Download as pdf or txt
Download as pdf or txt
You are on page 1of 27

Clin Pharmacokinet (2018) 57:1267–1293

https://doi.org/10.1007/s40262-018-0650-9

SYSTEMATIC REVIEW

Complex Drug–Drug–Gene–Disease Interactions Involving


Cytochromes P450: Systematic Review of Published Case Reports
and Clinical Perspectives
Flavia Storelli1,2 • Caroline Samer1,3,4 • Jean-Luc Reny3,5 • Jules Desmeules1,2,3,4 •

Youssef Daali1,2,3,4

Published online: 17 April 2018


Ó Springer International Publishing AG, part of Springer Nature 2018

Abstract Drug pharmacokinetics (PK) is influenced by renal impairment, cirrhosis, and/or inflammation. Current
multiple intrinsic and extrinsic factors, among which con- knowledge on the impact of each of these factors on drug
comitant medications are responsible for drug–drug inter- exposure and DDIs is summarized and future perspectives
actions (DDIs) that may have a clinical relevance, resulting for the management of such complex DDIs in clinical
in adverse drug reactions or reduced efficacy. The addition practice are discussed, including the use of advanced
of intrinsic factors affecting cytochromes P450 (CYPs) Computerized Physician Order Entry (CPOE) systems, the
activity and/or expression, such as genetic polymorphisms development of model-based dose optimization strategies,
and diseases, may potentiate the impact and clinical rele- and the education of healthcare professionals with respect
vance of DDIs. In addition, greater variability in drug to personalized medicine.
levels and exposures has been observed when such intrinsic
factors are present in addition to concomitant medications
perpetrating DDIs. This variability results in poor pre-
Key Points
dictability of DDIs and potentially dramatic clinical con-
sequences. The present review illustrates the issue of
Complex drug–drug interaction (DDI) scenarios
complex DDIs using systematically searched published
involving intrinsic factors are becoming increasingly
case reports of DDIs involving genetic polymorphisms,
frequent.
Genetic polymorphisms and diseases such as renal
Electronic supplementary material The online version of this impairment, cirrhosis, and inflammation affect drug
article (https://doi.org/10.1007/s40262-018-0650-9) contains supple- disposition and DDIs, resulting in increased
mentary material, which is available to authorized users.
variability and poor predictability of DDIs.
& Youssef Daali Further scientific research is needed to integrate
youssef.daali@hcuge.ch
intrinsic factors in the prediction of DDIs, together
1
Division of Clinical Pharmacology and Toxicology, Geneva with the development of advanced integrated
University Hospitals, University of Geneva, Geneva, e-patient medical dossiers and education of
Switzerland healthcare professionals with respect to personalized
2
Geneva-Lausanne School of Pharmacy, University of medicine.
Geneva, Geneva, Switzerland
3
Faculty of Medicine, University of Geneva, Geneva,
Switzerland
4
Swiss Center for Applied Human Toxicology, Geneva,
Switzerland
5
Department of Internal Medicine, Rehabilitation and
Geriatrics, Geneva University Hospitals, University of
Geneva, Geneva, Switzerland
1268 F. Storelli et al.

1 Introduction association of multiple perpetrators, and patient-related


complexities such as involvement of genetic variants and
With the increase of life expectancy and population aging, diseases affecting drug disposition.
patients tend to have multiple chronic diseases, such as Interaction trials are often performed on early-stage
diabetes, hypertension, hypercholesterolemia, and depres- clinical development with healthy volunteers or preselected
sion. Polymorbidity in elderly patients often involves the patients. Nevertheless, genetic variants of CYP isoforms of
prescription of several concomitant drug treatments, clinical interest are not rare and ‘real life’ patients often
increasing the risk of potentially harmful drug–drug inter- have comorbidities that may affect drug disposition, such
actions (DDIs), which are a major cause of adverse drug as renal failure, liver dysfunction, or inflammation. While
reactions (ADRs), frequently leading to hospitalization each individual factor may not have a major impact on the
[1–4]. Indeed, the risk of DDIs increases with the number PK and PD of drugs, the consequence of their combined
of concomitant prescriptions: from 13% with two con- effect might be clinically significant and deleterious.
comitant drugs, to 38% with four concomitant durgs and This review aims to focus on DDIs involving patient-
82% with eight or more concomitant drugs [5]. To note, it related complexities, namely genetic polymorphisms and
was reported that the frequency of elderly patients taking diseases affecting drug disposition from a clinical per-
eight or more concomitant medications was [ 10% [6]. spective. First, published case reports of DDIs complicated
The nature of DDIs can be pharmacokinetic (PK, affecting by genetic variants, renal failure, cirrhosis, or inflammation
the disposition of the drug) and/or pharmacodynamic (PD, are described, and the effect of genetic polymorphisms and
affecting the effect of the drug without affecting its con- diseases on drug disposition and their implication on DDIs
centration). Regarding the latter, a drug can affect the are then discussed. Finally, clinical perspectives regarding
absorption, distribution, metabolism and elimination of the management of such complex DDI scenarios are
another drug. Of particular interest, cytochromes P450 presented.
(CYPs) are involved in the metabolism of a large number
of drugs, in particular those belonging to families 1, 2 and
3. Their importance in clinical practice and drug develop- 2 Case Reports Review
ment lies in the large interindividual variability of both
their expression and activity, which might be explained by 2.1 Methods
intrinsic factors (age, disease, sex, weight, and genetics)
and/or extrinsic factors (xenobiotics). Case reports involving CYP-related DDIs and genetic
In the last decades, CYP-related DDIs have been polymorphisms, cirrhosis, renal failure, or inflammation
extensively studied. Currently, clinically relevant DDIs can were searched using the PubMed database and reviewed
be detected by both the prescribers and the delivering systematically. The PubMed database search algorithm is
pharmacists through interaction alert software such as described in the electronic supplementary material and
Lexi-Interact [7], ePocrates [8], and Theriaque [9], thus contains terms related to CYPs (families 1, 2 and 3), drug
allowing for the identification, management and prevention interactions, and case reports. Language was restricted to
of DDI-related ADRs and therapeutic inefficiency. Because English and French, but there were no restrictions on the
the association of two interacting drugs cannot always be year of publication. Search results were first screened based
avoided, increasing efforts have been provided to develop a on title and abstract in order to rule out off-topic publica-
mechanistic model to quantitatively predict DDIs, allowing tions, and full-texts of selected publications were then
for prospective dose adjustment [10]. Such models include reviewed in order to verify if they fulfilled all the following
basic static, mechanistic static, and dynamic physiologi- eligibility criteria: coherent PK interaction involving CYP,
cally-based PK (PBPK) models. However, complex DDI information on CYP genotype, renal function, cirrhotic
scenarios are becoming increasingly frequent, partly status, or inflammatory state. Renal function and inflam-
because our scientific knowledge is constantly developing matory state had to be assessed by biomarkers, such as
and partly because of patients’ increasingly complex clin- serum creatinine/glomerular filtration rate (GFR) and
ical features (aging and polymorbidity) and treatments, and C-reactive protein (CRP)/interleukin (IL)-6/white blood
still remain difficult to predict. These complex DDI sce- cell count, respectively.
narios may include drug-related complexities such as
simultaneous involvement of multiple enzymes and trans- 2.2 Results
porters, simultaneous inhibition and induction, mechanisms
of autoinhibition/induction, time-dependent inhibition, The systematic search workflow is presented in Fig. 1.
inhibition/induction caused by major metabolites, Overall, 877 records were output from the PubMed search
performed on 13 July 2017. Of these, 769 were excluded
Complex Drug–Drug–Gene–Disease Interactions 1269

Fig. 1 Systematic search in


accordance with the PRISMA
workflow. PRISMA Preferred
Reporting Items for Systematic
Reviews and Meta-Analyses,
DDGI drug–drug–gene
interaction, DDDI drug–drug–
disease interaction, DDGDI
drug–drug–gene–disease
interaction

based on the title and abstract because they were off-topic. substrates were simvastatin, tacrolimus, colchicine and
Of the 108 selected records, 46 were excluded because they voriconazole, while the most cited CYP perpetrators were
did not fulfill the eligibility criteria described in the the inhibitors clarithromycin, ritonavir, and diltiazem (see
Methods section. Tables 4 and 5 for a complete list of victim and perpetrator
Eligible publications were dated from 1998 to 2017. drugs). There were only a few induction-related DDIs,
Among the 62 eligible publications, only one was in which were perpetrated by the CYP inducers carbamaza-
French, while all others were in English. Language pine and rifampin.
restrictions were not considered to have a deleterious effect The mechanisms of these complex DDIs are numerous:
on the quality of the present review, with its aim being (1) enhancement of the magnitude of interaction due to a
illustrative rather than quantitative. Moreover, it has not genetic variant directly impacting the CYP isoform of
been proven that language restriction has any notable im- interest [12, 13]; (2) increased vulnerability to phenocon-
pact on the results of meta-analyses [11]. version (i.e. the phenomenon that occurs when a perpe-
The 62 eligible publications presented 34 cases of drug– trator drug of DDIs is strong enough to modify the
drug–gene interactions (DDGIs), 25 cases of drug–drug– phenotype of an individual [14, 15]) caused by a genetic
disease interactions (DDDIs), and 4 cases of drug–drug– variant or disease directly affecting the inhibited/induced
gene–disease interactions (DDGDIs), which are summa- metabolic pathway [16–62]; (3) increased exposure of the
rized in Tables 1, 2 and 3, respectively. perpetrator drug due to genetic polymorphisms
DDIs found in the selected literature included 33 victim [21, 25, 26, 60, 63–68]; and (4) modification of the relative
drugs and 44 perpetrator drugs. The most cited CYP contribution of a minor pathway by a genetic variant or
Table 1 Reported case reports of complex drug–drug interactions involving genetic polymorphisms
1270

Interacting drugsa Patient characteristics Effect of interaction Comment Reference


Victim Perpetrator Age, Sex Relevant genotype Other relevant
years information

Atorvastatin 20 mg/day Pantoprazole 75 M CYP2C19*2/*2 SLCO1B1 521CC C: Rhabdomyolysis and acute renal [66]
20 mg/day ABCB1 3435TT failure 3 weeks after statin initiation
Clomipramine Citalopram 28 F CYP2D6*1/*4 PK: Css desmethylclomipramine : CYP2D6 phenotype: [16]
75–100 mg/day 40 mg/day 82% (although clomipramine daily PM
Thioridazine dose was reduced from 100 to
5 mg/day 75 mg), plasma level 8-hydroxy-
desmethylclomipramine ; 42%
4 days after the addition of
citalopram
Clomipramine Modafinil 60 F CYP2D6 *4A/*4A PK: Css clomipramine and [69]
75–100 mg/day 200–400 mg/day desmethylclomipramine : 22% and
14%, respectively, after the addition
of modafinil 200 mg/day (although
clomipramine daily dose was
reduced from 100 to 75 mg), and
63% and 116%, respectively, when
modafinil daily dose was : to
400 mg/day
C: : of liver function tests
Clozapine 750 mg Fluvoxamine 31 M CYP1A2*1F/*1F (UM) CYP1A2 high PK: Clozapine and norclozapine Caffeine MR [13]
3 9/day 25–50 mg/day activity (caffeine concentrations : 2.18-fold and 1.44- decreased 50%
MR 17.9) fold, respectively, within 1 week of with fluvoxamine
fluvoxamine 25 mg/day 50 mg/day
C: Marked improvement in psychosis
with fluvoxamine 50 mg/day
Codeine 10–20 mg/4 h Fluoxetine NR F CYP2D6 *2/*4 UGT2B7 *1/*2 PK: Total codeine 840 lg/L, free Patient was involved [30]
Bupropion codeine 348 lg/L, morphine-3- in a car accident
glucuronide 17 lg/L, morphine-6-
Promethazine
glucuronide 3 lg/L and free
Valproic acid morphine not detectable
Dextromethorphan Amitriptyline 60 M CYP2D6*4/*4 PK: Accumulation of [62]
30 mg 3 9/day 50 mg/day dextromethorphan,
reaching [ 100 ng/mL after 3 days
C: Somnolence after 3 days of
dextromethorphan
Efavirenz 600 mg/day Fluconazole 33 F CYP2B6: 516TT PK: Efavirenz levels: 59.4 mg/L [71]
400 mg/day (approximately 30-fold more than
Ritonavir 100 mg the normal limit)
2 9/day C: Severe psychosis
F. Storelli et al.
Table 1 continued
Interacting drugsa Patient characteristics Effect of interaction Comment Reference
Victim Perpetrator Age, Sex Relevant genotype Other relevant
years information

Fluvastatin 40 mg/day Telmisartan 39 M CYP2C9*1/*3 ABCC2 -24CT C: Twofold : of creatine kinase levels [32]
20 mg/day with myalgia, cramps and
fasciculation in the legs
Fluticasone 27.5 lg/day per Ritonavir 6 F CYP3A4 *1B/*1G C: Cushing syndrome within a few [33]
nostril 80 mg/day CYP3A5 *3/*3 weeks after starting fluticasone
Haloperidol 9 mg/day 59 M CYP2D6*5/*10 C: Neuroleptic malignant syndrome [18]
Levomepromazine 100 mg/day
Chlorpromazine 250 mg/day
Hydrocodone 30 mg over Clarithromycin 5 M CYP2D6*2A/*41 PK: Hydrocodone fatal concentration The child received [20]
Complex Drug–Drug–Gene–Disease Interactions

24 h Valproic acid 0.14 lg/mL twice the


250 mg 2 9/day Hydromorphone \ 0.008 lg/mL recommended dose
Valproic acid 43 lg/mL (within the
therapeutic range)
C: Death
Lopinavir 180 mg 2 9/day Ritonavir 45 mg 6/12 F CYP2D6 UM PK: Plasma level of ritonavir and Ritonavir dose was [65]
2 9/day lopinavir not detectable increased threefold
C: Treatment failure up to 150 mg
2 9/day, and
lopinavir dose was
kept at 75 mg
2 9/day, resulting
in a
detectable plasma
level and
therapeutic
efficacy
Metoprolol 200 mg/day Terbinafine 63 M CYP2D6 *1/*4 C: Sinus bradycardia, confusion, falls [31]
250 mg/day
Metoprolol 200 mg/day Propafenone 66 F CYP2D6*4/*9 PK: MR metoprolol/a- [27]
600 mg/day hydroxymetoprolol 104.3 (PM
phenotype) 3 h after metoprolol
intake
After propafenone withdrawal: 1.4
(phenotype EM)
C: Tiredness and dyspnea on exertion
Nortriptyline 75 mg/day Fluoxetine 24 F CYP2D6*1/*81 VKORC1*3/*3 PK: C12h 1830 nmol/L (therapeutic [28]
60 mg/day (truncated protein) range 200–600 nmol/L)
CYP2C19*1/*2D C: Minimal adverse effects apart from
1271

dry mouth
Table 1 continued
1272

Interacting drugsa Patient characteristics Effect of interaction Comment Reference


Victim Perpetrator Age, Sex Relevant genotype Other relevant
years information

Oxycodone 40–60 mg/day Rifampin 300 mg 60 M CYP2D6*1/*4 CYP3A5*3/*3 PK: Oxycodone not detected; Patient was [72]
oxymorphone not detected free, suspected of lack
190 lg/L conjugated; noroxycodone of compliance
700 ug/mL free because of
C: Not adequate pain relief negative
oxycodone testings
Risperidone 6 mg/day Carbamazepine 50 M CYP2D6*4/*5 PK: Plasma levels of risperidone and [70]
titrated up to active metabolite ; 3.67- and 2.73-
800 mg/day over fold, respectively, after 4 weeks of
5 days carbamazepine
C: Exacerbation of psychotic
symptoms (auditory hallucinations,
paranoid delusions, ideas of
reference, and mild excitement
Risperidone 4 mg/day Haloperidol 6 mg 17 M CYP2D6*4/*4 PK: 8 days after haloperidol initiation, [17]
2 9/day risperidone Css : from 29–37 to 59
ug/L, 9-hydroxyrisperidone Css :
from 12–13 to 18 lg/L, and active
moiety Css : from 41–50 to 77 lg/L
C: Severe akathisia
Simvastatin 20 mg/day Cyclosporin 55 F CYP3A5*3/*3 C: Rhabdomylosis and elevation of [24]
350–500 mg/day CYP3AP1*3/*3 liver function tests
according to TDM
Simvastatin 40 mg/day for Diltiazem 90 mg 63 F CYP3A5 *3/*3 Other genetic C: : of creatine kinase and alanine [22]
4 weeks, then 80 mg/day 2 9/day variants: aminotransferase levels
SLCO1B1 521T/
C and ABCB1
2677TT
Tacrolimus 0.06 mg/kg/day Itraconazole 51 F CYP3A5*3/*3 PK: Tacrolimus C0 : 2.8-fold 2 days [19]
200 mg/day after introduction of itraconazole
Tacrolimus 1 mg/day Itraconazole 53 F CYP2C19*1/*2 PK: : in tacrolimus blood Voriconazole levels [25]
100 mg/day CYP3A5*3/*3 concentration after antifungal switch 5.5 lg/mL
replaced by from 6.1 to 34.2 ng/mL (reference
voriconazole 400 1.4–1.8 lg/mL)
mg/day
F. Storelli et al.
Table 1 continued
Interacting drugsa Patient characteristics Effect of interaction Comment Reference
Victim Perpetrator Age, Sex Relevant genotype Other relevant
years information

Tacrolimus 9 mg 2 9/day, Omeprazole started 17 NR CYP2C19*2/*2 ABCB1 1236TT, PK: C0 with a 9 mg 2 9/day regimen: [26]
days 1–4, then 7 mg on day 5 CYP3A5*3/*3 677TT and 19.6 ng/ml at day 3 and 29.2 ng/ml
2 9/day from day 5 3435TT at day 4
C0 with a 7 mg 2 9/day regimen:
92 ng/mL day 6
C: Acute nephrotoxicity (serum
creatinine 160 lmol/L associated
with proteinuria)
Tacrolimus 17 mg/day Lansoprazole 34 M CYP2C19*1/*2 PK: Tacrolimus C0 : 30% 1 day after [63]
Complex Drug–Drug–Gene–Disease Interactions

30 mg/day lansoprazole introduction, and


continued to increase although the
tacrolimus dose was gradually
decreased
Tacrolimus 22 mg/day Lansoprazole 57 F CYP2C19*1/*2 PK: Tacrolimus C0 : 1.57-fold 3 days [67, 68]
30 mg/day after lansoprazole initiation
Tacrolimus 2 mg/day Lansoprazole 51 M CYP2C19*2/*3 PK: Tacrolimus C/D ratio : Interaction occurred [64]
30 mg/day CYP3A5*3/*3 approximately twofold after switching
from omeprazole
to lansoprazole
Tramadol 4.5 g (intentional Ketoconazole 22 F CYP2D6*1/*1X2 PK: Tramadol initial concentration Tramadol/O- [73]
overdose) (plasma 3.1 mg/L at admission (low value desmethyltramadol
concentration at considering the dose intake) MR 2.54
admission 200 ng/ Tramadol/N-
mL) desmethyltramadol
MR 11.4
Venlafaxine 112.5 mg/day Cotrimoxazole 46 F CYP2C19*1/*2 CYP2D6 and PK: Css,plasma venlafaxine ? O- Tremor is probably [74]
800 ? 160 mg/day CYP3A4 desmethylvenlafaxine : from 344 to the consequence of
phenotypes: EM 444 ng/mL with the introduction of both PK and PD
cotrimoxazole (therapeutic interaction (known
recommended range: 100–400 ng/ side effect of
mL) cotrimoxazole)
N-desmethylvenlafaxine was not
modified: Css 8 ng/mL (expected
17–37 ng/mL)
C: Severe bilateral resting tremor of
the hands
1273
Table 1 continued
1274

Interacting drugsa Patient characteristics Effect of interaction Comment Reference


Victim Perpetrator Age, Sex Relevant genotype Other relevant
years information

Voriconazole 400 mg day Carbamazepine 62 F CYP2C19*17/*17 PK: C0 \ 0.3 mg/L at day 5 and [23]
1, then 200 mg 2 9/day, 200 mg 2 9/day 0.5 mg/L at day 9 (therapeutic range
day 2.5, then 510 mg IV 1–5 mg/L)
loading dose followed by C: Therapeutic failure
425 mg 2x/day
Voriconazole 400 mg Esomeprazole 43 F CYP2C19*1/*2 PK: 7.8 mg/L (therapeutic range [21]
2x/day day 1, followed by 1–5 mg/L; target 1.5 mg/L)
200 mg 2 9/day C: Hallucinations 7 days after
voriconazole initiation, : of liver
function tests
Voriconazole 2.5–5.5 mg/ Esomeprazole 35 F CYP2C19*1/*17 CYP2C19 PK: Subtherapeutic levels of [12]
kg/12 h IV 40 mg 2 9/day IV phenotype: voriconazole (B 0.5 ug/L), : up to
Ritonavir omeprazole 3.5 lg/L with concomitant
100 mg/day metabolic ratio administration of esomeprazole and
10.5 (CYP2C19 ritonavir (although voriconazole
activity dose reduced from 5.5 to 2.5 mg/kg/
decreased) 12 h)
Voriconazole 450 mg day Haloperidol 1 mg 43 M CYP2C19*2/*2 PK: Voriconazole C0 : from 3931 ng/ [75]
1, followed by 3 9/day from day mL at day 6 to 11,063 ng/mL at day
300 mg/day days 2–12 8 18
and 250 mg/day days C: : of liver function tests
13–18
Voriconazole 200 mg Ritonavir 50 M CYP2C19 *1/*2 PK: Overtherapeutic level of [34]
2 9/day 100 mg/day voriconazole requiring 25% dose
decrease
Warfarin 5 mg/day Amiodarone 73 M CYP2C9*3/*3 PD: INR 10.7 with 5 mg daily dose, [29]
400 mg/day 6.8 with 2.5 mg daily dose
C/D concentration over dose ratio, C clinical outcomes, Css concentration at steady-state, EM extensive metabolizer, INR international normalized ratio, MR metabolic ratio, NR not reported,
PK pharmacokinetic outcomes, PD pharmacodynamic outcomes, PM poor metabolizer, UM ultrarapid metabolizer, M male, F female, CYP cytochrome P450, TDM therapeutic drug
monitoring, IV intravenously, C12h concentration at 12 h, C0 trough concentration, : indicates increase, ; indicates decrease
a
If the dosage is not specified, the data could not be found in the case report. If not specified, the administration route is orally
F. Storelli et al.
Complex Drug–Drug–Gene–Disease Interactions 1275

Table 2 Reported case reports of complex drug–drug interactions involving diseases affecting drug disposition
Interacting drugsa Patient characteristics Effect of interaction Comment Reference
Substrate Perpetrator Age, Sex Relevant disease
years

Budesonide Amiodarone 100 mg/day 81 M Chronic RI C: Cushing’s syndrome [36]


6 mg/day (serum
creatinine
196–207 lmol/
L)
Budesonide Ritonavir 100 mg/day 75 M Chronic kidney C: Cushing’s syndrome [50]
3 mg Atazanavir 300 mg/day disease (serum
3 9/day creatinine
1.6 mg/dL)
Colchicine Clarithromycin 500 mg 23 F RI (dialysis) C: Nausea, vomiting, [41]
0.5 mg 2 9/day Mediterranean weakness, abdominal pain,
2 9/day fever (CRP diarrhea, pancytopenia
8.05 mg/dL
Colchicine Clarithromycin 500 mg 61 M RI (serum C: Nausea, vomiting, [41]
0.5 mg 2 9/day creatinine diarrhea, worsening of
2 9/day 3.1 mg/dL) renal function (creatinine
clearance 10 mL/min)
Colchicine Clarithromycin 73 M Chronic RI C: Neuromyopathy [42]
0.5 mg/day
Colchicine Clarithromycin 500 mg 48 M Chronic RI C: Rhabdomyolysis [46]
0.6 mg/day 2 9/day
Colchicine Cyclosporin 175 mg/day 60 M Chronic RI PK: C36h,serum colchicine [44]
1 mg/day (dialysis) serum 7 ng/mL (reference
range 1–4 ng/mL)
Cyclosporin blood
concentration : from 130
to 475 ng/mL (reference
range 100–150 ng/mL)
C: Vomiting, diarrhea,
neutropenia,
rhabdomyolysis
Clozapine Valproic acid 500 mg/day 49 M Probable PK: C8h clozapine 1130 ng/ Non-smoker [55]
300 mg/day Escitalopram 10 mg/day inflammation dL, norclozapine 297 ng/ Past history of
(WBC 13,000 dL (ratio 3.81:1) chronic
Aripiprazole 15 mg/day
cells/mm3) C: Somnolence moderate
renal
insufficiency
Fentanyl Diltiazem 85 M RI (creatinine C: Hypoactive delirium [49]
25 lg/h IV 2.0 mg/dL)
drip
Fluticasone Fluconazole 100 mg/day 9 F Cystic fibrosis C: Cushing’s syndrome [52]
110 lg/ and cirrhosis
actuation
Glibenclamide Sorafenib 800 mg/day 70 M Cirrhosis PD: Severe hypoglycemia [54]
3.5 mg (Child–Pugh (blood glucose 37 mg/dL)
2 9/day A) C: Acute nocturnal
eGFR 55 mL/ disorientation and
min somnolence
Nifedipine Clarithromycin 500 mg 77 M Chronic RI C: Reduced consciousness [40]
60 mg 2 9/day with response to stimuli,
2 9/day hypotension (80/
40 mmHg), bradycardia
(40 bpm)
1276 F. Storelli et al.

Table 2 continued
Interacting drugsa Patient characteristics Effect of interaction Comment Reference
Substrate Perpetrator Age, Sex Relevant disease
years

Nifedipine, Clarithromycin 78 M Chronic kidney C: Hypotension (96/ Patient was [57]


diltiazem disease 38 mmHg), bradycardia also taking
(44 bpm), loss of carvedilol
consciousness
Repaglinide Cotrimoxazole 76 M RI (serum C: Symptomatic [47]
1 mg 800 ? 160 mg/day creatinine hypoglycemia (blood
3 9/day 1.84–2 mg/dL) glucose 34 mg/dL)
Simvastatin Amiodarone 1 mg/day 80 M Chronic RI C: Severe rhabdomyolysis, [51]
40 mg/day (serum elevation of liver function
creatinine tests 4 days after
2.2 mg/dL, amiodarone initiation
eGFR 31)
Simvastatin Azithromycin 500 mg/day and 73 M Chronic kidney C: Acute kidney injury and Comorbidity: [53]
80 mg/day 250 mg/day days 2–5 disease rhabdomyolysis obesity
(;CYP3A4
activity by
40%)
Simvastatin Clarithromycin 500 mg 64 M Severe RI C: Rhabdomyolysis [37]
80 mg/day 2 9/day
Simvastatin Colchicine 0.5 mg 2 9/day 70 M Chronic RI C: Rhabdomyolysis and : of [38]
aspartate aminotransferase
levels
Simvastatin Tacrolimus, sirolimus (dosage 56 M RI (serum C: Rhabdomyolysis and [45]
10 mg/day unknown) creatinine acute renal failure
2.33 mg/dL) resulting in patient’s death
Simvastatin Telaprevir 750 mg 3 9/day 46 M Hepatitis C: Rhabdomyolysis and : of [56]
80 mg/day C-related liver function tests
cirrhosis
Sirolimus Erythromycin 500 mg 2 9/day 70 M Chronic RI PK: Sirolimus C0 38.2 ng/ [39]
1 mg/day (hemodialysis) mL (target range 5-10 ng/
mL)
Sirolimus Fluconazole 100 mg/day IV for 60 M RI PK: Sirolimus C0 : greater [39]
2 mg/day 10 days, then 200 mg PO than fourfold (up to
48.44 ng/mL)
Sorafenib Felodipine 5 mg 2 9/day 80 M Cirrhosis PK: Sorafenib Css, plasma : [48]
400 mg (competition) (Child–Pugh threefold 15 days after
2 9/day A) felodipine introduction
Tacrolimus Diltiazem 5–10 mg/h 24 h 38 M RI (serum PK: Tacrolimus C0 : to [35]
8 mg infusion followed by 30 mg/ creatinine 55 ng/mL
2 9/day 8 h orally 2.1 mg/dL) C: Delirium, confusion and
Ciprofloxacin agitation
Venlafaxine 75 mg/day 55 F Cirrhosis C: Acute respiratory failure [43]
Propanolol 160 mg/day (Child–Pugh
C)
bpm beats per minute, C clinical outcome, C8h concentration every 8 h, C0 trough concentration, CRP C-reactive protein, Css concentration at
steady state, CYP cytochrome P450, eGFR estimated glomerular filtration rate, F female, IV intravenous, M male, PD pharmacodynamic
outcome, PK pharmacokinetic outcome, PO orally, RI renal insufficiency, WBC white blood cells, : indicates increase, ; indicates decrease
a
If the dosage is not specified, the data could not be found in the case report. If not specified, the administration route is orally
Complex Drug–Drug–Gene–Disease Interactions 1277

Table 3 Reported case reports of complex drug–drug interactions involving both genetic polymorphisms and any disease affecting drug
disposition
Interacting drugsa Patient characteristics Effect of interaction Comment Reference
Substrate Perpetrator Age, Sex Relevant Relevant
years disease genotype

Codeine 25 mg Clarithromycin 62 M RI CYP2D6 PK: High levels of Comorbidity: [58]


3 9/day Voriconazole UM morphine (80 ng/ chronic
Valproic acid mL) and lymphocytic
1500 mg/day morphine-6- leukemia
glucuronide
(136 ng/mL)
C: Respiratory
depression, coma
Dextromethorphan Metoprolol 64 M Chronic renal CYP2D6*1/ PK: High [59]
30 mg 40 mg/day failure *10 dextromethorphan
(peritoneal blood level
dialysis) (2.68 ng/mL) 60 h
after a 30 mg dose
C: Myoclonus
Escitalopram Esomeprazole 46 F Child–Pugh C CYP2C19*2/ PK: Day 3: Escitalopram level [60]
10 mg 2 9/day 40 mg/day cirrhosis *2 Escitalopram 3 days before
from day 1 CYP2D6*5*/ serum level ARV initiation
Darunavir 600 *10 619 nmol/L was in the
mg 2 9/day (: 12 9) therapeutic range
and ritonavir Day 8: Escitalopram (52 nmol/L)
100 mg serum level:
2 9/day from 695 nmol/L
day 4 (: 13 9)
Expected
therapeutic range
40–250 nmol/L
Estimated half-life
67–69 h
(literature:
27–33 h)
C: Serotonin
syndrome at day 8
Haloperidol 1 mg Ciprofloxacin 66 M Cirrhosis of CYP2D6*6/ C: Severe UGT2B7-161CT [61]
2 9/day alcoholic *6 extrapyramidal
origin symptoms
Acute
inflammation
(leukocytes
26.72 9 109/
L, CRP
108.48 mg/L)
ARV antiretrovirals, C clinical outcome, CRP C-reactive protein, F female, M male, PK pharmacokinetic outcome, RI renal insufficiency, UM
ultrarapid metabolizer, : indicates increase
a
If the dosage is not specified, the data could not be found in the case report. If not specified, the administration route is orally

disease affecting another major pathway negligible effects on CYP activity at therapeutic doses,
[58, 60, 61, 69–75]. such as colchicine, felodipine, pantoprazole, propanolol,
Interestingly, complex DDIs were perpetrated by drugs tacrolimus, azithromycin (CYP3A), amitriptyline, meto-
with CYP-inhibiting/inducing potencies ranging from weak prolol (CYP2D6), haloperidol (CYP3A), and aripiprazole
to strong. In some cases, drugs described as having null to (CYP2D6, CYP3A) might have been capable of inhibiting
1278 F. Storelli et al.

Table 4 Victim drugs Victim drug No. of occurrences CYP-related metabolic pathways References
implicated in the selected case
reports Atorvastatin 1 3A [76]
Budesonide 2 3A [77]
Chlorpromazine 1 1A2, 2D6 [78, 79]
Clomipramine 2 1A2, 2C19, 2D6, 3A [80]
Clozapine 2 1A2, 2D6, 3A [81]
Codeine 2 2D6, 3A [76]
Colchicine 5 3A [82]
Dextromethorphan 2 2D6, 3A [76]
Diltiazem 1 2D6, 3A [83–85]
Efavirenz 1 1A2, 2B6, 3A [86]
Escitalopram 1 2C19, 2D6, 3A [87]
Fentanyl 1 3A [76]
Fluticasone 2 3A [88]
Fluvastatin 1 2C9 [76]
Glibenclamide 1 2C9, 3A [89–91]
Haloperidol 2 1A2, 2D6, 3A [76]
Hydrocodone 1 2D6, 3A [92]
Lopinavir 1 3A [93]
Metoprolol 2 2D6 [76]
Nifedipine 2 3A [76]
Nortriptyline 1 1A2, 2C19, 2D6, 3A [94, 95]
Oxycodone 1 2D6, 3A [96]
Repaglinide 1 2C8, 3A [97]
Risperidone 2 2D6, 3A [76]
Simvastatin 7 3A [76]
Sirolimus 2 3A [76]
Sorafenib 1 3A [98]
Tacrolimus 7 3A [76]
Tramadol 1 2B6, 2D6, 3A [99]
Venlafaxine 2 2C19, 2D6, 3A [100]
Voriconazole 5 2C9, 2C19, 3A [101]
Warfarin 1 1A2, 2C8, 2C19, 2C9, 3A [102]
CYP cytochrome P450

CYP pathways due to either increased vulnerability to pathway would not be significant in most individuals, it
phenoconversion caused by genetic or disease particulari- might become clinically relevant when the major pathway
ties, or enhanced exposure related to genetic is non-functional.
polymorphisms.
Victim drugs were affected by CYP inhibitors and
inducers either through major or minor metabolic path- 3 Genetic Polymorphisms
ways. In the latter case, inhibition of a major metabolic
pathway by a genetic variant or disease allowed boosting of CYPs are subject to genetic polymorphisms, defined as
the minor pathway relative contribution to overall drug stable variations in a given locus of the genetic sequence,
metabolism (fm). The fm value has a high impact on the which are detected in 1% or more of a specific population
magnitude of DDIs. When a major elimination pathway is [129]. Among these CYPs, the most polymorphic genes are
inhibited, the contribution of the minor pathway is CYP2C9, CYP2C19 and CYP2D6; however, genetic vari-
enhanced; therefore, the magnitude of DDIs can be dra- ants of other enzymes, such as CYP1A2, CYP3A4,
matically increased when multiple elimination pathways CYP3A5, and CYP2B6, have also been described. Variant
are inhibited. Whereas the effect of inhibition of the minor alleles of CYP-coding genes may either lead to
Complex Drug–Drug–Gene–Disease Interactions 1279

Table 5 Perpetrator drugs implicated in the selected case reports


Perpetrator No. of occurrences Inhibited/induced pathways References

Amiodarone 3 2C9, 3A: strong inhibition [76, 103]


Amitriptyline 1 2D6: possible weak inhibition (competitive substrate) [76]
Aripiprazole 1 2D6, 3A: possible weak inhibition (competitive substrate) [104]
Atazanavir 1 3A: strong inhibition [105]
Azithromycin 1 3A: weak inhibition [106]
Bupropion 1 2D6: strong inhibition [76]
Carbamazepine 2 2C9, 3A: induction [76]
Chlorpromazine 1 2D6: moderate inhibition [107–109]
Ciprofloxacin 2 3A: moderate inhibition [110]
Citalopram 1 2D6: moderate inhibition [111]
Clarithromycin 8 3A: strong inhibition [76]
Colchicine 1 3A: possible weak inhibition (competitive substrate) [82]
Cotrimoxazole 2 2C8, 2C9: moderate inhibition [76]
Cyclosporin 2 3A: strong inhibition [112]
Darunavir 1 3A: strong inhibition [113]
Diltiazem 4 3A: moderate inhibition [76]
Erythromycin 1 3A: moderate inhibition [76]
Escitalopram 1 2D6: moderate inhibition [87]
Esomeprazole 3 2C19: strong inhibition [76]
Felodipine 1 3A: possible weak inhibition (competitive substrate) [76]
Fluconazole 3 2C9, 3A: strong inhibition [76]
Fluoxetine 2 2C19, 3A: moderate inhibition [76]
2D6: strong inhibition
Fluvoxamine 1 2C9, 3A: moderate inhibition [76, 114]
1A2, 2C19: strong inhibition
Haloperidol 3 CYP3A: weak inhibition [115, 116]
2D6: strong inhibition
Itraconazole 2 3A: strong inhibtion [76]
Ketoconazole 1 3A: strong inhibition [76]
Lansoprazole 3 2C19: strong inhibition [76, 117]
3A: possible weak inhibition (competitive substrate)
Levomepromazine 1 2D6: strong inhibition [118]
Metoprolol 1 2D6: weak inhibition (competitive substrate) [76]
Modafinil 1 2C19: moderate inhibition [119]
Omeprazole 1 2C19: moderate inhibition [76]
Pantoprazole 1 2C19: weak inhibition [76]
Promethazine 1 2D6: inhibition [120]
Propafenone 1 2D6: strong inhibition [27]
Propanolol 1 2D6: weak inhibition (competitive substrate) [121]
Rifampin 1 1A2, 2B6, 2C8, 2C9, 2C19, 3A: induction [76]
Ritonavir 7 3A: strong inhibition [76]
Sorafenib 1 3A: weak inhibition [98, 122]
Tacrolimus 1 3A: weak inhibition (competitive substrate) [76]
Telaprevir 1 3A: strong inhibition [123, 124]
Telmisartan 1 2C9: moderate inhibition [125]
Terbinafine 1 2D6: strong inhibition [76]
Thioridazine 1 2D6: strong inhibition [126]
Valproic acid 3 2C9, 3A: inhibition [127]
Voriconazole 1 3A: strong inhibition [128]
1280 F. Storelli et al.

significantly decreased or increased enzyme activity, or vulnerability to phenoconversion and increased exposure of
regulated gene expression leading to lack of or overex- the perpetrator was well-described in a 17-year-old trans-
pression of coded enzymes [130]. plant recipient, with overtherapeutic tacrolimus levels
The impact of CYP genetic variants on the PK of sub- becoming even higher and resulting in acute nephrotoxicity
strate drugs (such as CYP2C9 for vitamin K antagonists, after omeprazole initiation despite a dose reduction of the
CYP2C19 for clopidogrel, and CYP2D6 for opioid anal- immunosuppressant [26]. Genotyping revealed that the
gesics, tamoxifen, and antidepressants) have been exten- patient was a non-expressor of CYP3A5, the major enzyme
sively studied in the last decades. Genetic-based dosing involved in tacrolimus metabolism. In addition, the patient
guidelines constitute a useful tool for the management of was a poor metabolizer (PM) of CYP2C19 (CYP2C19*2/
gene–drug interactions (GDIs) in clinical practice. When a *2) and carried non-functional variants of ABCB1 coding
GDI cannot be avoided through modification of therapeutic for P-glycoprotein (P-gp). In this case, omeprazole likely
entity, dose adjustment or safety/efficacy monitoring might inhibited tacrolimus elimination through competition with
be proposed to manage such interactions and avoid thera- the CYP3A4 pathway (minor metabolic pathway of
peutic failure and/or ADRs. The clinical relevance of pre- omeprazole) due to genetic polymorphisms of both
emptive genotyping is supported by recent studies that CYP2C19 (major pathway of omeprazole elimination) and
estimate the proportion of the population carrying at least CYP3A5 (preferred metabolic pathway of tacrolimus). As
one actionable pharmacogenetic variant to be over 90% a last example of dramatic outcomes of DDGIs, lethal
[131–133]. The clinical benefit of pre-emptive genotyping levels of hydrocodone were found in a 5-year-old child
of the major genes involved in the PK and PD of drugs is who received twice the recommended dose of hydrocodone
currently assessed in several prospective implementation (30 mg over 24 h). In this case, the fatal outcome was a
studies [134–139]. A recent study by an American genetic consequence of overdose, reduced CYP2D6 activity
testing laboratory with more than 22,000 patients reported (CYP2D6 genotype *2A/*41), and concomitant adminis-
that among all patients with at least one drug interaction in tration of CYP3A inhibitors (clarithromycin and valproic
their medication list (69% of the cohort), 53% were DDIs, acid) [20].
25% were GDIs, and 22% were DDGIs [140]. Two scenarios can be considered when assessing the
The simultaneous presence of both GDIs and DDIs may impact of DDGIs: (1) introduction of a perpetrator drug in
be responsible for greater clinical relevance (efficacy/ a patient stabilized with a victim drug; and (2) addition of a
safety) of such interactions, as reported in the case reports victim drug in a patient treated by a perpetrator. In the first
described herein. In addition, the complexity of the latter case, one may consider the magnitude of DDIs relative to
type of drug interactions results in greater variability in genotype, while, in the second case, the point of view may
drug levels and difficulty of DDI predictions. The impact of focus on genotype-related vulnerability to phenoconversion
genetic polymorphisms on CYP-mediated DDIs has been and on the superposition of mechanisms of both DDIs and
highlighted in several studies in the past years. Bahar and GDIs.
colleagues recently published a comprehensive systematic In the first case, the dose of the victim drug is probably
review on DDGIs involving the most polymorphic already modified according to the genetic variant before
enzymes (CYP2C9, CYP2C19 and CYP2D6) that sum- introduction of the perpetrator, and the magnitude of the
marized the available clinical studies on this topic [141]. DDI will depend on the genotype-relative contribution of
The present review adds clinical illustrations of such drug the inhibited/induced CYP pathway. As previously dis-
interactions. cussed, the impact of the fm value of the inhibited clearance
Among the DDGI cases described in the present study, pathway to the extent of DDI is well known and the effect
some had severe or fatal outcomes. In a psychotic 50-year- of even slight variations in fm values can be clinically
old patient with inactive CYP2D6 (CYP2D6*4/*5), the important, especially when the fm value approaches 1
addition of carbamazepine to a stable risperidone regimen [142]. In the case of CYP inhibition, the higher the con-
resulted in a significant decrease of active moiety levels, tribution of the enzymatic pathway, the higher the magni-
leading to an exacerbation of psychotic symptoms [70]. tude of DDIs [108, 143–145]. A PM of a specific CYP
Severe psychosis related to toxic levels of efavirenz isoform will not be greatly affected by an inhibitor of the
resulted from the coadministration of efavirenz, ritonavir variant isoform of interest. In healthy CYP2D6 PM Cau-
and fluconazole in a 33-year old HIV-positive woman casian subjects, no increase in the AUC of metoprolol was
carrying the CYP2B6 516TT genetic variant. In this case, observed when coadministered with dronedarone, a mod-
the inhibition of CYP3A by fluconazole, which normally erate CYP2D6 inhibitor [146]. In CYP2D6 PM psychiatric
has a modest effect on efavirenz exposure, became sig- patients, the MR of debrisoquine, a historical phenotyping
nificant because of the altered CYP2B6 major pathway probe of CYP2D6, remained unchanged when increasing
[71]. A case of a DDGI explained by both increased the thioridazine dose [147]. In a clinical study with healthy
Complex Drug–Drug–Gene–Disease Interactions 1281

Japanese subjects, inhibition of aripiprazole metabolism by variants of CYP2C9 increased the risk of overanticoagu-
paroxetine was significantly greater in extensive metabo- lation in the presence of CYP2C9 inhibitors, from 52 to
lizers (EMs) than in intermediate metabolizers (IMs). 78% [151]. We recently performed a prospective clinical
There was no marked difference with fluvoxamine, a less trial that aimed at assessing the risk of phenoconversion in
potent inhibitor [143]. In Korean subjects, the frequent CYP2D6 genetically predicted EMs and found a higher
CYP2D6*10 allele, which leads to decreased activity of rate of phenoconversion to PMs with paroxetine in
CYP2D6, showed an impact on the extent of CYP inhibi- heterozygous carriers of a non-functional allele compared
tion by paroxetine, a strong time-dependent inhibitor [144]. with homozygous carriers of two functional alleles [152].
Another study highlighted the differential inhibition extent Therefore, the lower the CYP basal activity, the higher the
of haloperidol metabolism by chlorpromazine in Japanese incidence of phenoconversion and levels of drugs with
schizophrenic patients in relation to CYP2D6*5 and *10 concomitant administration of a CYP inhibitor. This might
alleles [108]. The interaction between fluconazole have clinical consequences when victim drugs have a
200–400 mg/day and flurbiprofen was studied in a clinical narrow therapeutic window.
study with healthy volunteers, which demonstrated that Complexity is even greater if the magnitude of a DDI
changes in flurbiprofen apparent oral clearance caused by and its clinical impact is modified when a genetic variant
fluconazole coadministration were gene dose-dependent, affects the concentrations of a perpetrator. This has been
with only modest change occurring in CYP2C9 PM sub- demonstrated with healthy volunteers genotyped for
jects [148]. When a major metabolic pathway is inhibited CYP2C19 and receiving concomitant administration of
by a genetic variant, such as CYP2C9 for warfarin, the voriconazole and tacrolimus. In this case, genetic defects of
contribution of other pathways increases. This has been CYP2C19 were associated with increased exposure to
demonstrated in CYP2C9-genotyped patients treated with voriconazole and corresponding interaction with the
warfarin [149] for which the coadministration of simvas- CYP3A substrate tacrolimus, which was greater in
tatin (a substrate of CYP3A) required an approximately CYP2C19 PMs than EMs [153]. Among the case reports
29% dose reduction in PMs compared with 5% in EMs described herein, many were related to the interaction
[149]. In such cases, an inhibitor of the minor pathway that between tacrolimus and proton pump inhibitors (PPIs).
would not have any significant impact in a wild-type Lansoprazole and omeprazole may affect the elimination of
patient can cause severe DDIs in patients with a gene- tacrolimus through competition for the CYP3A pathway. In
related defect of the major clearance pathway. the presence of a genetic defect of CYP2C19, the exposure
In the second scenario, where a victim drug is initiated of PPIs increases, which directly affects the magnitude of
in a patient treated with a perpetrator drug, the impact of interaction between the concomitantly administered drugs.
the interaction results from the superposition of GDI and This was described in patients carrying homozygous and
DDI mechanisms. In the absence of an inhibitor/inducer, heterozygous non-functional CYP2C19 alleles, such as *2
dose adjustment of a CYP substrate with a genetic variant and *3 [63, 64, 67, 68]. This interaction may even be
might be performed during treatment initiation. In the case worsened by the addition of other genetic variants affecting
of concomitant administration of a perpetrator of DDIs, tacrolimus elimination, such as CYP3A5*3 (no expression
dose adjustment is complicated and requires taking into of the CYP3A5 enzyme) or loss-of-function mutations of
account both GDIs and DDIs. A genetic variant decreasing the ABCB1 gene coding for the efflux transporter P-gp.
the activity of a specific CYP isoform will result in To sum, complex DDGIs involve several mechanisms:
increased vulnerability to DDIs. IM subjects are thought to effect on the exposure of the perpetrator, effect on the
be more vulnerable to phenoconversion than EMs, while exposure of the victim drug, and effect on the magnitude of
ultrarapid metabolizers (UMs) were less vulnerable DDIs. These three mechanisms may be concomitant and
because of higher basal CYP activity. Indeed, a higher cause unexpected serious ADRs.
proportion of subjects in an IM population will be con- In recent years, there has been growing interest for the
verted to PMs (and will therefore not be able to clear prediction of such complex DDGIs. Tod et al. published a
substrate drugs of the polymorphic enzyme) than in a UM mechanistic static approach to predict the magnitude of
population, whose basal capacity of drug metabolism is DDIs involving CYPs and several genotypes. This
higher. For example, in patients treated for depression by approach takes into account the contribution of each indi-
any known inhibitor of CYP2D6, phenoconversion to PM vidual isoenzyme to the clearance of the drug (CR), the
was observed in 24% of patients. After stratification of impact of genetic variation on the residual CYP activity,
study subjects by their genotype, it appeared that conver- and the potency of perpetrator drugs. [154] The DDI sim-
sion to the PM phenotype occurred in 21% of genetic EM ulator ‘DDI Predictor’, available online at www.
subjects and in only 3% of genetic UM subjects [150]. In ddipredictor.org, is based on this quantitative prediction
patients taking acenocoumarol, the presence of genetic [155]; however, this static approach does not take into
1282 F. Storelli et al.

account the genetic-related changes in perpetrator drug inflammatory cytokines) [174]. However, despite accu-
exposure. By defining drug-dependent and system-depen- mulating evidence, trends in the impact of renal impair-
dent parameters in a single prediction model, PBPK ment on CYP abundance and/or CYP intrinsic clearance
modeling offers the needed tools to allow for accurate are not consistent [160]. Physiological changes that occur
prediction of complex DDGIs. Vieira and colleagues have in kidney diseases may alter the clearance of non-renally
recently shown that the additive impact of genetic poly- eliminated drugs. In particular, hypoalbuminemia and drug
morphisms of CYP2D6 on the interaction between displacement by endogenous compounds (e.g. hippuric
risperidone and fluoxetine could be successfully predicted acid) that accumulate in patients with impaired renal
by PBPK modeling [156]. function reduce albumin binding and therefore raise the
non-renal clearance of drugs through an increase in the
unbound fraction [171]. Thus, a lack of change in total
4 Renal Failure clearance may obscure reduced enzyme activity and/or
abundance in case of concomitant change of the unbound
Chronic kidney disease (CKD) is an increasing burden fraction. Therefore, further investigations are needed to
worldwide, with a consistent estimated global prevalence understand the net effect of renal disease on CYP activity
of between 11 and 13% [157]. CKD is defined by a and expression.
decrease in kidney function, shown by a GFR \ 60 mL/ Based on these considerations, kidney disease may
min/1.73 m2 and/or markers of kidney damage of at least affect the clinical impact of DDIs by two mechanisms
3 months’ duration [158]. The primary pathological fea- affecting either the exposure of the victim drug or the
tures of most CKDs are interstitial fibrosis, tubular atrophy, extent of the DDI: (1) alteration of the elimination of drugs
and glomerulosclerosis, and are mainly caused by ever- whose clearance partially or mainly depends on renal
increasing pathologies such as diabetes and hypertension function; and (2) inhibition of CYP activity induced by the
[158]. Acute kidney injury (AKI) arises from various eti- disease. In addition, the extent of the DDI is impacted by
ologies such as tubular, glomerular, and vascular damages exposure of the perpetrator, which may increase in cases of
[159, 160]. renal dysfunction. Moreover, by affecting both renal and
It is well accepted and intuitive that the clearance of CYP-mediated hepatic clearance, kidney disease alters the
drugs whose elimination mainly depends on renal filtration relative contributions of CYP enzymes and therefore the
and/or renal transporters is decreased in CKD and AKI. In magnitude of CYP-related DDIs.
addition, an increasing number of in vitro [161–165] and Among the 29 cases of DDDIs, 21 involved renal fail-
in vivo (in rats [164, 166, 167] and humans [168, 169]) ure. Simvastatin, whose elimination is mainly cleared by
studies published in the last 15 years revealed that CKD CYP3A with negligible renal participation (\ 0.5% is
and end-stage renal disease (ESRD) may decrease the non- recovered unchanged in urine), exhibits reduced non-renal
renal clearance of drugs eliminated by metabolizing clearance in CKD patients [171]. Indeed, patients with
enzymes and transporters. These data have been incorpo- severe renal failure (GFR \ 30 mL/min) showed twofold
rated in a PBPK model of renal impairment in which CYP higher plasma concentrations after a single dose compared
abundance was reduced by 15–85% depending on CYP with healthy volunteers [175]. The present review descri-
isoform and renal disease severity compared with healthy bed six cases of simvastatin toxicity related to DDDIs
controls [170]. In a survey by the US FDA, it was reported (rhabdomyolysis, elevation of liver function tests, renal
that for over 23 new drugs for which applications included failure). The perpetrators were not only well-known strong
renal impairment studies, 13 exhibited an increased expo- CYP3A inhibitors, such as clarithromycin and amiodarone,
sure in patients with renal impairment (50% on average) but also weak inhibitors of CYP3A, such as azithromycin,
compared with healthy volunteers. Yeung and colleagues and drugs metabolized by the CYP3A pathway that might
reported 76 drugs that exhibited altered non-renal clearance competitively alter the CYP3A-mediated metabolism of
in patients with CKD [171], most of which were eliminated simvastatin, such as colchicine, tacrolimus, sirolimus, and
by CYP-mediated hepatic metabolism. Based on these telaprevir. In those cases, the association of both kidney
data, both the FDA and the European Medicines Agency disease and DDIs probably resulted in the development of
(EMA) currently recommend performing PK studies of ADRs. Of note, in three of these cases the daily dose of
non-renally cleared drugs in ESRD patients [172, 173]. To simvastatin was 80 mg, which is associated with a four-
date, underlying mechanisms of a drug non-renal clearance and tenfold higher risk of elevation of liver function tests
decrease in CKD patients are unclear but seem to be related and creatine kinase, respectively, compared with moderate
to downregulation of CYP gene expression and/or direct dosing [176].
inhibition of CYP by the accumulation of uremic toxins Colchicine is predominantly eliminated by biliary
(urea, parathyroid hormone, indoxyl sulfate, and excretion and is a substrate of P-gp. CYP3A-mediated
Complex Drug–Drug–Gene–Disease Interactions 1283

hepatic metabolism and renal elimination account only for the reduction of liver blood flow, the presence of portal-
5–20 and 10–20%, respectively, of drug disposition in systemic shunting, and a reduction in the number and
normal conditions [82]; however, their impact might activity of hepatocytes, as well as in impaired production
become critical in cases of DDIs or genetic variants of drug-binding proteins [178]. In addition, renal function
affecting P-gp [82]. The present review describes five cases might often become impaired in advanced liver disease,
of colchicine toxicity—four with clarithromycin and one often corresponding to hepatorenal syndrome [180].
with cyclosporin. The two perpetrators are both CYP3A For drugs metabolized primarily in the liver, the total
and P-gp inhibitors. Colchicine toxicity was precipitated by clearance of a compound is the hepatic clearance, which is
the superposition of three mechanisms: reduction of biliary the product of liver blood flow (QH) and the hepatic
excretion by inhibition of P-gp, inhibition of CYP3A-me- extraction ratio (EH), the latter being dependent on the
diated metabolism, and alteration of renal elimination unbound fraction of the drug in blood (fu,B), the unbound
related to kidney disease. The disposition of clarithromycin intrinsic clearance (CLu,int), and the liver blood flow, as
is dependent on renal elimination, and kidney dysfunction illustrated in Eq. 1:
may have led to increased exposure of the perpetrator, fu;B  CLu;int
therefore worsening the DDI. CLH ¼ QH  EH ¼ QH  : ð1Þ
QH þ fu;B  CLu;int
Overall, renal impairment was found to be a concomi-
tant disease in more than 20 cases of DDIs. The drugs The hepatic clearance of drugs with high extraction
involved in the DDDIs were primarily cleared by non-renal ratios (EH [ 0.7) is limited by hepatic blood flow;
routes. Renal impairment probably acted as a compounding therefore, the latter has a high impact on the clearance of
factor of DDIs, leading to clinical toxicity by increasing the highly extracted drugs. Oppositely, hepatic clearance of
victim drug’s exposure through inhibition of renal elimi- drugs with low extraction ratios (EH \ 0.3) is considered to
nation and/or decrease of CYP expression and/or activity. be limited by the metabolic capacity. Their clearance is
In addition, the magnitude of DDIs may have been wors- therefore impacted by changes in protein binding and
ened by an increase in the inhibitor’s exposure. intrinsic hepatic clearance, both occurring in liver disease.
System-dependent parameters such as enzyme abun- Finally, drugs with intermediate extraction ratios
dance, gastric emptying time, binding proteins in plasma, (0.3 \ EH \ 0.7) can be impacted by alterations of one
and hematocrit have been incorporated in renally impaired of the three components of the equation, i.e. liver blood
virtual populations in PBPK software (SimCYPÒ, Certara, flow, protein binding, and intrinsic hepatic clearance.
Princeton, NJ, USA). Provided that a PBPK model is As a result of a reduced first-pass effect, the bioavail-
capable of predicting comparable PK changes observed ability of intermediate to highly extracted drugs is signifi-
in vivo by both individual intrinsic (renal impairment) and cantly increased in cirrhotic patients. In addition, the
extrinsic (interacting drugs) factors, it can be used to pre- impaired production of albumin and a-glycoprotein related
dict the combined effect of those factors in common to liver dysfunction causes an increase in the unbound
complex scenarios. However, this is not always the case, as fraction of drugs. This change particularly affects the PK of
shown in the simulated interaction between telithromycin highly bound drugs (fu,B \ 0.1), impacting both their dis-
and ketoconazole that yielded a predicted AUC ratio lower tribution (increased volume of distribution) and clearance
than that observed in vivo [177]. These limited observa- (only the unbound fraction is cleared by the liver). The
tions (derived from only two subjects) warrant further intrinsic metabolic capacity is also altered in liver disease.
development and validation of current PBPK models. Indeed, it appears that the expression and activity of CYP
are selectively reduced in liver disease. The extent of
impairment of metabolic activity and enzyme expression
5 Hepatic Impairment: Cirrhosis depends on CYP isoform, and the type and severity of liver
disease [181]. In addition, the impairment of renal function
The liver plays a major role in the absorption, distribution, in advanced cirrhosis might explain the impact of cirrhosis
and elimination of many drugs and their active and inactive in the PK of drugs with predominant renal elimination.
metabolites. Liver diseases, particularly cirrhosis, result in It was postulated that the reduction of hepatic blood flow
numerous physiological changes that may influence drug in cirrhosis may lead to an increased sensitivity of flow-
PK [178]. Liver cirrhosis is characterized by the progres- limited clearance drugs with high extraction ratios to the
sive loss of functional hepatocytes, with concomitant for- action of enzymatic inhibitors. Indeed, flow-limited drugs
mation of connective tissues and nodules [179]. might become capacity-limited with reduction of hepatic
Progression of the disease might be classified using the blood flow [182]; however, it seems that the magnitude of
Child–Pugh score, which defines the severity of the disease DDIs decreases as the liver dysfunction progresses. Indeed,
(A, B, C) and the estimated prognosis. Cirrhosis results in the clearance of lidocaine, a flow-dependent drug with a
1284 F. Storelli et al.

high extraction ratio, was decreased by 60% in healthy metabolism due to hepatocellular carcinoma and cirrhosis.
volunteers, and by 44 and 9% in Child–Pugh A and C Indeed in vitro maximal velocities of sorafenib oxidation
cirrhotic patients, respectively, when coadministered with and glucuronidation were reduced by 25- and 2-fold,
fluvoxamine, an inhibitor of CYP1A2 and CYP3A [183]. respectively, in tumor liver microsomes compared with
The magnitude of the decrease of theophylline clearance by control hepatic microsomes [188]. Moreover, among hep-
fluvoxamine was also dependent on liver function, with atocellular carcinoma patients, those with Child–Pugh B
inhibition of 65, 55 and 14% in healthy volunteers and cirrhosis showed 20% higher sorafenib exposure compared
Child–Pugh A and C cirrhotic patients, respectively [184]. with Child–Pugh A cirrhosis [98].
The impact of clearance inhibition of the CYP3A substrate To summarize, cirrhosis is associated with decreased
quinine by erythromycin was also decreased in cirrhotic CYP expression, liver blood flow, and hepatic uptake,
patients but to a lesser extent, probably because of addi- which lead to alteration of drug hepatic clearance. It might
tional mechanism of competition for protein binding with also affect renal function through hepatorenal syndrome.
erythromycin that may mask the decrease in hepatic This might result in the achievement of toxic levels of
clearance [182]. Therefore, it seems that decreased uptake drugs, potentially leading to clinically relevant conse-
of the CYP inhibitor in hepatocytes, together with quences. Regarding the magnitude of DDIs, current
decreased enzyme expression (and thus decreased contri- knowledge indicates that the impact of CYP inhibitors
bution of the latter to drug clearance) are responsible for might be reduced by liver dysfunction. Therefore, while the
the decreased extent of CYP inhibition on liver cirrhosis. addition of a CYP inhibitor might require dose adaptation
The mechanism of inhibition, whether reversible or irre- in the presence of a CYP inhibitor in healthy volunteers, it
versible, might also play a role in the liver status-depen- might not be the case for advanced cirrhotic patients. Static
dency of DDIs [184]. [189] and dynamic models [179, 190] have been proposed
Regarding CYP induction, it was demonstrated in rats to predict the exposure of drugs in cirrhotic patients based
that severe liver dysfunction reduces the expression of the on the Child–Pugh classification. When incorporating cir-
aryl hydrocarbon receptor (AhR), and consequently the rhosis-related physiological changes into the semi-mecha-
expression and inducibility of CYP1A enzymes [185]. To nistic PBPK models, predicted drug concentrations
our knowledge, no human clinical study was properly corroborated with observed data [179, 190]. However,
designed to assess the impact of liver dysfunction on CYP recent work indicates underprediction of lopinavir expo-
induction. sure changes in cirrhotic patients by PBPK modeling,
Because the impact of liver dysfunction on drug PK and which highlights the need to provide additional efforts to
DDIs was mostly described with cirrhosis, only the latter address knowledge gaps in virtual hepatic impairment
was selected in the present review. Seven cases of DDDIs populations before being able to predict complex DDIs
involved cirrhosis as a concomitant disease. Given previ- involving liver diseases with PBPK modeling [191].
ous considerations related to the impact of liver dysfunc-
tion on the extent of DDIs, it seems that cirrhosis only
represents a compounding factor through elevation of basal 6 Inflammation
circulating levels of the victim drugs and not through an
increase of the magnitude of DDIs. While cirrhosis might Systemic inflammation markers have been identified in a
minimize the magnitude of DDIs, in the cases reported number of diseases, such as advanced cancer, inflammatory
herein, it was responsible for the decreased clearance of diseases (rheumatoid arthritis, psoriasis), and infection
victim drugs even before addition of the perpetrator, so that (influenza infection, sepsis) [192]. In addition, inflamma-
the toxic range was more easily reached in DDI conditions. tion is the result of oncologic treatment by recently mar-
Interestingly, the steady-state plasma levels of sorafenib, keted immunotherapy agents [193, 194]. With the recent
a CYP3A substrate whose liver metabolism is minor development of such immunotherapies, closer scientific
(\ 30%) [186], were increased threefold 2 weeks after the attention has been brought to the reduction of CYP-medi-
introduction of felodipine, another substrate of CYP3A, in ated drug metabolism and transport in inflammatory con-
a patient with hepatocellular carcinoma. While ketocona- ditions [194].
zole did not increase the AUC of single-dose sorafenib in The underlying mechanisms of reduced CYP activity
healthy volunteers [187], the introduction of the CYP3A and expression have been investigated in vitro [195–200]
substrate felodipine resulted in a clinically significant and are not only related to transcriptional suppression but
increase in sorafenib steady-state levels. The explanation also in part to post-translational protein modification
for such significant DDIs may involve other routes of caused by proinflammatory cytokines, with the most potent
elimination (sorafenib is also a substrate of UDP-glu- being IL-6, tumor necrosis factor (TNF)-a, IL-1b, and
curonyltransferases [48]), as well as alteration of sorafenib interferon (IFN)-c [201]. In rheumatoid arthritis patients,
Complex Drug–Drug–Gene–Disease Interactions 1285

tocilizumab, a humanized, monoclonal, antihuman IL-6 evidence is still scarce and is mostly limited to the impact
receptor (IL-6R) antibody, reversed IL-6-induced sup- of IL-6 on CYP3A activity. Further research is needed in
pression of CYP3A4 activity, as demonstrated by a order to investigate the impact of both chronic and acute
decrease of simvastatin exposure of approximately 60 and inflammation on other CYPs and transporters. Nonetheless,
38% 1 and 5 weeks after tocilizumab infusion, respectively available population PK (PopPK) and PBPK models have
[202]. Similarly, sarilumab, a human monoclonal antibody shown their ability to simulate interactions between
blocking IL-6R, also provoked a decrease in simvastatin inflammatory diseases and the PK of CYP substrates,
exposure of approximately 50% 7 days after a single-dose which paves the way for model-informed drug exposure
administration [203]. Using a cocktail approach, the predictions and dose-finding strategies. As with the other
activity of CYP3A (probe midazolam), CYP2C19 (probe diseases described in the present review, PBPK might
omeprazole), and CYP2C9 (probe warfarin) was increased represent a useful tool in order to predict complex sce-
after sirukumab administration in rheumatoid arthritis narios of DDIs in the context of systemic inflammation,
patients [204]. Consequently, PBPK models were devel- provided that the PK changes related to inflammatory sta-
oped in order to predict the impact of elevated IL-6 levels tus that are fed into the model are validated. Moreover, the
on CYP3A4 and the treatment effect of tocilizumab and impact of surrogate markers of systemic inflammation
sirukumab in rheumatoid arthritis patients [205, 206]. other than IL-6 on CYP activity and/or expression, as well
Moreover, cancer immunotherapy agents have also been as their combination, remains to be investigated.
associated with IL-6-mediated CYP suppression. [193]
While the impact of IL-6 as a marker of systemic inflam-
mation on CYP3A repression now appears to be clear, 7 Clinical Perspectives
additional knowledge is required in order to assess the
impact of disease-related inflammatory response on drug Computerized physician order entry (CPOE) systems cur-
exposure. rently include DDI checking tools, most of which are
The effect of inflammation on DDIs has only been integrating alert systems [209]. In some advanced systems,
investigated in relation to CYP induction, and it was evi- renal and hepatic functions can also be incorporated, and
denced in human hepatocytes that proinflammatory dose adaptation might be recommended according to the
cytokines such as IL-6 or TNFa inhibit the b-naph- estimated GFR and/or liver functional status [210]. A
thoflavone-mediated induction of CYP1A enzymes and the recent meta-analysis highlighted the discrepancy between
rifampin-mediated induction of CYP3A [207]. The the high prevalence of potential DDIs and the low preva-
inducibility of CYP2B6 and CYP2C was also decreased by lence of actual DDIs leading to clinical harm [211]. DDI
IL-6, probably through negative regulation of the nuclear checking tools included in CPOE systems only have poor
constitutive androstane receptor (CAR) and the pregnane X positive predictability and may cause annoyance and
receptor (PXR) gene expression [208]. frustration through overexposure to prescription alerts
In the present case reports review, only three cases of [212], especially when prescription assistance is lacking.
DDIs in inflammatory status patients were retained from While therapeutic drug monitoring might represent the
the literature search, mainly because inflammatory markers gold standard for assessing drug exposure, the approach is
such as CRP, IL-6, or white blood cell count were rarely not feasible for all drugs and patients, it is resource-con-
reported in reviewed publications. As reported in advanced suming, and it cannot predict exposure prior to the actual
cancer patients, CRP, IL-6, a1-acid glycoprotein, and drug dosing. Phenotyping of drug-metabolizing enzymes
TNFa seem to be the inflammatory markers that have the has the major advantage of taking into account both
greatest correlation between inflammatory response and intrinsic and extrinsic factors influencing enzyme activity
decreased CYP-mediated drug clearance [192]. and expression, and is currently often used in Geneva
In two of these cases, inflammation was a compounding University Hospitals in order to support the management of
factor of DDI-related ADRs, in addition to renal impair- complex drug interaction scenarios [213]. However, this
ment, cirrhosis, or genetic polymorphisms. As an example, approach is not yet performed routinely and requires time
severe extrapyramidal symptoms were related to haloperi- for analysis and administration of xenobiotics, even at low
dol toxicity when comedicated with the moderate CYP3A doses. The management of complex drug interaction sce-
inhibitor ciprofloxacin in a patient, combining four com- narios involving intrinsic (genetic polymorphisms and/or
pounding factors: CYP2D6*6/*6 (PM) and UGT2B7 -161 diseases affecting drug disposition) and extrinsic (con-
CT (IM) genotypes, cirrhosis, and acute inflammation as comitant medications) factors is thus challenging and relies
reported by high CRP levels [61]. on several prerequisites.
Although significant advances have been proposed in First, the prescriber needs to be aware of concomitant
the field of inflammation-induced CYP repression, clinical medications, diseases, or medical conditions that may
1286 F. Storelli et al.

affect the PK of the prescribed treatment. Therefore, pre- healthcare professionals, seems necessary. Pharmacoge-
and post-graduate education of physicians to PK, pharma- netic education to physicians allowed the successful
cogenetics, and DDIs is highly recommended to ensure that adoption of healthcare models of pre-emptive pharmaco-
prescribers are aware of intrinsic and extrinsic factors genetic testing [215]. In addition, the clinical validation of
leading to drug safety and efficacy issues. In addition, the pharmacogenetic markers is an important prerequisite for
use of advanced CPOE systems that integrate a DDI further implementation. Pharmacogenomics data may not
checking tool, laboratory data (genetic results, liver func- be sufficient to reliably predict a clinical response to a
tion tests, creatinine clearance/GFR, biomarkers of sys- drug, particularly when the impact of genetic variants on
temic inflammation), and clinical information (known the PD outcome is moderate [216] or when the relation
allergies, pregnancy, weight, sex) may represent a great with clinical outcome is affected by other patients’ char-
asset to gather all relevant data needed to optimize drug acteristics or clinical settings [217].
prescription. Current knowledge does not yet allow for the manage-
Moreover, the main issue remains the ability to accu- ment of complex drug interactions, even though static
rately predict the clinical outcome of such complex drug mechanistic models and dynamic PBPK models have been
interactions. In clinical practice, the prescriber may not developed to address this issue. While a static prediction
have sufficient PK knowledge and confidence to adjust tool might present the advantage of simplicity and appli-
drug dosage based on the patients’ characteristics, and may cability in a clinical setting, PBPK modeling seems more
refer to a clinical pharmacology consultancy. Although able to handle such complex scenarios, as demonstrated
pharmacogenetics is expanding in clinical practice, the with DDIs involving genetic polymorphisms or race-re-
prescriber is often confused when interpreting a pharma- lated changes in CYP abundance [156, 218]. In the past
cogenetic result. A US survey reported that 98% of years, model-informed approaches have been successively
physicians agree that genetics may predict drug response; used to optimize drug dosing in special populations and
however, only 10% feel adequately informed to actually appear to be efficient tools for improving the efficacy and
use genetic tests [214]. Therefore, providing more focus on safety of a drug treatment for an individual patient by
pharmacogenetics, which is expected to grow bigger and integrating its own characteristics in the model [219].
bigger in future years with the paradigm shifting from While a few issues currently hamper their widespread use
universal to personalized medicine, in the education of in clinical practice, including limited evidence of efficacy,

Fig. 2 Integrated PBPK model-based informed dose adaptation for metabolizing enzymes and transporters, GFR glomerular filtration
the management of complex drug interactions in clinical practice. rate, PBPK physiologically-based pharmacokinetics
CPOE Computerized Physician Order Entry, DMET drug-
Complex Drug–Drug–Gene–Disease Interactions 1287

weak awareness of the approach in the clinical community, meta-analysis. Pharmacoepidemiol Drug Saf.
and lack of cost–benefit analysis, we think that future 2012;21(11):1139–54.
2. Marengoni A, Pasina L, Concoreggi C, et al. Understanding
implementation of such comprehensive tools may help to adverse drug reactions in older adults through drug-drug inter-
manage complex clinical scenarios in relation to drug actions. Eur J Intern Med. 2014;25(9):843–6.
dosing (see Fig. 2). The development of international 3. Pirmohamed M, James S, Meakin S, et al. Adverse drug reac-
guidelines for the management of complex drug interac- tions as cause of admission to hospital: prospective analysis of
18 820 patients. BMJ. 2004;329(7456):15–9.
tions is however a prerequisite for further clinical 4. McDonnell PJ, Jacobs MR. Hospital admissions resulting from
implementation. preventable adverse drug reactions. Ann Pharmacother.
2002;36(9):1331–6.
5. Goldberg RM, Mabee J, Chan L, et al. Drug-drug and drug-
disease interactions in the ED: analysis of a high-risk popula-
8 Conclusions tion. Am J Emerg Med. 1996;14(5):447–50.
6. Hohl CM, Dankoff J, Colacone A, et al. Polypharmacy, adverse
More than 60 case reports of complex DDIs were identified drug-related events, and potential adverse drug interactions in
from our systematic literature search. The analysis of these elderly patients presenting to an emergency department. Ann
Emerg Med. 2001;38(6):666–71.
reports on DDIs involving intrinsic factors such as genetic 7. Lexi-Interact (database). 2017. Available at: www.lexi.com/.
polymorphisms or diseases showed their association with Accessed 23 Aug 2017.
drug exposure. Although the additive role of intrinsic 8. Multicheck Drug Interaction Checker (database) 2017. http://
factors in the clinical relevance of DDIs is not clearly online.epocrates.com/noFrame/. Accessed 23 Aug 2017.
9. Thériaque (database). 2017. www.theriaque.org/apps/recherche/
demonstrated in some case reports presented in this sys- rch_simple.php. Accessed 23 Aug 2017.
tematic review, there is evidence suggesting that both 10. Marsousi N, Samer CF, Fontana P, et al. Coadministration of
intrinsic and/or extrinsic factors have an impact on the ticagrelor and ritonavir: toward prospective dose adjustment to
extent of DDIs and the severity of clinical outcomes. maintain an optimal platelet inhibition using the PBPK
approach. Clin Pharmacol Ther. 2016;100(3):295–304.
Indisputably, the presented case reports are real-life illus- 11. Moher D, Pham B, Klassen TP, et al. What contributions do
trations of the issue of complex DDIs in clinical practice. languages other than English make on the results of meta-
Further research and dedicated clinical trials are needed to analyses? J Clin Epidemiol. 2000;53(9):964–72.
appraise the clinical relevance of complex drug interactions 12. Bouatou Y, Samer CF, Ing Lorenzini KR, et al. Therapeutic
drug monitoring of voriconazole: a case report of multiple drug
involving extrinsic and intrinsic factors. Current data interactions in a patient with an increased CYP2C19 activity.
indicate a greater variability in drug levels and exposure AIDS Res Ther. 2014;11:25.
when such complex DDIs are present, compared with 13. Ozdemir V, Kalow W, Okey AB, et al. Treatment-resistance to
simple DDIs. This variability results in poor predictability clozapine in association with ultrarapid CYP1A2 activity and
the C– [ A polymorphism in intron 1 of the CYP1A2 gene:
of the DDIs and to the potentially dramatic clinical con- effect of grapefruit juice and low-dose fluvoxamine. J Clin
sequences that were described in the present review. In Psychopharmacol. 2001;21(6):603–7.
clinical practice, the management of such complex sce- 14. Shah RR, Smith RL. Addressing phenoconversion: the Achilles’
narios of DDIs is currently empiric due to the lack of heel of personalized medicine. Br J Clin Pharmacol.
2015;79(2):222–40.
prediction tools. The future development of such tools 15. Zanger UM, Schwab M. Cytochrome P450 enzymes in drug
available for clinical practice, including PBPK model- metabolism: regulation of gene expression, enzyme activities,
based precision dosing tools, is expected to bring more and impact of genetic variation. Pharmacol Ther.
confidence in the management of complex scenarios and 2013;138(1):103–41.
16. Haffen E, Vandel P, Broly F, et al. Citalopram: an interaction
therefore to decreasing the rate of DDI-related adverse study with clomipramine in a patient heterozygous for CYP2D6
reactions. genotype. Pharmacopsychiatry. 1999;32(6):232–4.
17. Kohnke MD, Griese EU, Stosser D, et al. Cytochrome P450 2D6
Compliance with Ethical Standards deficiency and its clinical relevance in a patient treated with
risperidone. Pharmacopsychiatry. 2002;35(3):116–8.
Funding No funding was received for the preparation of this 18. Kato D, Kawanishi C, Kishida I, et al. CYP2D6 gene deletion
manuscript. allele in patients with neuroleptic malignant syndrome: prelim-
inary report. Psychiatry Clin Neurosci. 2005;59(4):504–7.
Conflict of interest Flavia Storelli, Caroline Samer, Jean-Luc Reny, 19. Nara M, Takahashi N, Miura M, et al. Effect of oral itraconazole
Jules Desmeules and Youssef Daali declare no conflicts of interest. on the pharmacokinetics of tacrolimus in a hematopoietic stem
cell transplant recipient with CYP3A5*3/*3. Am J Hematol.
2010;85(8):634–5.
20. Madadi P, Hildebrandt D, Gong IY, et al. Fatal hydrocodone
overdose in a child: pharmacogenetics and drug interactions.
References Pediatrics. 2010;126(4):e986–9.
21. Suan D, O’Connor K, Booth DR, et al. Voriconazole toxicity
1. Miguel A, Azevedo LF, Araujo M, et al. Frequency of adverse related to polymorphisms in CYP2C19. Intern Med J.
drug reactions in hospitalized patients: a systematic review and 2011;41(4):364–5.
1288 F. Storelli et al.

22. Hu M, Mak VW, Tomlinson B. Simvastatin-induced myopathy, 41. Akdag I, Ersoy A, Kahvecioglu S, et al. Acute colchicine
the role of interaction with diltiazem and genetic predisposition. intoxication during clarithromycin administration in patients
J Clin Pharm Ther. 2011;36(3):419–25. with chronic renal failure. J Nephrol. 2006;19(4):515–7.
23. Malingre MM, Godschalk PC, Klein SK. A case report of 42. van der Velden W, Huussen J, Ter Laak H, et al. Colchicine-
voriconazole therapy failure in a homozygous ultrarapid induced neuromyopathy in a patient with chronic renal failure:
CYP2C19*17/*17 patient comedicated with carbamazepine. Br the role of clarithromycin. Neth J Med. 2008;66(5):204–6.
J Clin Pharmacol. 2012;74(1):205–6. 43. Priou P, Gagnadoux F, Dehe C, et al. Drug-induced pneumonitis
24. Yang WH, Zeng ZS, Ren XW, et al. Simvastatin-induced in a patient treated with venlafaxine and propanolol [in French].
myopathy with concomitant use of cyclosporine: case report. Int Rev Mal Respir. 2008;25(5):610–3.
J Clin Pharmacol Ther. 2011;49(12):772–7. 44. Eleftheriou G, Bacis G, Fiocchi R, et al. Colchicine-induced
25. Fujita Y, Araki T, Okada Y, et al. Analysis of cytochrome P450 toxicity in a heart transplant patient with chronic renal failure.
gene polymorphism in a lupus nephritis patient in whom Clin Toxicol (Phila). 2008;46(9):827–30.
tacrolimus blood concentration was markedly elevated after 45. Dopazo C, Bilbao I, Lazaro JL, et al. Severe rhabdomyolysis
administration of azole antifungal agents. J Clin Pharm Ther. and acute renal failure secondary to concomitant use of sim-
2013;38(1):74–6. vastatin with rapamycin plus tacrolimus in liver transplant
26. Zhao W, Fakhoury M, Maisin A, et al. Pharmacogenetic patient. Transpl Proc. 2009;41(3):1021–4.
determinant of the drug interaction between tacrolimus and 46. McKinnell J, Tayek JA. Short term treatment with clar-
omeprazole. Ther Drug Monit. 2012;34(6):739–41. ithromycin resulting in colchicine-induced rhabdomyolysis.
27. Duricova J, Perinova I, Jurckova N, et al. Clinically important J Clin Rheumatol. 2009;15(6):303–5.
interaction between metoprolol and propafenone. Can Fam 47. Roustit M, Blondel E, Villier C, et al. Symptomatic hypo-
Phys. 2013;59(4):373–5. glycemia associated with trimethoprim/sulfamethoxazole and
28. Chua EW, Foulds J, Miller AL, et al. Novel CYP2D6 and repaglinide in a diabetic patient. Ann Pharmacother.
CYP2C19 variants identified in a patient with adverse reactions 2010;44(4):764–7.
towards venlafaxine monotherapy and dual therapy with nor- 48. Gomo C, Coriat R, Faivre L, et al. Pharmacokinetic interaction
triptyline and fluoxetine. Pharmacogenet Genomics. involving sorafenib and the calcium-channel blocker felodipine
2013;23(9):494–7. in a patient with hepatocellular carcinoma. Invest New Drugs.
29. Tomisti L, Del Re M, Bartalena L, et al. Effects of amiodarone, 2011;29(6):1511–4.
thyroid hormones and CYP2C9 and VKORC1 polymorphisms 49. Levin TT, Bakr MH, Nikolova T. Case report: delirium due to a
on warfarin metabolism: a review of the literature. Endocr Pract. diltiazem-fentanyl CYP3A4 drug interaction. Gen Hosp Psy-
2013;19(6):1043–9. chiatry. 2010;32(6):648e9–650e10.
30. Wu AH, Kearney T. Lack of impairment due to confirmed 50. Frankel JK, Packer CD. Cushing’s syndrome due to antiretro-
codeine use prior to a motor vehicle accident: role of pharma- viral-budesonide interaction. Ann Pharmacother.
cogenomics. J Forensic Leg Med. 2013;20(8):1024–7. 2011;45(6):823–4.
31. Bebawi E, Jouni SS, Tessier AA, et al. A metoprolol-terbinafine 51. Marot A, Morelle J, Chouinard VA, et al. Concomitant use of
combination induced bradycardia. Eur J Drug Metab Pharma- simvastatin and amiodarone resulting in severe rhabdomyolysis:
cokinet. 2015;40(3):295–9. a case report and review of the literature. Acta Clin Belg.
32. Meyer zu Schwabedissen HE, Siegmund W, Kroemer HK, et al. 2011;66(2):134–6.
Creatine kinase elevation caused by a combination of fluvastatin 52. Hoover WC, Britton LJ, Gardner J, et al. Rapid onset of iatro-
and telmisartan in a patient heterozygous for the CYP2C9*3 and genic adrenal insufficiency in a patient with cystic fibrosis-re-
ABCC2-24C [ T variants: a case report. BMC Res Notes. lated liver disease treated with inhaled corticosteroids and a
2014;7:688. moderate CYP3A4 inhibitor. Ann Pharmacother.
33. van den Berg SAA, van ‘t Veer NE, Emmen JMA, et al. Flu- 2011;45(7–8):e38.
ticasone furoate induced iatrogenic Cushing syndrome in a 53. Alreja G, Inayatullah S, Goel S, et al. Rhabdomyolysis caused
pediatric patient receiving anti-retroviral therapy. Endocrinol by an unusual interaction between azithromycin and simvastatin.
Diabetes Metab Case Rep. 2017;2017:16–0158. J Cardiovasc Dis Res. 2012;3(4):319–22.
34. Calcagno A, Baietto L, Pagani N, et al. Voriconazole and ata- 54. Holstein A, Kovacs P, Beil W. Severe hypoglycemia due to
zanavir: a CYP2C19-dependent manageable drug-drug interac- possible interaction between glibenclamide and sorafenib in a
tion. Pharmacogenomics. 2014;15(10):1281–6. patient with hepatocellular carcinoma. Curr Drug Saf.
35. Hebert MF, Lam AY. Diltiazem increases tacrolimus concen- 2013;8(2):148–52.
trations. Ann Pharmacother. 1999;33(6):680–2. 55. Leung JG, Nelson S, Takala CR, et al. Infection and inflam-
36. Ahle GB, Blum AL, Martinek J, et al. Cushing’s syndrome in an mation leading to clozapine toxicity and intensive care: a case
81-year-old patient treated with budesonide and amiodarone. series. Ann Pharmacother. 2014;48(6):801–5.
Eur J Gastroenterol Hepatol. 2000;12(9):1041–2. 56. Kanter CT, Luin M, Solas C, et al. Rhabdomyolysis in a hep-
37. Lee AJ, Maddix DS. Rhabdomyolysis secondary to a drug atitis C virus infected patient treated with telaprevir and sim-
interaction between simvastatin and clarithromycin. Ann Phar- vastatin. Ann Hepatol. 2014;13(4):452–5.
macother. 2001;35(1):26–31. 57. Takeuchi S, Kotani Y, Tsujimoto T. Hypotension induced by the
38. Hsu WC, Chen WH, Chang MT, et al. Colchicine-induced acute concomitant use of a calcium-channel blocker and clar-
myopathy in a patient with concomitant use of simvastatin. Clin ithromycin. BMJ Case Rep. 2017. https://doi.org/10.1136/bcr-
Neuropharmacol. 2002;25(5):266–8. 2016-218388.
39. Sadaba B, Campanero MA, Quetglas EG, et al. Clinical rele- 58. Gasche Y, Daali Y, Fathi M, et al. Codeine intoxication asso-
vance of sirolimus drug interactions in transplant patients. ciated with ultrarapid CYP2D6 metabolism. N Engl J Med.
Transpl Proc. 2004;36(10):3226–8. 2004;351(27):2827–31.
40. Geronimo-Pardo M, Cuartero-del-Pozo AB, Jimenez-Vizuete 59. Tanaka A, Nagamatsu T, Yamaguchi M, et al. Myoclonus after
JM, et al. Clarithromycin-nifedipine interaction as possible dextromethorphan administration in peritoneal dialysis. Ann
cause of vasodilatory shock. Ann Pharmacother. Pharmacother. 2011;45(1):e1.
2005;39(3):538–42.
Complex Drug–Drug–Gene–Disease Interactions 1289

60. Ing Lorenzini K, Calmy A, Ambrosioni J, et al. Serotonin syn- 77. Edsbacker S, Andersson T. Pharmacokinetics of budesonide
drome following drug-drug interactions and CYP2D6 and (Entocort EC) capsules for Crohn’s disease. Clin Pharma-
CYP2C19 genetic polymorphisms in an HIV-infected patient. cokinet. 2004;43(12):803–21.
AIDS. 2012;26(18):2417–8. 78. Wojcikowski J, Boksa J, Daniel WA. Main contribution of the
61. Simic I, Potocnjak I, Kraljickovic I, et al. CYP2D6 *6/*6 cytochrome P450 isoenzyme 1A2 (CYP1A2) to N-demethyla-
genotype and drug interactions as cause of haloperidol-induced tion and 5-sulfoxidation of the phenothiazine neuroleptic
extrapyramidal symptoms. Pharmacogenomics. chlorpromazine in human liver: a comparison with other phe-
2016;17(13):1385–9. nothiazines. Biochem Pharmacol. 2010;80(8):1252–9.
62. Forget P, le Polain de Waroux B B, Wallemacq P, et al. Life- 79. Yoshii K, Kobayashi K, Tsumuji M, et al. Identification of
threatening dextromethorphan intoxication associated with human cytochrome P450 isoforms involved in the 7-hydroxy-
interaction with amitriptyline in a poor CYP2D6 metabolizer: a lation of chlorpromazine by human liver microsomes. Life Sci.
single case re-exposure study. J Pain Symptom Manage. 2000;67(2):175–84.
2008;36(1):92–6. 80. Nielsen KK, Flinois JP, Beaune P, et al. The biotransformation
63. Takahashi K, Motohashi H, Yonezawa A, et al. Lansoprazole- of clomipramine in vitro, identification of the cytochrome
tacrolimus interaction in Japanese transplant recipient with P450 s responsible for the separate metabolic pathways. J Phar-
CYP2C19 polymorphism. Ann Pharmacother. macol Exp Ther. 1996;277(3):1659–64.
2004;38(5):791–4. 81. Clozaril Drug Information. Novartis. http://www.accessdata.fda.
64. Hosohata K, Masuda S, Ogura Y, et al. Interaction between gov/drugsatfda_docs/label/2014/019758s073lbl.pdf. Accessed 9
tacrolimus and lansoprazole, but not rabeprazole in living-donor Feb 2018.
liver transplant patients with defects of CYP2C19 and CYP3A5. 82. Terkeltaub RA. Colchicine update: 2008. Semin Arthritis
Drug Metab Pharmacokinet. 2008;23(2):134–8. Rheum. 2009;38(6):411–9.
65. Gorny M, Rohm S, Laer S, et al. Pharmacogenomic adaptation 83. Molden E, Asberg A, Christensen H. CYP2D6 is involved in
of antiretroviral therapy: overcoming the failure of lopinavir in O-demethylation of diltiazem. An in vitro study with transfected
an African infant with CYP2D6 ultrarapid metabolism. Eur J human liver cells. Eur J Clin Pharmacol. 2000;56(8):575–9.
Clin Pharmacol. 2010;66(1):107–8. 84. Molden E, Johansen PW, Boe GH, et al. Pharmacokinetics of
66. Marusic S, Lisicic A, Horvatic I, et al. Atorvastatin-related diltiazem and its metabolites in relation to CYP2D6 genotype.
rhabdomyolysis and acute renal failure in a genetically predis- Clin Pharmacol Ther. 2002;72(3):333–42.
posed patient with potential drug-drug interaction. Int J Clin 85. Sutton D, Butler AM, Nadin L, et al. Role of CYP3A4 in human
Pharm. 2012;34(6):825–7. hepatic diltiazem N-demethylation: inhibition of CYP3A4
67. Homma M, Itagaki F, Yuzawa K, et al. Effects of lansoprazole activity by oxidized diltiazem metabolites. J Pharmacol Exp
and rabeprazole on tacrolimus blood concentration: case of a Ther. 1997;282(1):294–300.
renal transplant recipient with CYP2C19 gene mutation. 86. Ward BA, Gorski JC, Jones DR, et al. The cytochrome P450
Transplantation. 2002;73(2):303–4. 2B6 (CYP2B6) is the main catalyst of efavirenz primary and
68. Itagaki F, Homma M, Yuzawa K, et al. Drug interaction of secondary metabolism: implication for HIV/AIDS therapy and
tacrolimus and proton pump inhibitors in renal transplant utility of efavirenz as a substrate marker of CYP2B6 catalytic
recipients with CYP2C19 gene mutation. Transplant Proc. activity. J Pharmacol Exp Ther. 2003;306(1):287–300.
2002;34(7):2777–8. 87. Lexapro Drug Information. Allergan. http://www.accessdata.
69. Grozinger M, Hartter S, Hiemke C, et al. Interaction of mod- fda.gov/drugsatfda_docs/label/2009/
afinil and clomipramine as comedication in a narcoleptic patient. 021323s032,021365s023lbl.pdf. Accessed 23 Aug 2017.
Clin Neuropharmacol. 1998;21(2):127–9. 88. Flonase Drug Information. GlaxoSmithKline. http://www.
70. Spina E, Scordo MG, Avenoso A, et al. Adverse drug interaction accessdata.fda.gov/drugsatfda_docs/label/2003/020121s028lbl.
between risperidone and carbamazepine in a patient with pdf. Accessed 23 Aug 2017.
chronic schizophrenia and deficient CYP2D6 activity. J Clin 89. Zhou L, Naraharisetti SB, Liu L, et al. Contributions of human
Psychopharmacol. 2001;21(1):108–9. cytochrome P450 enzymes to glyburide metabolism. Biopharm
71. Hasse B, Gunthard HF, Bleiber G, et al. Efavirenz intoxication Drug Dispos. 2010;31(4):228–42.
due to slow hepatic metabolism. Clin Infect Dis. 90. Zharikova OL, Fokina VM, Nanovskaya TN, et al. Identification
2005;40(3):e22–3. of the major human hepatic and placental enzymes responsible
72. Lee HK, Lewis LD, Tsongalis GJ, et al. Negative urine opioid for the biotransformation of glyburide. Biochem Pharmacol.
screening caused by rifampin-mediated induction of oxycodone 2009;78(12):1483–90.
hepatic metabolism. Clin Chim Acta. 2006;367(1–2):196–200. 91. Diabeta Drug information. Sanofi-Aventis. http://www.
73. Elkalioubie A, Allorge D, Robriquet L, et al. Near-fatal tra- accessdata.fda.gov/drugsatfda_docs/label/2009/017532s030lbl.
madol cardiotoxicity in a CYP2D6 ultrarapid metabolizer. Eur J pdf. Accessed 9 Feb 2018.
Clin Pharmacol. 2011;67(8):855–8. 92. Hutchinson MR, Menelaou A, Foster DJ, et al. CYP2D6 and
74. Geber C, Ostad Haji E, Schlicht K, et al. Severe tremor after CYP3A4 involvement in the primary oxidative metabolism of
cotrimoxazole-induced elevation of venlafaxine serum concen- hydrocodone by human liver microsomes. Br J Clin Pharmacol.
trations in a patient with major depressive disorder. Ther Drug 2004;57(3):287–97.
Monit. 2013;35(3):279–82. 93. Kumar GN, Dykstra J, Roberts EM, et al. Potent inhibition of
75. Motta I, Calcagno A, Baietto L, et al. A probable drug-to-drug the cytochrome P-450 3A-mediated human liver microsomal
interaction between voriconazole and haloperidol in a CYP2C19 metabolism of a novel HIV protease inhibitor by ritonavir: a
poor metabolizing patient [corrected]. Infez Med. positive drug-drug interaction. Drug Metab Dispos.
2015;23(4):367–9. 1999;27(8):902–8.
76. Flockhart DA. Drug Interactions: Cytochromes P450 Drug 94. Olesen OV, Linnet K. Hydroxylation and demethylation of the
Interaction Table. Indiana University School of Medicine tricyclic antidepressant nortriptyline by cDNA-expressed human
(2007). http://medicine.iupui.edu/clinpharm/ddis/main-table. cytochrome P-450 isozymes. Drug Metab Dispos.
Accessed 8 Feb 2018. 1997;25(6):740–4.
1290 F. Storelli et al.

95. Venkatakrishnan K, von Moltke LL, Greenblatt DJ. Nortripty- between CYP2C19 genotypes. Br J Clin Pharmacol.
line E-10-hydroxylation in vitro is mediated by human CYP2D6 2004;57(4):487–94.
(high affinity) and CYP3A4 (low affinity): implications for 115. Galetin A, Clarke SE, Houston JB. Quinidine and haloperidol as
interactions with enzyme-inducing drugs. J Clin Pharmacol. modifiers of CYP3A4 activity: multisite kinetic model approach.
1999;39(6):567–77. Drug Metab Dispos. 2002;30(12):1512–22.
96. Samer CF, Daali Y, Wagner M, et al. The effects of CYP2D6 116. Shin JG, Kane K, Flockhart DA. Potent inhibition of CYP2D6
and CYP3A activities on the pharmacokinetics of immediate by haloperidol metabolites: stereoselective inhibition by reduced
release oxycodone. Br J Pharmacol. 2010;160(4):907–18. haloperidol. Br J Clin Pharmacol. 2001;51(1):45–52.
97. Sall C, Houston JB, Galetin A. A comprehensive assessment of 117. Yanagida Y, Watanabe M, Takeba Y, et al. Potential of lanso-
repaglinide metabolic pathways: impact of choice of in vitro prazole as a novel probe for cytochrome P450 3A activity by
system and relative enzyme contribution to in vitro clearance. measuring lansoprazole sulfone in human liver microsomes.
Drug Metab Dispos. 2012;40(7):1279–89. Biol Pharm Bull. 2009;32(8):1422–6.
98. Nevaxar FDA Drug Approval Package. Bayer. http://www. 118. Syvalahti EK, Lindberg R, Kallio J, et al. Inhibitory effects of
accessdata.fda.gov/drugsatfda_docs/nda/2005/021923_s000_ neuroleptics on debrisoquine oxidation in man. Br J Clin Phar-
NexavarTOC.cfm. Accessed 12 Feb 2018. macol. 1986;22(1):89–92.
99. Grond S, Sablotzki A. Clinical pharmacology of tramadol. Clin 119. Robertson P, DeCory HH, Madan A, et al. In vitro inhibition and
Pharmacokinet. 2004;43(13):879–923. induction of human hepatic cytochrome P450 enzymes by
100. Fogelman SM, Schmider J, Venkatakrishnan K, et al. O- and modafinil. Drug Metab Dispos. 2000;28(6):664–71.
N-demethylation of venlafaxine in vitro by human liver micro- 120. Hamelin BA, Bouayad A, Drolet B, et al. In vitro characteri-
somes and by microsomes from cDNA-transfected cells: effect zation of cytochrome P450 2D6 inhibition by classic histamine
of metabolic inhibitors and SSRI antidepressants. Neuropsy- H1 receptor antagonists. Drug Metab Dispos. 1998;26(6):536–9.
chopharmacology. 1999;20(5):480–90. 121. Johnson JA, Herring VL, Wolfe MS, et al. CYP1A2 and
101. Hyland R, Jones BC, Smith DA. Identification of the cyto- CYP2D6 4-hydroxylate propranolol and both reactions exhibit
chrome P450 enzymes involved in the N-oxidation of racial differences. J Pharmacol Exp Ther.
voriconazole. Drug Metab Dispos. 2003;31(5):540–7. 2000;294(3):1099–105.
102. Ufer M. Comparative pharmacokinetics of vitamin K antago- 122. Filppula AM, Neuvonen PJ, Backman JT. In vitro assessment of
nists: warfarin, phenprocoumon and acenocoumarol. Clin time-dependent inhibitory effects on CYP2C8 and CYP3A
Pharmacokinet. 2005;44(12):1227–46. activity by fourteen protein kinase inhibitors. Drug Metab Dis-
103. O’Reilly RA, Trager WF, Rettie AE, et al. Interaction of pos. 2014;42(7):1202–9.
amiodarone with racemic warfarin and its separated enan- 123. Chapron B, Risler L, Phillips B, et al. Reversible, time-depen-
tiomorphs in humans. Clin Pharmacol Ther. 1987;42(3):290–4. dent inhibition of CYP3A-mediated metabolism of midazolam
104. Abilify Drug Information. Otsuka. http://www.accessdata.fda. and tacrolimus by telaprevir in human liver microsomes.
gov/drugsatfda_docs/label/2014/ J Pharm Pharm Sci. 2015;18(1):101–11.
021436s038,021713s030,021729s022,021866s023lbl.pdf. 124. Garg V, Kauffman RS, Beaumont M, et al. Telaprevir: phar-
Accessed 23 Aug 2017. macokinetics and drug interactions. Antivir Ther.
105. Perloff ES, Duan SX, Skolnik PR, et al. Atazanavir: effects on 2012;17(7):1211–21.
P-glycoprotein transport and CYP3A metabolism in vitro. Drug 125. Kamiyama E, Yoshigae Y, Kasuya A, et al. Inhibitory effects of
Metab Dispos. 2005;33(6):764–70. angiotensin receptor blockers on CYP2C9 activity in human
106. Westphal JF. Macrolide-induced clinically relevant drug inter- liver microsomes. Drug Metab Pharmacokinet.
actions with cytochrome P-450A (CYP) 3A4: an update focused 2007;22(4):267–75.
on clarithromycin, azithromycin and dirithromycin. Br J Clin 126. Ll A, Berecz R, de la Rubia A, et al. Effect of thioridazine
Pharmacol. 2000;50(4):285–95. dosage on the debrisoquine hydroxylation phenotype in psy-
107. Shin JG, Soukhova N, Flockhart DA. Effect of antipsychotic chiatric patients with different CYP2D6 genotypes. Ther Drug
drugs on human liver cytochrome P-450 (CYP) isoforms Monit. 2001;23(6):616–20.
in vitro: preferential inhibition of CYP2D6. Drug Metab Dispos. 127. Wen X, Wang JS, Kivisto KT, et al. In vitro evaluation of
1999;27(9):1078–84. valproic acid as an inhibitor of human cytochrome P450 iso-
108. Suzuki Y, Someya T, Shimoda K, et al. Importance of the forms: preferential inhibition of cytochrome P450 2C9
cytochrome P450 2D6 genotype for the drug metabolic inter- (CYP2C9). Br J Clin Pharmacol. 2001;52(5):547–53.
action between chlorpromazine and haloperidol. Ther Drug 128. Saari TI, Laine K, Leino K, et al. Effect of voriconazole on the
Monit. 2001;23(4):363–8. pharmacokinetics and pharmacodynamics of intravenous and
109. Spina E, Martines C, Caputi AP, et al. Debrisoquine oxidation oral midazolam. Clin Pharmacol Ther. 2006;79(4):362–70.
phenotype during neuroleptic monotherapy. Eur J Clin Phar- 129. Zhou SF, Liu JP, Chowbay B. Polymorphism of human cyto-
macol. 1991;41(5):467–70. chrome P450 enzymes and its clinical impact. Drug Metab Rev.
110. Vlase L, Popa A, Neag M, et al. Pharmacokinetic interaction 2009;41(2):89–295.
between zolpidem and ciprofloxacin in healthy volunteers. Eur J 130. Valenzuela B, Rebollo J, Perez T, et al. Effect of grapefruit juice
Drug Metab Pharmacokinet. 2011;35(3–4):83–7. on the pharmacokinetics of docetaxel in cancer patients: a case
111. Gram LF, Hansen MG, Sindrup SH, et al. Citalopram: interac- report. Br J Clin Pharmacol. 2011;72(6):978–81.
tion studies with levomepromazine, imipramine, and lithium. 131. Dunnenberger HM, Crews KR, Hoffman JM, et al. Preemptive
Ther Drug Monit. 1993;15(1):18–24. clinical pharmacogenetics implementation: current programs in
112. Kajosaari LI, Niemi M, Neuvonen M, et al. Cyclosporine five US medical centers. Annu Rev Pharmacol Toxicol.
markedly raises the plasma concentrations of repaglinide. Clin 2015;55:89–106.
Pharmacol Ther. 2005;78(4):388–99. 132. Van Driest SL, Shi Y, Bowton EA, et al. Clinically actionable
113. Back D, Sekar V, Hoetelmans RM. Darunavir: pharmacokinetics genotypes among 10,000 patients with preemptive pharma-
and drug interactions. Antivir Ther. 2008;13(1):1–13. cogenomic testing. Clin Pharmacol Ther. 2014;95(4):423–31.
114. Yasui-Furukori N, Takahata T, Nakagami T, et al. Different
inhibitory effect of fluvoxamine on omeprazole metabolism
Complex Drug–Drug–Gene–Disease Interactions 1291

133. Goh LL, Lim CW, Sim WC, et al. Analysis of genetic variation 150. Preskorn SH, Kane CP, Lobello K, et al. Cytochrome P450 2D6
in CYP450 genes for clinical implementation. PLoS One. phenoconversion is common in patients being treated for
2017;12(1):e0169233. depression: implications for personalized medicine. J Clin Psy-
134. van der Wouden CH, Cambon-Thomsen A, Cecchin E, et al. chiatry. 2013;74(6):614–21.
Implementing pharmacogenomics in Europe: design and 151. Gschwind L, Rollason V, Boehlen F, et al. Impact of CYP2C9
implementation strategy of the ubiquitous pharmacogenomics polymorphisms on the vulnerability to pharmacokinetic drug-
consortium. Clin Pharmacol Ther. 2017;101(3):341–58. drug interactions during acenocoumarol treatment. Pharma-
135. Gottesman O, Scott SA, Ellis SB, et al. The CLIPMERGE PGx cogenomics. 2013;14(7):745–53.
Program: clinical implementation of personalized medicine 152. Storelli F, Matthey A, Lenglet S, et al. Impact of CYP2D6
through electronic health records and genomics-pharmacoge- functional allelic variations on phenoconversion and drug-drug
nomics. Clin Pharmacol Ther. 2013;94(2):214–7. interactions. Clin Pharmacol Ther. 2017. https://doi.org/10.
136. Rasmussen-Torvik LJ, Stallings SC, Gordon AS, et al. Design 1002/cpt.889.
and anticipated outcomes of the eMERGE-PGx project: a mul- 153. Imamura CK, Furihata K, Okamoto S, et al. Impact of cyto-
ticenter pilot for preemptive pharmacogenomics in electronic chrome P450 2C19 polymorphisms on the pharmacokinetics of
health record systems. Clin Pharmacol Ther. 2014;96(4):482–9. tacrolimus when coadministered with voriconazole. J Clin
137. Eadon MT, Desta Z, Levy KD, et al. Implementation of a Pharmacol. 2016;56(4):408–13.
pharmacogenomics consult service to support the INGENIOUS 154. Tod M, Nkoud-Mongo C, Gueyffier F. Impact of genetic poly-
trial. Clin Pharmacol Ther. 2016;100(1):63–6. morphism on drug-drug interactions mediated by cytochromes: a
138. Hoffman JM, Haidar CE, Wilkinson MR, et al. PG4KDS: a general approach. AAPS J. 2013;15(4):1242–52.
model for the clinical implementation of pre-emptive pharma- 155. Bolley R, Zulke C, Kammerl M, et al. Tacrolimus-induced
cogenetics. Am J Med Genet C Semin Med Genet. nephrotoxicity unmasked by induction of the CYP3A4 system
2014;166C(1):45–55. with St John’s wort. Transplantation. 2002;73(6):1009.
139. Bielinski SJ, Olson JE, Pathak J, et al. Preemptive genotyping 156. Vieira MD, Kim MJ, Apparaju S, et al. PBPK model describes
for personalized medicine: design of the right drug, right dose, the effects of comedication and genetic polymorphism on sys-
right time-using genomic data to individualize treatment proto- temic exposure of drugs that undergo multiple clearance path-
col. Mayo Clin Proc. 2014;89(1):25–33. ways. Clin Pharmacol Ther. 2014;95(5):550–7.
140. Hocum BT, White JR Jr, Heck JW, et al. Cytochrome P-450 157. Hill NR, Fatoba ST, Oke JL, et al. Global prevalence of chronic
gene and drug interaction analysis in patients referred for kidney disease—a systematic review and meta-analysis. PLoS
pharmacogenetic testing. Am J Health Syst Pharm. One. 2016;11(7):e0158765.
2016;73(2):61–7. 158. Webster AC, Nagler EV, Morton RL, et al. Chronic kidney
141. Bahar MA, Setiawan D, Hak E, et al. Pharmacogenetics of drug- disease. Lancet. 2017;389(10075):1238–52.
drug interaction and drug-drug-gene interaction: a systematic 159. Basile DP, Anderson MD, Sutton TA. Pathophysiology of acute
review on CYP2C9, CYP2C19 and CYP2D6. Pharmacoge- kidney injury. Compr Physiol. 2012;2(2):1303–53.
nomics. 2017;18(7):701–39. 160. Miners JO, Yang X, Knights KM, et al. The role of the kidney in
142. Venkatakrishnan K, Obach RS. In vitro-in vivo extrapolation of drug elimination: transport, metabolism, and the impact of
CYP2D6 inactivation by paroxetine: prediction of nonstationary kidney disease on drug clearance. Clin Pharmacol Ther.
pharmacokinetics and drug interaction magnitude. Drug Metab 2017;102(3):436–49.
Dispos. 2005;33(6):845–52. 161. Guevin C, Michaud J, Naud J, et al. Down-regulation of hepatic
143. Azuma J, Hasunuma T, Kubo M, et al. The relationship between cytochrome p450 in chronic renal failure: role of uremic
clinical pharmacokinetics of aripiprazole and CYP2D6 genetic mediators. Br J Pharmacol. 2002;137(7):1039–46.
polymorphism: effects of CYP enzyme inhibition by coadmin- 162. Rege B, Krieg R, Gao N, et al. Down-regulation of hepatic
istration of paroxetine or fluvoxamine. Eur J Clin Pharmacol. CYP3A in chronic renal insufficiency. Pharm Res.
2012;68(1):29–37. 2003;20(10):1600–6.
144. Lim KS, Cho JY, Jang IJ, et al. Pharmacokinetic interaction of 163. Michaud J, Dube P, Naud J, et al. Effects of serum from patients
flecainide and paroxetine in relation to the CYP2D6*10 allele in with chronic renal failure on rat hepatic cytochrome P450. Br J
healthy Korean subjects. Br J Clin Pharmacol. Pharmacol. 2005;144(8):1067–77.
2008;66(5):660–6. 164. Leblond FA, Giroux L, Villeneuve JP, et al. Decreased in vivo
145. Lee LS, Nafziger AN, Bertino JS Jr. Evaluation of inhibitory metabolism of drugs in chronic renal failure. Drug Metab Dis-
drug interactions during drug development: genetic polymor- pos. 2000;28(11):1317–20.
phisms must be considered. Clin Pharmacol Ther. 165. Velenosi TJ, Fu AY, Luo S, et al. Down-regulation of hepatic
2005;78(1):1–6. CYP3A and CYP2C mediated metabolism in rats with moderate
146. Damy T, Pousset F, Caplain H, et al. Pharmacokinetic and chronic kidney disease. Drug Metab Dispos.
pharmacodynamic interactions between metoprolol and drone- 2012;40(8):1508–14.
darone in extensive and poor CYP2D6 metabolizers healthy 166. Leblond FA, Petrucci M, Dube P, et al. Downregulation of
subjects. Fundam Clin Pharmacol. 2004;18(1):113–23. intestinal cytochrome p450 in chronic renal failure. J Am Soc
147. LLerena A, Berecz R, de la Rubia A, et al. Effect of thioridazine Nephrol. 2002;13(6):1579–85.
dosage on the debrisoquine hydroxylation phenotype in psy- 167. Dani M, Boisvert C, Michaud J, et al. Down-regulation of liver
chiatric patients with different CYP2D6 genotypes. Ther Drug drug-metabolizing enzymes in a murine model of chronic renal
Monit. 2001;23(6):616–20. failure. Drug Metab Dispos. 2010;38(3):357–60.
148. Kumar V, Brundage RC, Oetting WS, et al. Differential geno- 168. Dreisbach AW, Japa S, Gebrekal AB, et al. Cytochrome
type dependent inhibition of CYP2C9 in humans. Drug Metab P4502C9 activity in end-stage renal disease. Clin Pharmacol
Dispos. 2008;36(7):1242–8. Ther. 2003;73(5):475–7.
149. Andersson ML, Eliasson E, Lindh JD. A clinically significant 169. Dowling TC, Briglia AE, Fink JC, et al. Characterization of
interaction between warfarin and simvastatin is unique to car- hepatic cytochrome p4503A activity in patients with end-stage
riers of the CYP2C9*3 allele. Pharmacogenomics. renal disease. Clin Pharmacol Ther. 2003;73(5):427–34.
2012;13(7):757–62.
1292 F. Storelli et al.

170. Rowland Yeo K, Aarabi M, Jamei M, et al. Modeling and pre- 189. Steelandt J, Jean-Bart E, Goutelle S, et al. A prediction model of
dicting drug pharmacokinetics in patients with renal impairment. drug exposure in cirrhotic patients according to child-pugh
Expert Rev Clin Pharmacol. 2011;4(2):261–74. classification. Clin Pharmacokinet. 2015;54(12):1245–58.
171. Yeung CK, Shen DD, Thummel KE, et al. Effects of chronic 190. Johnson TN, Boussery K, Rowland-Yeo K, et al. A semi-
kidney disease and uremia on hepatic drug metabolism and mechanistic model to predict the effects of liver cirrhosis on
transport. Kidney Int. 2014;85(3):522–8. drug clearance. Clin Pharmacokinet. 2010;49(3):189–206.
172. US FDA. Guidance for Industry. Pharmacokinetics in patients 191. Wagner C, Zhao P, Arya V, et al. Physiologically based phar-
with impaired renal function—study design, data analysis, and macokinetic modeling for predicting the effect of intrinsic and
impact on dosing and labeling. 2010. http://www.fda.gov/ extrinsic factors on darunavir or lopinavir exposure coadminis-
downloads/drugs/guidances/ucm204959.pdf. Accessed 19 Sep tered with ritonavir. J Clin Pharmacol. 2017;57(10):1295–304.
2017. 192. Coutant DE, Kulanthaivel P, Turner PK, et al. Understanding
173. European Medicines Agency. Guideline on the evaluatin of the disease-drug interactions in cancer patients: implications for
pharmacokinetics of medicinal products in patients with dosing within the therapeutic window. Clin Pharmacol Ther.
decreased renal function. 2014. http://www.ema.europa.eu/docs/ 2015;98(1):76–86.
en_GB/document_library/Scientific_guideline/2014/02/ 193. Xu Y, Hijazi Y, Wolf A, et al. Physiologically based pharma-
WC500162133.pdf. Accessed 13 Sep 2017. cokinetic model to assess the influence of blinatumomab-me-
174. Nolin TD, Naud J, Leblond FA, et al. Emerging evidence of the diated cytokine elevations on cytochrome P450 enzyme activity.
impact of kidney disease on drug metabolism and transport. Clin CPT Pharmacom Syst Pharmacol. 2015;4(9):507–15.
Pharmacol Ther. 2008;83(6):898–903. 194. Morgan ET, Goralski KB, Piquette-Miller M, et al. Regulation
175. Zocor Drug Information. MSD Merck Sharp & Dohme. http:// of drug-metabolizing enzymes and transporters in infection,
www.swissmedicinfo.ch/default.aspx. Accessed 13 Sep 2017. inflammation, and cancer. Drug Metab Dispos.
176. Silva M, Matthews ML, Jarvis C, et al. Meta-analysis of drug- 2008;36(2):205–16.
induced adverse events associated with intensive-dose statin 195. Klein M, Thomas M, Hofmann U, et al. A systematic compar-
therapy. Clin Ther. 2007;29(2):253–60. ison of the impact of inflammatory signaling on absorption,
177. Zhao P, Vieira Mde L, Grillo JA, et al. Evaluation of exposure distribution, metabolism, and excretion gene expression and
change of nonrenally eliminated drugs in patients with chronic activity in primary human hepatocytes and HepaRG cells. Drug
kidney disease using physiologically based pharmacokinetic Metab Dispos. 2015;43(2):273–83.
modeling and simulation. J Clin Pharmacol. 2012;52(1 196. Rubin K, Janefeldt A, Andersson L, et al. HepaRG cells as
Suppl):91S–108S. human-relevant in vitro model to study the effects of inflam-
178. Verbeeck RK. Pharmacokinetics and dosage adjustment in matory stimuli on cytochrome P450 isoenzymes. Drug Metab
patients with hepatic dysfunction. Eur J Clin Pharmacol. Dispos. 2015;43(1):119–25.
2008;64(12):1147–61. 197. Dickmann LJ, Patel SK, Wienkers LC, et al. Effects of inter-
179. Edginton AN, Willmann S. Physiology-based simulations of a leukin 1beta (IL-1beta) and IL-1beta/interleukin 6 (IL-6) com-
pathological condition: prediction of pharmacokinetics in binations on drug metabolizing enzymes in human hepatocyte
patients with liver cirrhosis. Clin Pharmacokinet. culture. Curr Drug Metab. 2012;13(7):930–7.
2008;47(11):743–52. 198. Mimura H, Kobayashi K, Xu L, et al. Effects of cytokines on
180. Gines P, Guevara M, Arroyo V, et al. Hepatorenal syndrome. CYP3A4 expression and reversal of the effects by anti-cytokine
Lancet. 2003;362(9398):1819–27. agents in the three-dimensionally cultured human hepatoma cell
181. George J, Murray M, Byth K, et al. Differential alterations of line FLC-4. Drug Metab Pharmacokinet. 2015;30(1):105–10.
cytochrome P450 proteins in livers from patients with severe 199. Dickmann LJ, Patel SK, Rock DA, et al. Effects of interleukin-6
chronic liver disease. Hepatology. 1995;21(1):120–8. (IL-6) and an anti-IL-6 monoclonal antibody on drug-metabo-
182. Palatini P, De Martin S. Pharmacokinetic drug interactions in lizing enzymes in human hepatocyte culture. Drug Metab Dis-
liver disease: an update. World J Gastroenterol. pos. 2011;39(8):1415–22.
2016;22(3):1260–78. 200. Yang J, Hao C, Yang D, et al. Pregnane X receptor is required
183. Orlando R, Piccoli P, De Martin S, et al. Cytochrome P450 1A2 for interleukin-6-mediated down-regulation of cytochrome P450
is a major determinant of lidocaine metabolism in vivo: effects 3A4 in human hepatocytes. Toxicol Lett. 2010;197(3):219–26.
of liver function. Clin Pharmacol Ther. 2004;75(1):80–8. 201. Harvey RD, Morgan ET. Cancer, inflammation, and therapy:
184. Orlando R, Padrini R, Perazzi M, et al. Liver dysfunction effects on cytochrome p450-mediated drug metabolism and
markedly decreases the inhibition of cytochrome P450 1A2- implications for novel immunotherapeutic agents. Clin Phar-
mediated theophylline metabolism by fluvoxamine. Clin Phar- macol Ther. 2014;96(4):449–57.
macol Ther. 2006;79(5):489–99. 202. Schmitt C, Kuhn B, Zhang X, et al. Disease-drug-drug interac-
185. Floreani M, De Martin S, Gabbia D, et al. Severe liver cirrhosis tion involving tocilizumab and simvastatin in patients with
markedly reduces AhR-mediated induction of cytochrome P450 rheumatoid arthritis. Clin Pharmacol Ther. 2011;89(5):735–40.
in rats by decreasing the transcription of target genes. PLoS 203. Lee EB, Daskalakis N, Xu C, et al. Disease-drug interaction of
One. 2013;8(4):e61983. sarilumab and simvastatin in patients with rheumatoid arthritis.
186. Iyer R, Fetterly G, Lugade A, et al. Sorafenib: a clinical and Clin Pharmacokinet. 2017;56(6):607–15.
pharmacologic review. Expert Opin Pharmacother. 204. Zhuang Y, de Vries DE, Xu Z, et al. Evaluation of disease-
2010;11(11):1943–55. mediated therapeutic protein-drug interactions between an anti-
187. Lathia C, Lettieri J, Cihon F, et al. Lack of effect of keto- interleukin-6 monoclonal antibody (sirukumab) and cytochrome
conazole-mediated CYP3A inhibition on sorafenib clinical P450 activities in a phase 1 study in patients with rheumatoid
pharmacokinetics. Cancer Chemother Pharmacol. arthritis using a cocktail approach. J Clin Pharmacol.
2006;57(5):685–92. 2015;55(12):1386–94.
188. Ye L, Yang X, Guo E, et al. Sorafenib metabolism is signifi- 205. Jiang X, Zhuang Y, Xu Z, et al. Development of a physiologi-
cantly altered in the liver tumor tissue of hepatocellular carci- cally based pharmacokinetic model to predict disease-mediated
noma patient. PLoS One. 2014;9(5):e96664. therapeutic protein-drug interactions: modulation of multiple
Complex Drug–Drug–Gene–Disease Interactions 1293

cytochrome P450 enzymes by interleukin-6. AAPS J. 213. Bosilkovska M, Samer CF, Deglon J, et al. Geneva cocktail for
2016;18(3):767–76. cytochrome p450 and P-glycoprotein activity assessment using
206. Machavaram KK, Almond LM, Rostami-Hodjegan A, et al. A dried blood spots. Clin Pharmacol Ther. 2014;96(3):349–59.
physiologically based pharmacokinetic modeling approach to 214. Stanek EJ, Sanders CL, Taber KA, et al. Adoption of pharma-
predict disease-drug interactions: suppression of CYP3A by IL- cogenomic testing by US physicians: results of a nationwide
6. Clin Pharmacol Ther. 2013;94(2):260–8. survey. Clin Pharmacol Ther. 2012;91(3):450–8.
207. Muntane-Relat J, Ourlin JC, Domergue J, et al. Differential 215. O’Donnell PH, Danahey K, Jacobs M, et al. Adoption of a
effects of cytokines on the inducible expression of CYP1A1, clinical pharmacogenomics implementation program during
CYP1A2, and CYP3A4 in human hepatocytes in primary cul- outpatient care: initial results of the University of Chicago
ture. Hepatology. 1995;22(4 Pt 1):1143–53. ‘‘1,200 Patients Project’’. Am J Med Genet C Semin Med Genet.
208. Pascussi JM, Gerbal-Chaloin S, Pichard-Garcia L, et al. Inter- 2014;166C(1):68–75.
leukin-6 negatively regulates the expression of pregnane X 216. Fontana P, Cattaneo M, Combescure C, et al. Tailored
receptor and constitutively activated receptor in primary human thienopyridine therapy: no urgency for CYP2C19 genotyping.
hepatocytes. Biochem Biophys Res Commun. J Am Heart Assoc. 2013;2(2):e000131.
2000;274(3):707–13. 217. Reny JL, Fontana P, Hochholzer W, et al. Vascular risk levels
209. Missiakos O, Baysari MT, Day RO. Identifying effective com- affect the predictive value of platelet reactivity for the occur-
puterized strategies to prevent drug-drug interactions in hospital: rence of MACE in patients on clopidogrel. Systematic review
a user-centered approach. Int J Med Inform. and meta-analysis of individual patient data. Thromb Haemost.
2015;84(8):595–600. 2016;115(4):844–55.
210. Boussadi A, Caruba T, Karras A, et al. Validity of a clinical 218. Patel C, Rathi C, Venkatakrishnan K. Should race-genotype
decision rule-based alert system for drug dose adjustment in interactions be considered in the global development of
patients with renal failure intended to improve pharmacists’ CYP2C19 Substrates? A proposed framework using physiolog-
analysis of medication orders in hospitals. Int J Med Inform. ically based pharmacokinetic modeling. J Clin Pharmacol.
2013;82(10):964–72. 2017;57(4):417–21.
211. Zheng WY, Richardson LC, Li L, et al. Drug-drug interactions 219. Darwich AS, Ogungbenro K, Vinks AA, et al. Why has model-
and their harmful effects in hospitalised patients: a systematic informed precision dosing not yet become common clinical
review and meta-analysis. Eur J Clin Pharmacol. reality? lessons from the past and a roadmap for the future. Clin
2018;74(1):15–27. Pharmacol Ther. 2017;101(5):646–56.
212. Carli D. Impact des aides à la décision informatisée sur la
qualité des soins. Geneva: University of Geneva; 2016.

You might also like