Metabolic Drug Interactions With Immunosuppressants
Metabolic Drug Interactions With Immunosuppressants
Metabolic Drug Interactions With Immunosuppressants
74524
Provisional chapter
Chapter 20
Katalin Monostory
Katalin Monostory
Additional information is available at the end of the chapter
Additional information is available at the end of the chapter
http://dx.doi.org/10.5772/intechopen.74524
Abstract
Organ transplantation has become a routine clinical practice for patients with end-
stage disease of liver, kidney, heart, or lung. Although improved immunosuppressant
therapy substantially contributes to the success of transplantation, clinicians continue
to face challenges because of wide interindividual variations in blood concentrations
resulting in subtherapeutic or supratherapeutic levels. Many undesired side-effects or
therapeutic failure of immunosuppressants as a consequence are the results of differ-
ences or changes in drug metabolism. Considering genetic and nongenetic factors, such
as co-medication, can refine the immunosuppressant therapy, facilitating personalized
treatments to individual recipients. This review provides an up-to-date summary of
functional polymorphisms of enzymes involved in the metabolism of immunosuppres-
sants with low molecular weight and of the clinical significance of metabolic drug inter-
actions between immunosuppressive agents and other drugs in therapeutic regimens of
transplant recipients.
1. Introduction
Many undesired side-effects or therapeutic failures of drugs are the results of differences
or changes in drug metabolism. A patient’s drug metabolizing capacity, highly influenced
by genetic variations or alterations in the expression and activities of drug-metabolizing
enzymes, can substantially modify the pharmacokinetics of a drug and eventually its efficacy
or toxicity [1]. Even if the routine clinical practice applies blood concentration guided dosing,
© 2016 The Author(s). Licensee InTech. This chapter is distributed under the terms of the Creative Commons
© 2018 The Author(s). Licensee IntechOpen. This chapter is distributed under the terms of the Creative
Attribution License (http://creativecommons.org/licenses/by/3.0), which permits unrestricted use,
Commons Attribution License (http://creativecommons.org/licenses/by/3.0), which permits unrestricted use,
distribution, and reproduction in any medium, provided the original work is properly cited.
distribution, and reproduction in any medium, provided the original work is properly cited.
410 Organ Donation and Transplantation - Current Status and Future Challenges
the interindividual variability in drug metabolism calls for personalized medication primarily
for drugs with narrow therapeutic index [2, 3]. The identification of genetic and nongenetic
factors that can potentially affect the pharmacokinetics of a particular drug is a prerequisite
of tailored pharmacotherapy [4, 5].
CYP enzymes are the key players in the metabolism of most drugs; therefore, interindividual
and intraindividual variations in CYP activities are of significant importance in clinical practice.
The pharmacokinetic variability can divide the population into poor, intermediate, extensive,
and ultra-rapid metabolizer phenotypes. The loss-of-function mutations in CYP genes result
in permanent poor metabolism, whereas nongenetic (internal or environmental) factors can
substantially modify the expression and activities of CYP enzymes, evoking transient poor or
extensive/ultra-rapid metabolism [6, 7]. The clinical relevance for many CYP genetic variants,
regarding drug efficacy, adverse drug reactions, or dose requirement, has been clearly evi-
denced [6–9]; however, the heritable genetic polymorphisms are not the only determinant fac-
tors in interindividual differences in drug metabolism. CYP genotype determines the potential
for the expression of functional or nonfunctional enzymes; and nongenetic host factors (age,
sex, and disease states) and environmental factors (nutrition, medication, smoking, and alcohol
consumption) can alter the expression and activities of CYP enzymes [10]. Homozygous wild
genotype, predicted to be translated to functional CYP enzyme, can be transiently switched
into poor or extensive metabolizer phenotype, due to phenoconversion [1, 11]. Consequently,
both the CYP genotype and the current CYP expression or activity should be considered for the
estimation of a patient’s drug-metabolizing capacity.
Beside the genetic polymorphisms, one of the major sources of interindividual or intraindi-
vidual variability in drug metabolism is concomitant medication and co-morbidities, evoking
phenoconversion, notably CYP induction and enzyme inhibition [17]. CYP induction leads to
an increase in the expression and activity of CYP enzymes and contributes to the increased
elimination of drugs metabolized by the particular enzyme. Several pathways involving the
activation of various nuclear receptors (PXR pregnane X receptor, CAR constitutive andro-
stane receptor, and glucocorticoid receptor) have been reported to enhance the transcription
of CYP3A genes and to contribute to the complex regulation of CYP3A enzymes by drugs
such as rifampicin, phenobarbital, carbamazepine, and synthetic or natural steroids [18–21].
Reduced drug concentration as a consequence of CYP3A induction leads to the lack of the
pharmacological effect and drug failure. Phenoconversion converting genotypic extensive
metabolism into phenotypic poor metabolism of drugs may occur during inflammation (ster-
ile or infection-induced inflammation). Elevated release of proinflammatory cytokines (IL-6,
IL-1β, TNF-α) has been associated with downregulation of several drug-metabolizing CYPs,
including CYP3A enzymes. The mechanism of downregulation is the repression of PXR and
CAR that are involved in transcriptional regulation of CYP3A expression [22–26]. As a conse-
quence, transient poor metabolizer phenotype is developed, significantly increasing the risk
of adverse drug reactions and impacting the clinical outcome [1, 27]. Likewise, co-medication
can also give rise to poor metabolism. Several drugs or food components (e.g., bergamot-
tin) are known to inhibit the function of drug-metabolizing CYPs; therefore, the concomitant
treatment with a CYP inhibitor is expected to increase the exposure of those pharmacons
that are metabolized by the particular enzyme. As a consequence of CYP inhibition, the risk
of increased exposure and drug-induced adverse reactions can be anticipated, primarily for
drugs with narrow therapeutic index, such as tacrolimus and ciclosporin.
By recognizing individual differences in drug metabolism, personalized drug therapy
adjusted to the patient’s drug-metabolizing capacity can help to avoid the potential side
effects of drugs. The graft and recipient survival are highly influenced by drug-metabolizing
capacity of the liver, and it is essential to predict potential drug-drug interactions and to tailor
medication at both early and late postoperative periods.
3. Metabolism of immunosuppressants
In recent decades, transplantation (liver, kidney, heart, and lung) has become a routine pro-
cedure for patients with end stage disease. Advances in surgical techniques and postopera-
tive therapy have led to increasing numbers of transplantation and extended survival among
these patients. The final outcome of transplantation and the long-term graft function have
been improved mainly due to the development of potent and specific immunosuppressive
drugs. Immunosuppressants efficiently decrease the risk of rejection, blocking the recipient’s
immune system and protecting the transplanted organ. Because of the narrow therapeutic
indexes and increased risk of adverse drug reactions, it is essential to apply personalized
immunosuppressive therapy adjusted to patient’s drug-metabolizing capacity.
mTOR inhibitors:
Purine analogues:
Inosine monophosphate
dehydrogenase inhibitors:
Corticosteroids:
according to their molecular weights (agents with low or high molecular weights). High-
molecular-weight agents, such as polyclonal and monoclonal antibodies (e.g., thymoglobulin,
basiliximab, belatacept), that are not substrates for drug-metabolizing enzymes, are metabolized
in common protein degradation pathways (intracellular catabolism by endosomal-lysosomal
system) [28]; therefore, they are not subjects of metabolic drug interactions and not subjects of
this review. In the metabolism of immunosuppressants with low molecular weight, drug-metab-
olizing CYP enzymes are involved which may entail metabolic drug interactions (Table 1).
For solid organ transplant recipients, the mainstay of the immunosuppressive regimens is
calcineurin inhibitor (CNI) therapy with ciclosporin or tacrolimus which selectively blocks
several signaling processes, resulting in the inhibition of T-cell activation and proliferation
(Figure 1) [29, 30]. These drugs effectively treat allograft rejection; however, they display large
interindividual variability in their pharmacokinetics, requiring monitoring of blood concen-
trations for optimal safety and therapeutic efficacy.
Ciclosporin A is an 11-amino acid cyclopeptide that blocks the production of IL-2 by inhibi-
tion of calcineurin and, as a consequence, the activation of T-cells (Figure 1) [31]. Ciclosporin
undergoes extensive metabolism by CYP3A enzymes, producing more than 30 metabolites.
The major metabolic pathways are N-demethylation to 4-N-demethyl ciclosporin, hydroxyl-
ation at several positions (1-, 6-, 9-monohydroxy and 1,9- or 6,9-dihydroxy-metabolites), and
oxidation to carboxylic acid [32]. Some of the metabolites (e.g., 1,9-dihydroxy-ciclosporin,
Figure 1. Molecular action of calcineurin inhibitors and corticosteriods. AP-1 activator protein 1, CAR constitutive
androstane receptor, CSA ciclosporin, FKBP tarcolimus binding protein, GR glucocoticoid receptor, IL-2 interleukin 2,
JNK c-Jun N-terminal kinase, MAP3K mitogen-activated protein 3 kinase, MPK-1 mitogen-activated protein kinase 1,
NFAT nuclear factor of activated T-cells, PXR pregnane X receptor, Tacr tacrolimus.
414 Organ Donation and Transplantation - Current Status and Future Challenges
The mTOR inhibitors prevent cell proliferation by blocking cell cycle progression from the
G1-phase to the S-phase. The immunosuppressive activity is mediated via blocking mTOR
protein kinases, resulting in inhibition of growth factor–mediated T-cell proliferation in
response to IL-2 trigger [42]. Sirolimus is a 31-membered macrolide, whereas everolimus is
a sirolimus derivative having a 2-hydroxyethyl chain substitution at position 40. Although
the chemical structures of sirolimus and everolimus are similar to that of tacrolimus, the
mechanism of action of mTOR inhibitors is distinct from calcineurin inhibitors, which allows
the application of combination regimens. Additionally, the main advantages of mTOR inhibi-
tors are their nonnephrotoxic properties; therefore, mTOR inhibitors in combination with
reduced dose calcineurin inhibitors can augment the calcineurin inhibitor–induced nephro-
toxicity [43–45].
Metabolic Drug Interactions with Immunosuppressants 415
http://dx.doi.org/10.5772/intechopen.74524
The structural similarities can explain some common metabolic pathways of mTOR inhibitors
and tacrolimus, such as O-dealkylation and hydroxylation at several positions [42]. Sirolimus is
primarily metabolized by CYP3A enzymes and by CYP2C8 at lower extent, producing hydrox-
ylated and O-demethylated metabolites (e.g., 12-hydroxy-, 16-O-demethyl-, 39-O-demethyl-,
27–39-O-didemethyl- and dihydroxy-sirolimus as major metabolites) [46, 47]. The metabolism
of sirolimus leads to inactivation, despite the fact that some metabolites display some phar-
macological activity less than one tenth of the parent drug. Everolimus is also metabolized by
CYP3A and CYP2C8 enzymes; however, the elimination rate of everolimus is more rapid than
sirolimus (with 30 h vs. 62 h elimination half-lives, respectively). Everolimus is O-demethylated
and hydroxylated at several positions (forming both mono- and dihydroxy-metabolites); further-
more, a ring-opened metabolite is also formed from everolimus [46]. Everolimus-induced adverse
effects are associated with the exposure rather to the parent compound than to its metabolites.
One of the oldest agents with immunosuppressive activity introduced for kidney transplant
recipients was the purine analogue 6-mercaptopurine, which acts by inhibiting purine nucle-
otide synthesis and, as a consequence, cell proliferation. The prodrug of 6-mercaptopurine,
azathioprine with more favorable side-effect profile was later introduced to prevent rejection.
Azathioprine is converted to 6-mercaptopurine by nonenzymatic cleavage of the thioether in
enterocytes and hepatocytes or in erythrocytes. The major active metabolites, 6-thioguanine
nucleotides, are formed via 6-thioinosine monophosphate in natural purine synthetic pathways.
Inhibition of cell proliferation is mediated by incorporation of the thiopurine nucleotide ana-
logues into DNA (and RNA), causing DNA damage [48]. 6-Mercaptopurine, independently from
either direct administration or production from azathioprine, undergoes metabolic inactivation
by xanthine oxidase and thiopurine S-methyl transferase and is excreted in the urine, leaving less
parent compound available to form thiopurine nucleotides [49]. Due to genetic polymorphism,
the thiopurine S-methyl transferase activity is highly variable in patients; namely, those subjects
who carry one or two nonfunctional thiopurine S-methyl transferase alleles are unable to toler-
ate normal doses of azathioprine and can experience serious myelosuppression [50]. Therefore,
genotyping assay is recommended before starting azathioprine therapy to identify high-risk
patients, and dosage reduction or alternative therapy is recommended for these patients.
3.5. Corticosteroids
Calcineurin inhibitors are often applied in combination with mTOR inhibitors. Since both
mTOR inhibitors and calcineurin inhibitors are substrates of CYP3A enzymes and can inhibit
CYP3A activities, reduction of calcineurin inhibitor doses is recommended. Standard doses of
ciclosporin were observed to decrease the clearance of sirolimus or everolimus more substan-
tially than the doses of tacrolimus [45]. The major drawback of calcineurin inhibitor therapy is
the risk of nephrotoxicity which appears to be dose dependent. The combination of low calci-
neurin inhibitor doses with mTOR inhibitors was found to be beneficial regarding retaining low
rejection rates and lowering the risk of nephrotoxicity [44, 63]. To avoided renal dysfunction,
the complete substitution of calcineurin inhibitors for mTOR inhibitors was attempted; how-
ever, the substitution showed an increase in graft failure in patients treated with merely mTOR
inhibitors [64].
Corticosteroids have been demonstrated to induce the expression of the efflux pump trans-
porter ABCB1 (P-glycoprotein) playing a main role in intestinal drug absorption and of CYP3A
enzymes responsible for the metabolism of the majority of drugs [18, 65]. Therefore, the con-
comitant treatment of calcineurin inhibitors or mTOR inhibitors with corticosteroids can be
expected to decrease the blood concentrations of tacrolimus/ciclosporin or of sirolimus/evero-
limus. Although the evidence for clinically significant interactions between corticosteroids and
ciclosporin or mTOR inhibitors is limited, clear clinical effect of corticosteroids on tacrolimus
exposure has been demonstrated [66, 67]. This also implies that dose reduction or cessation
of corticosteroids leads to an increase in blood concentrations of tacrolimus, requiring dose
418 Organ Donation and Transplantation - Current Status and Future Challenges
Antifungals:
Antibiotics:
Antiviral agents:
Lipid-lowering agents:
Antihypertensive agents:
Antidiabetic agents:
Psychopharmacons:
Herbs:
Antifungals:
Antibiotics:
Antiviral agents:
Antihypertensive agents:
Antidiabetic agents:
Psychopharmacons:
Herbs:
Prednisolone Antifungals:
Methylprednisolone
Antibiotics:
Antiviral agents:
adjustment [68]. Interestingly, CYP3A5 nonexpressers with CYP3A5*3/*3 genotype are more
susceptible to glucocorticoid induction than CYP3A5*1 carriers [69]; thus, more pronounced
increase in tacrolimus exposure can be expected in CYP3A5 nonexpressers after glucocorticoid
withdrawal.
Clinically significant interaction between mycophenolic acid, the active metabolite of myco-
phenolate mofetil, and ciclosporin has been reported [70]. The mycophenolate-glucuronide
metabolite eliminated into bile undergoes enterohepatic cycling because of intestinal bacterial
metabolism and reabsorption of mycophenolic acid. The enterohepatic circulation, contrib-
uting to overall pharmacokinetics of mycophenolic acid by 37% in human, is inhibited by
concomitant administration of ciclosporin but does not interfere with tacrolimus or sirolimus
[71, 72]. In ciclosporin-mycophenolate combination therapy, the reduced blood concentration
of mycophenolic acid is necessary to ameliorate by increasing dose of mycophenolate mofetil.
Furthermore, special attention on optimal dosing is required during switching ciclosporin-
mycophenolate to tacrolimus-mycophenolate therapy and vice versa.
primarily in the early postoperative period. Since fungal infections are a threatening cause
of morbidity and mortality, the antifungal prophylaxis is an important element of posttrans-
plant medication. The antifungal azole-derivatives are potent (some of them are very strong)
CYP3A inhibitors, predicting potential metabolic drug interactions with calcineurin inhibi-
tors, mTOR inhibitors, or corticosteroids. The most potent CYP3A inhibitor is ketoconazole,
able to increase blood concentrations (AUC) of ciclosporin (> 4-fold), tacrolimus (> 2-fold),
sirolimus (11-fold), everolimus (15-fold), and methylprednisolone (> 2-fold) [73, 74]. Because
of the substantial increase in blood concentrations of several immunosuppressants that can
be avoided by drastic reduction of immunosuppressant doses and because of other adverse
effects of ketoconazole, the concomitant medication is discouraged. Fluconazole, itraconazole,
and voriconazole are alternative regimens for antifungal therapy or prophylaxis; however, all
three drugs are azole derivatives and have the capability to inhibit CYP3A function, albeit
at a lower extent than ketoconazole [75–77]. Although the continuous immunosuppressant
monitoring is highly recommended and dose adjustment (reduction) is generally required,
the antifungal treatment with fluconazole, itraconazole, or voriconazole can be safely applied
except for voriconazole-sirolimus combination [78]. Because of an extreme (7-fold) increase
of sirolimus blood concentrations as a consequence of concomitant use of voriconazole, this
combination is contraindicated. Amphotericin B, the nonazole type antifungal agent, does
not influence CYP activities; therefore, no metabolic drug interactions can be expected in con-
comitant treatment with immunosuppressants. However, the widespread use of amphoteri-
cin B is limited because of its toxicity profile, primarily because its nephrotoxic side-effect can
contribute to the renal injury by ciclosporin or tacrolimus.
Organ transplant patients are at high risk for developing bacterial infections that occur in
20–40% of transplants. Potential sources of infection are from hospital and community expo-
sures, as well as from endogenous flora of patients. Among the antibiotics used for treatment
of infections, the macrolide erythromycin and clarithromycin have been reported to interact
with immunosuppressive agents. These macrolides are CYP3A substrates and bind to CYP3A4
enzymes, leading to a complex formation that completely inactivates CYP3A4 enzyme [79–82].
The in vitro findings were confirmed by clinical observations that blood concentrations of ciclo-
sporin/tacrolimus or sirolimus/everolimus increased as a consequences of concomitant treat-
ment with erythromycin or clarithromycin [73, 83–86]. Page et al. [87] and Mori et al. [88] have
reported some potential of azithromycin for drug interaction with ciclosporin and tacrolimus;
however, in vitro experiments demonstrated that azithromycin poorly interfere with CYP3A4
[89]. When concomitant therapy with these macrolides is necessary, blood concentrations of
calcineurin inhibitors or mTOR inhibitors should be carefully monitored, and the immunosup-
pressant doses should be adjusted. In contrast, the macrolide rifampicin is a potent CYP3A4
inducer and can activate PXR, resulting in a substantial increase in CYP3A4 expression [90].
The increased CYP3A4 activity consequently enhances the metabolism and elimination of cal-
cineurin inhibitors, mTOR inhibitors, and corticosteroids [91–93]. However, blood concentra-
tion–guided dose-adjustment of immunosuppressants should be applied carefully because
increased metabolism can evoke elevation of toxic metabolite formation (e.g., ciclosporin).
A significant cause of graft failure still remains viral infections, which are acquired as new
infection or reactivation of latent viruses. After transplantation, cytomegalovirus (CMV) is the
422 Organ Donation and Transplantation - Current Status and Future Challenges
most common viral infection in recipients, primarily in those CMV-seronegative patients who
were transplanted with graft from CMV-seropositive donors, resulting in viral reactivation.
For prophylaxis and treatment of CMV infection, aciclovir, ganciclovir, and valganciclovir
(the prodrug of ganciclovir) are generally applied. None of these antiviral drugs influences
the function of drug-metabolizing CYPs or UDP-glucuronyl transferases, and consequently,
they do not modify the pharmacokinetic properties of immunosuppressants. Aciclovir and
ganciclovir are eliminated primarily in the urine as unchanged compounds. Increased risk
of nephrotoxicity and leukopenia has been reported in patients who were co-medicated with
a drug that can reduce renal clearance of aciclovir or ganciclovir. During co-administration
with mycophenolate or mycophenolate-mofetil, mycophenolate-glucuronide and aciclovir or
ganciclovir can significantly compete for renal tubular secretion, resulting in an increase in
aciclovir/ganciclovir and mycophenolate-glucuronide exposure, as well as the risk of neph-
rotoxicity or leukopenia [94–96]. Management of potent metabolic drug interactions between
antiviral protease inhibitors and immunosuppressants is a major challenge because most of
the protease inhibitors are clinically significant CYP3A4 inhibitors. Ritonavir-boosted thera-
pies require substantial reduction of immunosuppressant doses (to 5–20% for ciclosporin; to
1–3.5% for tacrolimus) with continuous monitoring of blood concentrations [97–101].
mTOR inhibitors and corticosteroids, increasing the risk of rejection [133]. Immunosuppressant
dose adjustment is required to avoid subtherapeutic blood concentrations, and careful moni-
toring of immunosuppressant blood concentrations is recommended during withdrawal of
troglitazone or rosiglitazone and during switching to other antihyperglycemic agent.
The most common psychiatric disorders encountered in transplant patients are anxiety,
depression, mood disorders, behavior problems, and insomnia that are reversible in most
cases; however, they often require psychotherapy with antidepressants, mood stabilizers,
anxiolytic agents, or even with antipsychotics. Many of these pharmacons are metabolized
by enzymes other than CYP3A4 and do not influence the drug-metabolizing activities of
CYP3A4; consequently, metabolic drug interactions with immunosuppressants cannot be
expected. Nevertheless, the CYP3A4 inducing or inhibitory properties of some of these psy-
chopharmacons should be considered. The mood stabilizer carbamazepine and valproic acid
have been clearly evidenced to be able to activate CAR and PXR. The nuclear receptor acti-
vation leads to an increase in transcription of CYP3A4 gene and CYP3A4 metabolic activity
[134, 135], anticipating decrease of immunosuppressant blood concentrations [136]. To reduce
the risk of organ rejection, adjustment (increase) of immunosuppressant doses is required
with continuous monitoring of immunosuppressant blood levels. Furthermore, the CYP3A4
deinduction process can last for about 2 weeks after cessation of carbamazepine or valproic
acid [137]; thus, careful monitoring of immunosuppressant blood concentrations during with-
drawal is essential. The comedication with the antidepressant fluvoxamine is contraindicated
because of its strong inhibitory properties for CYP3A4 substrates and potential drug interac-
tions with ciclosporin/tacrolimus or with sirolimus/everolimus [80, 138, 139]. For psychother-
apeutic agents that are CYP3A substrates (haloperidol, quetiapine, clonazepam, midazolam,
alprazolam), continuous monitoring of immunosuppressant blood levels is highly recom-
mended to avoid metabolic drug interactions.
Pharmacokinetic herb-drug interactions can also significantly influence the outcome of immu-
nosuppressive therapy and long-term graft survival [145]. St John’s wort (Hypericum perfora-
tum) extract and grapefruit juice are well described as modifiers of pharmacokinetic properties
of ciclosporin and tacrolimus [146–148]. St John’s wort extract is a herbal product for treatment
of symptoms of mild or moderate depression, including anxiety, fatigue, and sleeping prob-
lems. The extract contains a number of biologically active components, e.g., hyperforin of high
interest. Hyperforin has a strong affinity for PXR and significantly increases the expression
and activities of CYP3A4 enzyme, which is involved in metabolism of many drugs [149, 150].
Consequently, chronic consumption of St John’s wort extract can decrease the blood concentra-
tions of CYP3A substrates, such as calcineurin inhibitors, mTOR inhibitors, and corticosteroids
[151–154]. In addition, St John’s wort extract has been reported to induce the expression of
ABCB1 transporter that reduces the absorption of ABCB1-ligand drugs from the gut. The hyper-
forin contents of commercially available St John’s wort preparations are variables that appear
to significantly affect the extent of pharmacokinetic interactions [150, 155]. Coadministration of
ciclosporin with St John’s wort extract has been reported to lead a 40–60% decrease of ciclospo-
rin blood concentrations, increasing the risk of rejection; therefore, substantial dose adjustment
is required [151, 152, 155–159]. Since clinicians are often unaware of concomitant consump-
tion of herbal supplements, transplant patients should be informed about the drug interaction
potential of St John’s wort that can endanger the success of organ transplantation.
Concomitant intake of grapefruit (Citrus paradisii) or pomelo (Citrus grandis) has been dem-
onstrated to increase the bioavailability of immunosuppressants [147, 160, 161]. Some com-
ponents of these citrus fruits, bergamottin and naringenin responsible for the bitter taste,
can inhibit the activities of CYP3A4 and CYP3A5 enzymes both in the intestinal wall and
in the liver, resulting in significant reduction of first-pass metabolism of CYP3A substrates,
including ciclosporin and tacrolimus [162–164]. Significant reduction of ciclosporin/tacroli-
mus doses is necessary to avoid the risk of nephrotoxicity or other adverse events associated
with immunosuppressive therapy. The furanocoumarin bergamottin is a “suicide substrate,”
namely it is metabolized by CYP3A4 to an epoxid metabolite that covalently binds to and
inactivates the enzyme [165]. The flavonoid naringenin was found to be a less-potent CYP3A4
inhibitor than bergamottin [166]; however, during consumption of grapefruit, the inhibitory
effects of naringenin and bergamottin are added together. Since clear evidence of bergamottin
content and CYP3A4 inhibitory potential of citruses other than grapefruit and pomelo was
provided [167], the transplantation centers do not recommend citrus consumption for trans-
plant patients during immunosuppressive therapy.
5. Concluding remarks
Acknowledgements
This work was supported by MedInProt Synergy VIII program of the Hungarian Academy of
Sciences and by the Society of Hungarian Toxicologists.
Conflict of interest
Author details
Katalin Monostory
References
[1] Shah RR, Smith RL. Addressing phenoconversion: The Achilles' heel of personalized
medicine. British Journal of Clinical Pharmacology. 2015;79:222-240. DOI: 10.1111/bcp.
12441
[3] Gervasini G, Benítez J, Carrillo JA. Pharmacogenetic testing and therapeutic drug moni-
toring are complementary tools for optimal individualization of drug therapy. European
Journal of Clinical Pharmacology. 2010;66:755-774. DOI: 10.1007/s00228-010-0857-7
[4] Shah RR, Shah DR. Personalized medicine: Is it a pharmacogenetic mirage? British
Journal of Clinical Pharmacology. 2012;74:698-721. DOI: 10.1111/j.1365-2125.2012.04328.x
[6] Zhou SF, Liu JP, Chowbay B. Polymorphism of human cytochrome P450 enzymes and
its clinical impact. Drug Metabolism Reviews. 2009;41:89-295. DOI: 10.1021/tx0100439
[7] Zanger UM, Schwab M. Cytochrome P450 enzymes in drug metabolism: Regulation of
gene expression, enzyme activities, and impact of genetic variation. Pharmacology &
Therapeutics. 2013;138:103-141. DOI: 10.1016/j.pharmthera.2012.12.007
[8] Samer CF, Lorenzini KI, Rollason V, Daali Y, Desmeules JA. Applications of CYP450
testing in the clinical setting. Molecular Diagnosis & Therapy. 2013;17:165-184. DOI:
10.1007/s40291-013-0028-5
[9] Zhou ZW, Chen XW, Sneed KB, Yang YX, Zhang X, He ZX, Chow K, Yang T, Duan W,
Zhou SF. Clinical association between pharmacogenomics and adverse drug reactions.
Drugs. 2015;75:589-631. DOI: 10.1007/s40265-015-0375-0
[10] Rendic S, Guengerich FP. Update information on drug metabolism systems - 2009, part
II: Summary of information on the effects of diseases and environmental factors on
human cytochrome P450 (CYP) enzymes and transporters. Current Drug Metabolism.
2010;11:4-84. DOI: 10.2174/138920010791110917
[11] Shah RR, Gaedigk A, LLerena A, Eichelbaum M, Stingl J, Smith RL. CYP450 genotype
and pharmacogenetic association studies: A critical appraisal. Pharmacogenomics.
2016;17:259-275. DOI: 10.2217/pgs.15.172
[14] Amirimani B, Ning B, Deitz AC, Weber BL, Kadlubar FF, Rebbeck TR. Increased tran-
scriptional activity of the CYP3A4*1B promoter variant. Environmental and Molecular
Mutagenesis. 2003;42:299-305. DOI: 10.1002/em.10199
[17] Zhou SF. Drugs behave as substrates, inhibitors and inducers of human cytochrome
P450 3A4. Current Drug Metabolism. 2008;9:310-322. DOI: 10.2174/138920008784220664
[19] Hukkanen J. Induction of cytochrome P450 enzymes: A view on human in vivo findings.
Expert Review of Clinical Pharmacology. 2012;5:569-585. DOI: 10.1586/ecp.12.39
[21] Matoulková P, Pávek P, Malý J, Vlček J. Cytochrome P450 enzyme regulation by glu-
cocorticoids and consequences in terms of drug interaction. Expert Opinion on Drug
Metabolism & Toxicology. 2014;10:425-435. DOI: 10.1517/17425255.2014.878703
[23] Dickmann LJ, Patel SK, Rock DA, Wienkers LC, Slatter JG. Effects of interleukin-6 (IL-6)
and an anti-IL-6 monoclonal antibody on drug-metabolizing enzymes in human hepa-
tocyte culture. Drug Metabolism and Disposition. 2011;39:1415-1422. DOI: 10.1124/dmd.
111.038679
[24] Dickmann LJ, Patel SK, Wienkers LC, Slatter JG. Effects of interleukin 1β (IL-1β) and IL-1β/
interleukin 6 (IL-6) combinations on drug metabolizing enzymes in human hepatocyte
culture. Current Drug Metabolism. 2012;13:930-937. DOI: 10.2174/138920012802138642
Metabolic Drug Interactions with Immunosuppressants 429
http://dx.doi.org/10.5772/intechopen.74524
[29] Penninga L, Wettergren A, Chan AW, Steinbrüchel DA, Gluud C. Calcineurin inhibi-
tor minimisation versus continuation of calcineurin inhibitor treatment for liver trans-
plant recipients. Cochrane Database of Systematic Reviews. 2012;3:CD008852. DOI: 10.
1002/14651858.CD008852.pub2
[30] Choudhary NS, Saigal S, Shukla R, Kotecha H, Saraf N, Soin AS. Current status of immu-
nosuppression in liver transplantation. Journal of Clinical and Experimental Hepatology.
2013;3:150-158. DOI: 10.1016/j.jceh.2013.04.005
[31] Azzi JR, Sayegh MH, Mallat SG. Calcineurin inhibitors: 40 years later, can't live without.
Journal of Immunology. 2013;191:5785-5791. DOI: 10.4049/jimmunol.1390055
[33] Christians U, Kohlhaw K, Budniak J, Bleck JS, Schottmann R, Schlitt HJ, Almeida VM,
Deters M, Wonigeit K, Pichlmayr R, Sewing KF. Ciclosporin metabolite pattern in blood
and urine of liver graft recipients. I. Association of ciclosporin metabolites with nephro-
toxicity. European Journal of Clinical Pharmacology. 1991;41:285-290
[34] Christians U, Kohlhaw K, Sürig T, Bader A, Schottmann R, Linck A, Ringe B, Sewing KF.
Parallel blood concentrations of second-generation cyclosporine metabolites and bili-
rubin in liver graft recipients. Therapeutic Drug Monitoring. 1995;17:487-498. DOI: 10.
1097/00007691-199510000-00009
[35] Monostory K, Tóth K, Kiss Á, Háfra E, Csikány N, Paulik J, Sárváry E, Kóbori L.
Personalizing calcineurin inhibitor therapy by adjusting to donor CYP3A-status in liver
transplant patients. British Journal of Clinical Pharmacology. 2015;80:1429-1437. DOI:
10.1111/bcp.12747
[36] Iwasaki K. Metabolism of tacrolimus (FK506) and recent topics in clinical pharmacokinet-
ics. Drug Metabolism and Pharmacokinetics. 2007;22:328-335. DOI: 10.2133/dmpk.22.328
430 Organ Donation and Transplantation - Current Status and Future Challenges
[51] Staatz CE, Tett SE. Pharmacology and toxicology of mycophenolate in organ transplant
recipients: An update. Archives of Toxicology. 2014;88:1352-1389. DOI: 10.1007/s00204-
014-1247-1
[54] Lamba V, Sangkuhl K, Sanghavi K, Fish A, Altman RB, Klein TE. PharmGKB summary:
Mycophenolic acid pathway. Pharmacogenetics and Genomics. 2014;24:73-79. DOI: 10.
1097/FPC.0000000000000010
[58] Wong TC, Lo CM, Fung JY. Emerging drugs for prevention of T-cell mediated rejec-
tion in liver and kidney transplantation. Expert Opinion on Emerging Drugs. 2017;22:
123-136. DOI: 10.1080/14728214.2017.1330884
[60] Ling SY, Huizinga RB, Mayo PR, Larouche R, Freitag DG, Aspeslet LJ, Foster RT. Cyto
chrome P450 3A and P-glycoprotein drug-drug interactions with voclosporin. British
Journal of Clinical Pharmacology. 2014;77:1039-1050. DOI: 10.1111/bcp.12309
432 Organ Donation and Transplantation - Current Status and Future Challenges
[63] van Gelder T, Fisher L, Shihab F, Shipkova M. Optimizing everolimus exposure when
combined with calcineurin inhibitors in solid organ transplantation. Transplantation
Reviews (Orlando, Fla.). 2017;31:151-157. DOI: 10.1016/j.trre.2017.02.007
[65] Martin P, Riley R, Back DJ, Owen A. Comparison of the induction profile for drug dis-
position proteins by typical nuclear receptor activators in human hepatic and intestinal
cells. British Journal of Pharmacology. 2008;153:805-819. DOI: 10.1038/sj.bjp.0707601
[66] Hesselink DA, Ngyuen H, Wabbijn M, Gregoor PJ, Steyerberg EW, van Riemsdijk IC,
Weimar W, van Gelder T. Tacrolimus dose requirement in renal transplant recipients is
significantly higher when used in combination with corticosteroids. British Journal of
Clinical Pharmacology. 2003;56:327-330. DOI: 10.1046/j.0306-5251.2003.01882.x
[68] van Duijnhoven EM, Boots JM, Christiaans MH, Stolk LM, Undre NA, van Hooff JP.
Increase in tacrolimus trough levels after steroid withdrawal. Transplant International.
2003;16:721-725. DOI: 10.1007/s00147-003-0615-1
[69] Roberts PJ, Rollins KD, Kashuba AD, Paine MF, Nelsen AC, Williams EE, Moran C,
Lamba JK, Schuetz EG, Hawke RL. The influence of CYP3A5 genotype on dexamethasone
induction of CYP3A activity in African Americans. Drug Metabolism and Disposition.
2008;36:1465-1469. DOI: 10.1124/dmd.107.020065
[72] Dalal P, Shah G, Chhabra D, Gallon L. Role of tacrolimus combination therapy with
mycophenolate mofetil in the prevention of organ rejection in kidney transplant patients.
International Journal of Nephrology and Renovascular Disease. 2010;3:107-115. DOI:
10.2147/IJNRD.S7044
[73] Kovarik JM, Beyer D, Schmouder RL. Everolimus drug interactions: Application of a
classification system for clinical decision making. Biopharmaceutics & Drug Disposition.
2006;27:421-426. DOI: 10.1002/bdd.524
[74] Lu C, Hatsis P, Berg C, Lee FW, Balani SK. Prediction of pharmacokinetic drug-drug
interactions using human hepatocyte suspension in plasma and cytochrome P450 phe-
notypic data. II. In vitro-in vivo correlation with ketoconazole. Drug Metabolism and
Disposition. 2008;36:1255-1260. DOI: 10.1124/dmd.107.018796
[75] Lu C, Berg C, Prakash SR, Lee FW, Balani SK. Prediction of pharmacokinetic drug-drug
interactions using human hepatocyte suspension in plasma and cytochrome P450 phe-
notypic data. III. In vitro-in vivo correlation with fluconazole. Drug Metabolism and
Disposition. 2008;36:1261-1266. DOI: 10.1124/dmd.107.019000
[77] Zhang S, Pillai VC, Mada SR, Strom S, Venkataramanan R. Effect of voriconazole and
other azole antifungal agents on CYP3A activity and metabolism of tacrolimus in human
liver microsomes. Xenobiotica. 2012;42:409-416. DOI: 10.3109/00498254.2011.631224
[78] Saad AH, DePestel DD, Carver PL. Factors influencing the magnitude and clinical signif-
icance of drug interactions between azole antifungals and select immunosuppressants.
Pharmacotherapy. 2006;26:1730-1744. DOI: 10.1592/phco.26.12.1730
[79] Galetin A, Burt H, Gibbons L, Houston JB. Prediction of time-dependent CYP3A4 drug-drug
interactions: Impact of enzyme degradation, parallel elimination pathways, and intestinal
inhibition. Drug Metabolism and Disposition. 2006;34:166-175. DOI: 10.1124/dmd/105.006874
[80] Watanabe A, Nakamura K, Okudaira N, Okazaki O, Sudo K. Risk assessment for drug-
drug interaction caused by metabolism-based inhibition of CYP3A using automated
in vitro assay systems and its application in the early drug discovery process. Drug
Metabolism and Disposition. 2007;35:1232-1238. DOI: 10.1124/dmd.107.015016
[82] Kosaka M, Kosugi Y, Hirabayashi H. Risk assessment using cytochrome P450 time-depen-
dent inhibition assays at single time and concentration in the early stage of drug discovery.
Journal of Pharmaceutical Sciences. 2017;106:2839-2846. DOI: 10.1016/j.xphs.2017.04.077
[83] Westphal JF. Macrolide - induced clinically relevant drug interactions with cytochrome P-450A
(CYP) 3A4: An update focused on clarithromycin, azithromycin and dirithromycin. British
Journal of Clinical Pharmacology. 2000;50:285-295. DOI: 10.1046/j.1365-2125.2000.00261.x
434 Organ Donation and Transplantation - Current Status and Future Challenges
[86] Pea F, Cojutti P, Tursi V, Livi U, Baraldo M. Everolimus overexposure in a heart transplant
patient receiving clarithromycin for the treatment of pneumonia. Transplant Infectious
Disease. 2015;17:926-928. DOI: 10.1111/tid.12446
[87] Page RL 2nd, Ruscin JM, Fish D, Lapointe M. Possible interaction between intravenous
azithromycin and oral cyclosporine. Pharmacotherapy. 2001;21:1436-1443. DOI: 10.1592/
phco.21.17.1436.34434
[91] Hebert MF, Fisher RM, Marsh CL, Dressler D, Bekersky I. Effects of rifampin on tacro-
limus pharmacokinetics in healthy volunteers. Journal of Clinical Pharmacology.
1999;39:91-96. DOI: 10.1177/00912709922007499
[92] Bhaloo S, Prasad GV. Severe reduction in tacrolimus levels with rifampin despite mul-
tiple cytochrome P450 inhibitors: A case report. Transplantation Proceedings. 2003;35:
2449-2451. DOI: 10.1016/j.transproceed.2003.08.019
[93] Lee YT, Hwang S, Lee SG, Kim KW, Choi NK, Park GC, Yu YD, Yoo JW, Kim WS,
Shim TS. Living-donor liver transplantation in patients with concurrent active tuber-
culosis at transplantation. The International Journal of Tuberculosis and Lung Disease.
2010;14:1039-1044
[95] Brum S, Nolasco F, Sousa J, Ferreira A, Possante M, Pinto JR, Barroso E, Santos JR.
Leukopenia in kidney transplant patients with the association of valganciclovir and
mycophenolate mofetil. Transplantation Proceedings. 2008;40:752-754. DOI: 10.1016/j.
transproceed.2008.02.048
Metabolic Drug Interactions with Immunosuppressants 435
http://dx.doi.org/10.5772/intechopen.74524
[97] Sheikh AM, Wolf DC, Lebovics E, Goldberg R, Horowitz HW. Concomitant human immu-
nodeficiency virus protease inhibitor therapy markedly reduces tacrolimus metabolism
and increases blood levels. Transplantation. 1999;68:307-309. DOI: 10.1097/00007890-
199907270-00027
[98] Vogel M, Voigt E, Michaelis HC, Sudhop T, Wolff M, Türler A, Sauerbruch T, Rockstroh JK,
Spengler U. Management of drug-to-drug interactions between cyclosporine a and the
protease-inhibitor lopinavir/ritonavir in liver-transplanted HIV-infected patients. Liver
Transplantation. 2004;10:939-944. DOI: 10.1002/lt.20165
[102] Mathis AS, Davé N, Knipp GT, Friedman GS. Drug-related dyslipidemia after renal
transplantation. American Journal of Health-System Pharmacy. 2004;61:565-585
[104] Asberg A. Interactions between cyclosporin and lipid-lowering drugs: Implications for
organ transplant recipients. Drugs. 2003;63:367-378. DOI: 10.2165/00003495-200363040-
00003
[105] Neuvonen PJ, Backman JT, Niemi M. Pharmacokinetic comparison of the potential
over-the-counter statins simvastatin, lovastatin, fluvastatin and pravastatin. Clinical
Pharmacokinetics. 2008;47:463-474. DOI: 10.2165/00003088-200847070-00003
[107] Gertz M, Cartwright CM, Hobbs MJ, Kenworthy KE, Rowland M, Houston JB, Galetin A.
Cyclosporine inhibition of hepatic and intestinal CYP3A4, uptake and efflux transport-
ers: Application of PBPK modeling in the assessment of drug-drug interaction poten-
tial. Pharmaceutical Research. 2013;30:761-780. DOI: 10.1007/s11095-012-0918-y
436 Organ Donation and Transplantation - Current Status and Future Challenges
[108] Christians U, Jacobsen W, Benet LZ, Lampen A. Mechanisms of clinically relevant drug
interactions associated with tacrolimus. Clinical Pharmacokinetics. 2002;41:813-851.
DOI: 10.2165/00003088-200241110-00003
[109] Lemahieu WP, Hermann M, Asberg A, Verbeke K, Holdaas H, Vanrenterghem Y,
Maes BD. Combined therapy with atorvastatin and calcineurin inhibitors: No inter-
actions with tacrolimus. American Journal of Transplantation. 2005;5:2236-2243. DOI:
10.1111/j.1600-6143.2005.01005.x
[110] Ma B, Prueksaritanont T, Lin JH. Drug interactions with calcium channel blockers:
Possible involvement of metabolite-intermediate complexation with CYP3A. Drug
Metabolism and Disposition. 2000;28:125-130
[111] Wang YH, Jones DR, Hall SD. Prediction of cytochrome P450 3A inhibition by verapamil
enantiomers and their metabolites. Drug Metabolism and Disposition. 2004;32:259-266.
DOI: 10.1124/dmd.32.2.259
[112] Wang YH, Jones DR, Hall SD. Differential mechanism-based inhibition of CYP3A4
and CYP3A5 by verapamil. Drug Metabolism and Disposition. 2005;33:664-671. DOI:
10.1124/dmd/104.001834
[113] Zhao P, Lee CA, Kunze KL. Sequential metabolism is responsible for diltiazem-induced
time-dependent loss of CYP3A. Drug Metabolism and Disposition. 2007;35:704-712.
DOI: 10.1124/dmd.106.013847
[115] Crocker JF, Renton KW, LeVatte TL, McLellan DH. The interaction of the calcium chan-
nel blockers verapamil and nifedipine with cyclosporin a in pediatric renal transplant
patients. Pediatric Nephrology. 1994;8:408-411. DOI: 10.1007/BF00856514
[116] Campana C, Regazzi MB, Buggia I, Molinaro M. Clinically significant drug interac-
tions with cyclosporin. An update. Clinical Pharmacokinetics. 1996;30:141-179. DOI:
10.2165/00003088-199630020-00004
[117] Yildiz A, Sever MS, Türkmen A, Ecder T, Türk S, Akkaya V, Ark E. Interaction between
cyclosporine a and verapamil, felodipine, and isradipine. Nephron. 1999;81:117-118
[118] Hebert MF, Lam AY. Diltiazem increases tacrolimus concentrations. The Annals of
Pharmacotherapy. 1999;33:680-682. DOI: 10.1345/aph.18356
[119] Böttiger Y, Säwe J, Brattström C, Tollemar J, Burke JT, Häss G, Zimmerman JJ. Pharma
cokinetic interaction between single oral doses of diltiazem and sirolimus in healthy
volunteers. Clinical Pharmacology and Therapeutics. 2001;69:32-40. DOI: 10.1067/mcp.
2001.112513
[120] Jones TE, Morris RG. Pharmacokinetic interaction between tacrolimus and diltiazem:
Dose-response relationship in kidney and liver transplant recipients. Clinical Pharma
cokinetics. 2002;41:381-388
Metabolic Drug Interactions with Immunosuppressants 437
http://dx.doi.org/10.5772/intechopen.74524
[121] Kovarik JM, Beyer D, Bizot MN, Jiang Q, Allison MJ, Schmouder RL. Pharmacokinetic
interaction between verapamil and everolimus in healthy subjects. British Journal of
Clinical Pharmacology. 2005;60:434-437. DOI: 10.1111/j.1365-2125.2005.02434.x
[124] Zuo XC, Zhou YN, Zhang BK, Yang GP, Cheng ZN, Yuan H, Ouyang DS, Liu SK,
Barrett JS, Li PJ, Liu Z, Tan HY, Guo R, Zhou LY, Xie YL, Li ZJ, Li J, Wang CJ, Wang JL.
Effect of CYP3A5*3 polymorphism on pharmacokinetic drug interaction between tacro-
limus and amlodipine. Drug Metabolism and Pharmacokinetics. 2013;28:398-405. DOI:
10.2133/dmpk.DMPK-12-RG-148
[126] Bader FM, Hagan ME, Crompton JA, Gilbert EM. The effect of beta-blocker use on
cyclosporine level in cardiac transplant recipients. The Journal of Heart and Lung
Transplantation. 2005;24:2144-2147. DOI: 10.1016/j.healun.2005.05.002
[127] Oldham HG, Clarke SE. Vitro identification of the human cytochrome P450 enzymes
involved in the metabolism of R(+)- and S(−)-carvedilol. Drug Metabolism and Dispo
sition. 1997;25:970-977
[129] Ramachandran V, Kostrubsky VE, Komoroski BJ, Zhang S, Dorko K, Esplen JE, Strom SC,
Venkataramanan R. Troglitazone increases cytochrome P-450 3A protein and activity
in primary cultures of human hepatocytes. Drug Metabolism and Disposition. 1999;
27:1194-1199
[130] Sahi J, Black CB, Hamilton GA, Zheng X, Jolley S, Rose KA, Gilbert D, LeCluyse EL, Sinz
MW. Comparative effects of thiazolidinediones on in vitro P450 enzyme induction and
inhibition. Drug Metabolism and Disposition. 2003;31:439-446. DOI: 10.1124/dmd.31.4.439
[134] Oscarson M, Zanger UM, Rifki OF, Klein K, Eichelbaum M, Meyer UA. Transcriptional
profiling of genes induced in the livers of patients treated with carbamazepine. Clinical
Pharmacology and Therapeutics. 2006;80:440-456. DOI: 10.1016/j.clpt.2006.08.013
[136] Cooney GF, Mochon M, Kaiser B, Dunn SP, Goldsmith B. Effects of carbamazepine
on cyclosporine metabolism in pediatric renal transplant recipients. Pharmacotherapy.
1995;15:353-356. DOI: 10.1002/j.1875-9114.1995.tb04373.x
[137] Punyawudho B, Cloyd JC, Leppik IE, Ramsay RE, Marino SE, Pennell PB, White JR,
Birnbaum AK. Characterization of the time course of carbamazepine deinduction by
an enzyme turnover model. Clinical Pharmacokinetics. 2009;48:313-320. DOI: 10.2165/
00003088-200948050-00003
[138] Kashuba AD, Nafziger AN, Kearns GL, Leeder JS, Gotschall R, Rocci ML Jr, Kulawy RW,
Beck DJ, Bertino JS Jr. Effect of fluvoxamine therapy on the activities of CYP1A2,
CYP2D6, and CYP3A as determined by phenotyping. Clinical Pharmacology and Ther
apeutics. 1998;64:257-268. DOI: 10.1016/S0009-9236(98)90174-6
[139] Jones DR, Ekins S, Li L, Hall SD. Computational approaches that predict metabolic inter-
mediate complex formation with CYP3A4 (+b5). Drug Metabolism and Disposition.
2007;35:1466-1475. DOI: 10.1124/dmd.106.014613
[140] Cummins D, Sekar M, Halil O, Banner N. Myelosuppression associated with azathi-
oprine-allopurinol interaction after heart and lung transplantation. Transplantation.
1996;61:1661-1662. DOI: 10.1097/00007890-199606150-00023
[141] Weiler S, Aellig N, Fauchère I, Jetter A, Corti N. Treatment of gout in a renal trans-
plant patient leading to severe thrombocytopenia. Journal of Clinical Pharmacy and
Therapeutics. 2014;39:571-572. DOI: 10.1111/jcpt.12190
[143] Trück J, Laube GF, von Vigier RO, Goetschel P. Gout in pediatric renal transplant recip-
ients. Pediatric Nephrology. 2010;25:2535-2538. DOI: 10.1007/s00467-010-1599-6
[144] Stamp LK, Chapman PT. Gout and organ transplantation. Current Rheumatology
Reports. 2012;14:165-172. DOI: 10.1007/s11926-012-0235-9
Metabolic Drug Interactions with Immunosuppressants 439
http://dx.doi.org/10.5772/intechopen.74524
[158] Alscher DM, Klotz U. Drug interaction of herbal tea containing St. John's wort with
cyclosporine. Transplant International. 2003;16:543-544. DOI: 10.1007/s00147-003-0560-z
[159] Mai I, Störmer E, Bauer S, Krüger H, Budde K, Roots I. Impact of St John's wort treat-
ment on the pharmacokinetics of tacrolimus and mycophenolic acid in renal transplant
patients. Nephrology, Dialysis, Transplantation. 2003;18:819-822. DOI: 10.1093/ndt/
gfg002
[160] Hermann M, Asberg A, Reubsaet JL, Sather S, Berg KJ, Christensen H. Intake of grape-
fruit juice alters the metabolic pattern of cyclosporin a in renal transplant recipients.
International Journal of Clinical Pharmacology and Therapeutics. 2002;40:451-456.
DOI: 10.5414/CPP40451
[162] Lin HL, Kent UM, Hollenberg PF. The grapefruit juice effect is not limited to cytochrome
P450 (P450) 3A4: Evidence for bergamottin-dependent inactivation, heme destruction,
and covalent binding to protein in P450s 2B6 and 3A5. The Journal of Pharmacology
and Experimental Therapeutics. 2005;313:154-164. DOI: 10.1124/jpet.104.079608
[163] Messer A, Raquet N, Lohr C, Schrenk D. Major furocoumarins in grapefruit juice II:
Phototoxicity, photogenotoxicity, and inhibitory potency vs. cytochrome P450 3A4
activity. Food and Chemical Toxicology. 2012;50:756-760. DOI: 10.1016/j.fct.2011.11.023
[164] Burkina V, Zlabek V, Halsne R, Ropstad E, Zamaratskaia G. In vitro effects of the citrus
flavonoids diosmin, naringenin and naringin on the hepatic drug-metabolizing CYP3A
enzyme in human, pig, mouse and fish. Biochemical Pharmacology. 2016;110-111:109-116.
DOI: 10.1016/j.bcp.2016.04.011
[165] Lin HL, Kenaan C, Hollenberg PF. Identification of the residue in human CYP3A4 that
is covalently modified by bergamottin and the reactive intermediate that contributes to
the grapefruit juice effect. Drug Metabolism and Disposition. 2012;40:998-1006. DOI:
10.1124/dmd.112.044560
[166] Ho PC, Saville DJ, Wanwimolruk S. Inhibition of human CYP3A4 activity by grape-
fruit flavonoids, furanocoumarins and related compounds. Journal of Pharmacy &
Pharmaceutical Sciences. 2001;4:217-227