI study may not be conducted and an in silico method may be used to assess such transient effect, and label language should incorporate the potential DDIs due to cytokine‐drug effect.

CONCLUSION

Potential DDI risk for TPs with pro‐inflammatory activities via cytokine‐drug interaction has been well‐recognized. Pro‐inflammatory cytokines are generally associated with suppression of CYP enzymes but have varied effects on P‐gp expression levels and activities. The approved TPs with pro‐inflammatory activities generally contain label languages for potential DDI risks due to cytokine elevation. Cocktail studies have been used to evaluate the complicated DDIs involving multiple CYP enzymes and transporters with the advantage of efficiency and cost saving, and it is a viable approach to assess the DDI for TPs with pro‐inflammatory activities. In silico methods are also viable approaches to assess the DDIs for TPs with pro‐inflammatory activities.

FUNDING INFORMATION

No funding was received for this work.

CONFLICT OF INTEREST STATEMENT

Y.Y. was employed at Pfizer when work was conducted and received salary and stock; currently employed at Genentech and owns stock in Roche. D.W. is an employee of Pfizer, Inc. and owns stock in Pfizer Inc. C.H. has no competing interests to declare.

Supporting information

Table S1

Yu Y, Henrich C, Wang D. Assessment of the drug–drug interaction potential for therapeutic proteins with pro‐inflammatory activities. Clin Transl Sci. 2023;16:922‐936. doi: 10.1111/cts.13507

REFERENCES

  • 1. Janeway's Immunobiology. Garland Science; 2017. [Google Scholar]
  • 2. Haas CE. Drug‐cytokine interaction. In: Piscitelli SC, Rodvold KA, eds. Drug interactions in infectious diseases. Humana Press Inc; 2001:287‐310. [Google Scholar]
  • 3. Chang KC, Bell TD, Lauer BA, Chai H. Altered theophylline pharmacokinetics during acute respiratory viral illness. Lancet. 1978;1:1132‐1133. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4. Trenholme GM, Williams RL, Rieckmann KH, Frischer H, Carson PE. Quinine disposition during malaria and during induced fever. Clin Pharmacol Ther. 1976;19:459‐467. [DOI] [PubMed] [Google Scholar]
  • 5. Siewert E, Bort R, Kluge R, Heinrich PC, Castell J, Jover R. Hepatic cytochrome P450 down‐regulation during aseptic inflammation in the mouse is interleukin 6 dependent. Hepatology. 2000;32:49‐55. [DOI] [PubMed] [Google Scholar]
  • 6. Jing X, Ji P, Schrieber SJ, Fletcher EP, Sahajwalla C. Update on therapeutic protein‐drug interaction: information in labeling. Clin Pharmacokinet. 2020;59:25‐36. [DOI] [PubMed] [Google Scholar]
  • 7. Coutant DE, Boulton DW, Dahal UP, et al. Therapeutic protein drug interactions: a white paper from the international consortium for innovation and quality in pharmaceutical development. Clin Pharmacol Ther. 2022. Online ahead of print. [DOI] [PubMed] [Google Scholar]
  • 8. US FDA draft guidance: Drug‐drug interaction assessment for therapeutic proteins. 2020.
  • 9. Rosenberg SA. IL‐2: the first effective immunotherapy for human cancer. J Immunol. 2014;192:5451‐5458. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10. Dallas S, Sensenhauser C, Batheja A, et al. De‐risking bio‐therapeutics for possible drug interactions using cryopreserved human hepatocytes. Curr Drug Metab. 2012;13:923‐929. [DOI] [PubMed] [Google Scholar]
  • 11. Sunman JA, Hawke RL, LeCluyse EL, Kashuba AD. Kupffer cell‐mediated IL‐2 suppression of CYP3A activity in human hepatocytes. Drug Metab Dispos. 2004;32:359‐363. [DOI] [PubMed] [Google Scholar]
  • 12. Piscitelli SC, Vogel S, Figg WD, et al. Alteration in indinavir clearance during interleukin‐2 infusions in patients infected with the human immunodeficiency virus. Pharmacotherapy. 1998;18:1212‐1216. [PubMed] [Google Scholar]
  • 13. Elkahwaji J, Robin MA, Berson A, et al. Decrease in hepatic cytochrome P450 after interleukin‐2 immunotherapy. Biochem Pharmacol. 1999;57:951‐954. [DOI] [PubMed] [Google Scholar]
  • 14. Walther W, Stein U. Influence of cytokines on mdr1 expression in human colon carcinoma cell lines: increased cytotoxicity of MDR relevant drugs. J Cancer Res Clin Oncol. 1994;120:471‐478. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15. Stein U, Walther W, Shoemaker RH. Modulation of mdr1 expression by cytokines in human colon carcinoma cells: an approach for reversal of multidrug resistance. Br J Cancer. 1996;74:1384‐1391. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16. Veau C, Faivre L, Tardivel S, et al. Effect of interleukin‐2 on intestinal P‐glycoprotein expression and functionality in mice. J Pharmacol Exp Ther. 2002;302:742‐750. [DOI] [PubMed] [Google Scholar]
  • 17. Liptrott NJ, Penny M, Bray PG, et al. The impact of cytokines on the expression of drug transporters, cytochrome P450 enzymes and chemokine receptors in human PBMC. Br J Pharmacol. 2009;156:497‐508. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Tsujimura S, Saito K, Nakayamada S, et al. Transcriptional regulation of multidrug resistance‐1 gene by interleukin‐2 in lymphocytes. Genes Cells. 2004;9:1265‐1273. [DOI] [PubMed] [Google Scholar]
  • 19. Tanaka T, Narazaki M, Kishimoto T. IL‐6 in inflammation, immunity, and disease. Cold Spring Harb Perspect Biol. 2014;6:a016295. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20. Aitken AE, Morgan ET. Gene‐specific effects of inflammatory cytokines on cytochrome P450 2C, 2B6 and 3A4 mRNA levels in human hepatocytes. Drug Metab Dispos. 2007;35:1687‐1693. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21. Klein M, Thomas M, Hofmann U, Seehofer D, Damm G, Zanger UM. A systematic comparison of the impact of inflammatory signaling on absorption, distribution, metabolism, and excretion gene expression and activity in primary human hepatocytes and HepaRG cells. Drug Metab Dispos. 2015;43:273‐283. [DOI] [PubMed] [Google Scholar]
  • 22. Bertilsson PM, Olsson P, Magnusson KE. Cytokines influence mRNA expression of cytochrome P450 3A4 and MDRI in intestinal cells. J Pharm Sci. 2001;90:638‐646. [DOI] [PubMed] [Google Scholar]
  • 23. Lee G, Piquette‐Miller M. Cytokines alter the expression and activity of the multidrug resistance transporters in human hepatoma cell lines; analysis using RT‐PCR and cDNA microarrays. J Pharm Sci. 2003;92:2152‐2163. [DOI] [PubMed] [Google Scholar]
  • 24. Le Vee M, Lecureur V, Stieger B, Fardel O. Regulation of drug transporter expression in human hepatocytes exposed to the proinflammatory cytokines tumor necrosis factor‐alpha or interleukin‐6. Drug Metab Dispos. 2009;37:685‐693. [DOI] [PubMed] [Google Scholar]
  • 25. Poller B, Drewe J, Krahenbuhl S, Huwyler J, Gutmann H. Regulation of BCRP (ABCG2) and P‐glycoprotein (ABCB1) by cytokines in a model of the human blood‐brain barrier. Cell Mol Neurobiol. 2010;30:63‐70. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26. Simon F, Guyot L, Garcia J, et al. Impact of interleukin‐6 on drug transporters and permeability in the hCMEC/D3 blood‐brain barrier model. Fundam Clin Pharmacol. 2021;35:397‐409. [DOI] [PubMed] [Google Scholar]
  • 27. Bhat MY, Solanki HS, Advani J, et al. Comprehensive network map of interferon gamma signaling. J Cell Commun Signal. 2018;12:745‐751. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28. Puddu P, Fais S, Luciani F, et al. Interferon‐gamma up‐regulates expression and activity of P‐glycoprotein in human peripheral blood monocyte‐derived macrophages. Lab Investig. 1999;79:1299‐1309. [PubMed] [Google Scholar]
  • 29. Belliard AM, Lacour B, Farinotti R, Leroy C. Effect of tumor necrosis factor‐alpha and interferon‐gamma on intestinal P‐glycoprotein expression, activity, and localization in Caco‐2 cells. J Pharm Sci. 2004;93:1524‐1536. [DOI] [PubMed] [Google Scholar]
  • 30. Dixit SG, Zingarelli B, Buckley DJ, Buckley AR, Pauletti GM. Nitric oxide mediates increased P‐glycoprotein activity in interferon‐{gamma}‐stimulated human intestinal cells. Am J Physiol Gastrointest Liver Physiol. 2005;288:G533‐G540. [DOI] [PubMed] [Google Scholar]
  • 31. Lee NY, Rieckmann P, Kang YS. The changes of P‐glycoprotein activity by interferon‐gamma and tumor necrosis factor‐alpha in primary and immortalized human brain microvascular endothelial cells. Biomol Ther (Seoul). 2012;20:293‐298. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Palladino MA, Bahjat FR, Theodorakis EA, Moldawer LL. Anti‐TNF‐alpha therapies: the next generation. Nat Rev Drug Discov. 2003;2:736‐746. [DOI] [PubMed] [Google Scholar]
  • 33. Idriss HT, Naismith JH. TNF alpha and the TNF receptor superfamily: structure‐function relationship(s). Microsc Res Tech. 2000;50:184‐195. [DOI] [PubMed] [Google Scholar]
  • 34. Pestka S, Krause CD, Sarkar D, Walter MR, Shi Y, Fisher PB. Interleukin‐10 and related cytokines and receptors. Annu Rev Immunol. 2004;22:929‐979. [DOI] [PubMed] [Google Scholar]
  • 35. Gorski JC, Hall SD, Becker P, Affrime MB, Cutler DL, Haehner‐Daniels B. In vivo effects of interleukin‐10 on human cytochrome P450 activity. Clin Pharmacol Ther. 2000;67:32‐43. [DOI] [PubMed] [Google Scholar]
  • 36. PEGASYS® (peginterferon alfa‐2a): package insert. Genentech In; 2021. https://www.gene.com/download/pdf/pegasys_prescribing.pdf [Google Scholar]
  • 37. Brennan BJ, Xu ZX, Grippo JF. Effect of peginterferon alfa‐2a (40KD) on cytochrome P450 isoenzyme activity. Br J Clin Pharmacol. 2013;75:497‐506. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38. PEGINTRON® (peginterferon alfa‐2b): package insert. Merck Inc.; 2019. https://www.merck.com/product/usa/pi_circulars/p/pegintron/pegintron_5ml_pi.pdf [Google Scholar]
  • 39. Gupta SK, Kolz K, Cutler DL. Effects of multiple‐dose pegylated interferon alfa‐2b on the activity of drug‐metabolizing enzymes in persons with chronic hepatitis C. Eur J Clin Pharmacol. 2011;67:591‐599. [DOI] [PubMed] [Google Scholar]
  • 40. Islam M, Frye RF, Richards TJ, et al. Differential effect of IFNalpha‐2b on the cytochrome P450 enzyme system: a potential basis of IFN toxicity and its modulation by other drugs. Clin Cancer Res. 2002;8:2480‐2487. [PubMed] [Google Scholar]
  • 41. Infergen® (interferon alfacon‐1): package insert. 2010. https://www.accessdata.fda.gov/drugsatfda_docs/label/2010/103663s5069lbl.pdf
  • 42. Actimmune® (interferon gamma‐1b): USPI. Horizon Therapeutics Inc; 2021. https://www.hzndocs.com/ACTIMMUNE‐Prescribing‐Information.pdf [Google Scholar]
  • 43. Extavia® (interferon beta‐1b): USPI. Novartis; 2021. https://www.novartis.com/us‐en/sites/novartis_us/files/extavia.pdf [Google Scholar]
  • 44. Betaferon® (interferon beta‐1b): SmPC. Bayer AG; 2021. https://www.ema.europa.eu/en/documents/product‐information/betaferon‐epar‐product‐information_en.pdf [Google Scholar]
  • 45. Beromun® (TNF‐alpha): SmPC. Belpharma; 2022. https://www.ema.europa.eu/en/documents/product‐information/beromun‐epar‐product‐information_en.pdf [Google Scholar]
  • 46. Proleukin® (aldesleukin): USPI. Clinigen Inc; 2019. https://proleukin.com/pi/proleukin%20prescribing%20information.pdf [Google Scholar]
  • 47. Ontak® (denileukin diftitox): USPI. Eisai Inc; 2008. https://www.accessdata.fda.gov/drugsatfda_docs/label/2008/103767s5094lbl.pdf [Google Scholar]
  • 48. Blincyto® (blinatumomab): USPI. Amgen; 2022. https://www.pi.amgen.com/‐/media/Project/Amgen/Repository/pi‐amgen‐com/blincyto/blincyto_pi_hcp_english.pdf [Google Scholar]
  • 49. Kimmtrak® (tebentafusp‐tebn): USPI. Immunocore Limited; 2022. https://www.accessdata.fda.gov/drugsatfda_docs/label/2022/761228s000lbl.pdf [Google Scholar]
  • 50. Lunsumio® (mosunetuzumab): SmPC. Roche; 2022. https://www.ema.europa.eu/en/documents/product‐information/lunsumio‐epar‐product‐information_en.pdf [Google Scholar]
  • 51. Tecvayli® (teclistamab‐cqyv): USPI. Janssen Biotech; 2022. https://www.accessdata.fda.gov/drugsatfda_docs/label/2022/761291s000lbl.pdf [Google Scholar]
  • 52. Frye RF, Matzke GR, Adedoyin A, Porter JA, Branch RA. Validation of the five‐drug "Pittsburgh cocktail" approach for assessment of selective regulation of drug‐metabolizing enzymes. Clin Pharmacol Ther. 1997;62:365‐376. [DOI] [PubMed] [Google Scholar]
  • 53. Zgheib NK, Frye RF, Tracy TS, Romkes M, Branch RA. Validation of incorporating flurbiprofen into the Pittsburgh cocktail. Clin Pharmacol Ther. 2006;80:257‐263. [DOI] [PubMed] [Google Scholar]
  • 54. Zhu B, Ou‐Yang DS, Chen XP, et al. Assessment of cytochrome P450 activity by a five‐drug cocktail approach. Clin Pharmacol Ther. 2001;70:455‐461. [DOI] [PubMed] [Google Scholar]
  • 55. Christensen M, Andersson K, Dalén P, et al. The Karolinska cocktail for phenotyping of five human cytochrome P450 enzymes. Clin Pharmacol Ther. 2003;73:517‐528. [DOI] [PubMed] [Google Scholar]
  • 56. Streetman DS, Bleakley JF, Kim JS, et al. Combined phenotypic assessment of CYP1A2, CYP2C19, CYP2D6, CYP3A, N‐acetyltransferase‐2, and xanthine oxidase with the "Cooperstown cocktail". Clin Pharmacol Ther. 2000;68:375‐383. [DOI] [PubMed] [Google Scholar]
  • 57. Chainuvati S, Nafziger AN, Leeder JS, et al. Combined phenotypic assessment of cytochrome p450 1A2, 2C9, 2C19, 2D6, and 3A, N‐acetyltransferase‐2, and xanthine oxidase activities with the "Cooperstown 5+1 cocktail". Clin Pharmacol Ther. 2003;74:437‐447. [DOI] [PubMed] [Google Scholar]
  • 58. Blakey GE, Lockton JA, Perrett J, et al. Pharmacokinetic and pharmacodynamic assessment of a five‐probe metabolic cocktail for CYPs 1A2, 3A4, 2C9, 2D6 and 2E1. Br J Clin Pharmacol. 2004;57:162‐169. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59. Ryu JY, Song IS, Sunwoo YE, et al. Development of the "Inje cocktail" for high‐throughput evaluation of five human cytochrome P450 isoforms in vivo. Clin Pharmacol Ther. 2007;82:531‐540. [DOI] [PubMed] [Google Scholar]
  • 60. Turpault S, Brian W, van Horn R, et al. Pharmacokinetic assessment of a five‐probe cocktail for CYPs 1A2, 2C9, 2C19, 2D6 and 3A. Br J Clin Pharmacol. 2009;68:928‐935. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61. Donzelli M, Derungs A, Serratore MG, et al. The Basel cocktail for simultaneous phenotyping of human cytochrome P450 isoforms in plasma, saliva and dried blood spots. Clin Pharmacokinet. 2014;53:271‐282. [DOI] [PubMed] [Google Scholar]
  • 62. Bosilkovska M, Clement M, Dayer P, Desmeules J, Daali Y. Incorporation of flurbiprofen in a 4‐drug cytochrome p450 phenotyping cocktail. Basic Clin Pharmacol Toxicol. 2014;115:465‐466. [DOI] [PubMed] [Google Scholar]
  • 63. Bosilkovska M, Samer C, Déglon J, et al. Evaluation of mutual drug‐drug interaction within Geneva cocktail for cytochrome P450 phenotyping using innovative dried blood sampling method. Basic Clin Pharmacol Toxicol. 2016;119:284‐290. [DOI] [PubMed] [Google Scholar]
  • 64. Miura M, Uchida S, Tanaka S, et al. Verification of a cocktail approach for quantitative drug‐drug interaction assessment: a comparative analysis between the results of a single drug and a cocktail drug. Xenobiotica. 2021;51:404‐412. [DOI] [PubMed] [Google Scholar]
  • 65. Prueksaritanont T, Tatosian DA, Chu X, et al. Validation of a microdose probe drug cocktail for clinical drug interaction assessments for drug transporters and CYP3A. Clin Pharmacol Ther. 2017;101:519‐530. [DOI] [PubMed] [Google Scholar]
  • 66. Stopfer P, Giessmann T, Hohl K, et al. Pharmacokinetic evaluation of a drug transporter cocktail consisting of digoxin, furosemide, metformin, and Rosuvastatin. Clin Pharmacol Ther. 2016;100:259‐267. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67. Stopfer P, Giessmann T, Hohl K, et al. Optimization of a drug transporter probe cocktail: potential screening tool for transporter‐mediated drug‐drug interactions. Br J Clin Pharmacol. 2018;84:1941‐1949. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68. Wiebe ST, Giessmann T, Hohl K, et al. Validation of a drug transporter probe cocktail using the prototypical inhibitors rifampin, probenecid, verapamil, and cimetidine. Clin Pharmacokinet. 2020;59:1627‐1639. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69. Trueck C, Hsin CH, Scherf‐Clavel O, et al. A clinical drug‐drug interaction study assessing a novel drug transporter phenotyping cocktail with Adefovir, Sitagliptin, metformin, Pitavastatin, and digoxin. Clin Pharmacol Ther. 2019;106:1398‐1407. [DOI] [PubMed] [Google Scholar]
  • 70. Machavaram KK, Almond LM, Rostami‐Hodjegan A, et al. A physiologically based pharmacokinetic modeling approach to predict disease‐drug interactions: suppression of CYP3A by IL‐6. Clin Pharmacol Ther. 2013;94:260‐268. [DOI] [PubMed] [Google Scholar]
  • 71. Schmitt C, Kuhn B, Zhang X, Kivitz AJ, Grange S. Disease‐drug‐drug interaction involving tocilizumab and simvastatin in patients with rheumatoid arthritis. Clin Pharmacol Ther. 2011;89:735‐740. [DOI] [PubMed] [Google Scholar]
  • 72. Chen YL, Vraux VL, Leneveu A, et al. Acute‐phase response, interleukin‐6, and alteration of cyclosporine pharmacokinetics. Clin Pharmacol Ther. 1994;55:649‐660. [DOI] [PubMed] [Google Scholar]
  • 73. Machavaram KK, Endo‐Tsukude C, Terao K, et al. Simulating the impact of elevated levels of Interleukin‐6 on the pharmacokinetics of various CYP450 substrates in patients with Neuromyelitis Optica or Neuromyelitis Optica Spectrum disorders in different ethnic populations. AAPS J. 2019;21:42. [DOI] [PubMed] [Google Scholar]
  • 74. Jiang X, Zhuang Y, Xu Z, Wang W, Zhou H. Development of a physiologically based pharmacokinetic model to predict disease‐mediated therapeutic protein‐drug interactions: modulation of multiple cytochrome P450 enzymes by Interleukin‐6. AAPS J. 2016;18:767‐776. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75. Xu Y, Hijazi Y, Wolf A, Wu B, Sun YN, Zhu M. Physiologically based pharmacokinetic model to assess the influence of Blinatumomab‐mediated cytokine elevations on cytochrome P450 enzyme activity. CPT Pharmacometrics Syst Pharmacol. 2015;4:507‐515. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76. Zhuang Y, de Vries DE, Xu Z, et al. Evaluation of disease‐mediated therapeutic protein‐drug interactions between an anti‐interleukin‐6 monoclonal antibody (sirukumab) and cytochrome P450 activities in a phase 1 study in patients with rheumatoid arthritis using a cocktail approach. J Clin Pharmacol. 2015;55:1386‐1394. [DOI] [PubMed] [Google Scholar]
  • 77. Okuno H, Takasu M, Kano H, Seki T, Shiozaki Y, Inoue K. Depression of drug‐metabolizing activity in the human liver by interferon‐beta. Hepatology. 1993;17:65‐69. [PubMed] [Google Scholar]
  • 78. Hellman K, Roos E, Österlund A, et al. Interferon‐beta treatment in patients with multiple sclerosis does not alter CYP2C19 or CYP2D6 activity. Br J Clin Pharmacol. 2003;56:337‐340. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79. Brennan MJ. The clinical implications of cytochrome p450 interactions with opioids and strategies for pain management. J Pain Symptom Manag. 2012;44:S15‐S22. [DOI] [PubMed] [Google Scholar]
  • 80. Khalilieh S, Hussain A, Montgomery D, et al. Effect of tildrakizumab (MK‐3222), a high affinity, selective anti‐IL23p19 monoclonal antibody, on cytochrome P450 metabolism in subjects with moderate to severe psoriasis. Br J Clin Pharmacol. 2018;84:2292‐2302. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81. Zhu Y, Xu Y, Zhuang Y, et al. Evaluating potential disease‐mediated protein‐drug interactions in patients with moderate‐to‐severe plaque psoriasis receiving subcutaneous Guselkumab. Clin Transl Sci. 2020;13:1217‐1226. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82. Khatri A, Cheng L, Camez A, Ignatenko S, Pang Y, Othman AA. Lack of effect of 12‐week treatment with Risankizumab on the pharmacokinetics of cytochrome P450 probe substrates in patients with moderate to severe chronic plaque psoriasis. Clin Pharmacokinet. 2019;58:805‐814. [DOI] [PubMed] [Google Scholar]
  • 83. Kwon M, Jeon JH, Choi MK, Song IS. The development and validation of a novel "dual cocktail" probe for cytochrome P450s and transporter functions to evaluate pharmacokinetic drug‐drug and herb‐drug interactions. Pharmaceutics. 2020;12:938‐963. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84. Bosilkovska M, Magliocco G, Desmeules J, Samer C, Daali Y. Interaction between fexofenadine and CYP phenotyping probe drugs in Geneva cocktail. J Pers Med. 2019;9:45‐52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85. Adedoyin A, Frye RF, Mauro K, Branch RA. Chloroquine modulation of specific metabolizing enzymes activities: investigation with selective five drug cocktail. Br J Clin Pharmacol. 1998;46:215‐219. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86. Zhang X, Lalezari JP, Badley AD, et al. Assessment of drug‐drug interaction potential of enfuvirtide in human immunodeficiency virus type 1‐infected patients. Clin Pharmacol Ther. 2004;75:558‐568. [DOI] [PubMed] [Google Scholar]
  • 87. Tran JQ, Othman AA, Wolstencroft P, Elkins J. Therapeutic protein‐drug interaction assessment for daclizumab high‐yield process in patients with multiple sclerosis using a cocktail approach. Br J Clin Pharmacol. 2016;82:160‐167. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88. Goh BC, Reddy NJ, Dandamudi UB, et al. An evaluation of the drug interaction potential of pazopanib, an oral vascular endothelial growth factor receptor tyrosine kinase inhibitor, using a modified Cooperstown 5+1 cocktail in patients with advanced solid tumors. Clin Pharmacol Ther. 2010;88:652‐659. [DOI] [PubMed] [Google Scholar]
  • 89. Derks M, Fowler S, Kuhlmann O. In vitro and in vivo assessment of the effect of dalcetrapib on a panel of CYP substrates. Curr Med Res Opin. 2009;25:891‐902. [DOI] [PubMed] [Google Scholar]
  • 90. Säll C, Alifrangis L, Dahl K, Friedrichsen MH, Nygård SB, Kristensen K. In vitro CYP450 enzyme down‐regulation by GLP‐1/glucagon co‐agonist does not translate to observed drug‐drug interactions in the clinic. Drug Metab Dispos. 2022;50:1087‐1097. [DOI] [PubMed] [Google Scholar]
  • 91. Ermer J, Corcoran M, Martin P. Lisdexamfetamine Dimesylate effects on the pharmacokinetics of cytochrome P450 substrates in healthy adults in an open‐label, randomized. Crossover Study Drugs R D. 2015;15:175‐185. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92. Ma JD, Nafziger AN, Villano SA, Gaedigk A, Bertino JS Jr. Maribavir pharmacokinetics and the effects of multiple‐dose maribavir on cytochrome P450 (CYP) 1A2, CYP 2C9, CYP 2C19, CYP 2D6, CYP 3A, N‐acetyltransferase‐2, and xanthine oxidase activities in healthy adults. Antimicrob Agents Chemother. 2006;50:1130‐1135. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93. Johnson BM, Song IH, Adkison KK, et al. Evaluation of the drug interaction potential of aplaviroc, a novel human immunodeficiency virus entry inhibitor, using a modified Cooperstown 5 + 1 cocktail. J Clin Pharmacol. 2006;46:577‐587. [DOI] [PubMed] [Google Scholar]
  • 94. Inui N, Akamatsu T, Uchida S, et al. Chronological effects of rifampicin discontinuation on cytochrome P450 activity in healthy Japanese volunteers, using the cocktail method. Clin Pharmacol Ther. 2013;94:702‐708. [DOI] [PubMed] [Google Scholar]
  • 95. Williams D, Tao X, Zhu L, et al. Use of a cocktail probe to assess potential drug interactions with cytochrome P450 after administration of belatacept, a costimulatory immunomodulator. Br J Clin Pharmacol. 2017;83:370‐380. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96. Treijtel N, Collins C, Bruijnsvoort M, et al. A cocktail interaction study evaluating the drug‐drug interaction potential of the perpetrator drug ASP8477 at multiple ascending dose levels. Clin Pharmacol Drug Dev. 2019;8:529‐540. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97. Vassiliou D, Sardh E, Harper P, et al. A drug‐drug interaction study evaluating the effect of Givosiran, a small interfering ribonucleic acid, on cytochrome P450 activity in the liver. Clin Pharmacol Ther. 2021;110:1250‐1260. [DOI] [PubMed] [Google Scholar]
  • 98. Rowland A, van Dyk M, Warncken D, Mangoni AA, Sorich MJ, Rowland A. Evaluation of modafinil as a perpetrator of metabolic drug‐drug interactions using a model informed cocktail reaction phenotyping trial protocol. Br J Clin Pharmacol. 2018;84:501‐509. [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.

Supplementary Materials

Table S1


Articles from Clinical and Translational Science are provided here courtesy of Wiley

RESOURCES