j. antimicrob. chemother. 2008 sousa 872 8 (1)

Upload: julianda-dini-halim

Post on 03-Apr-2018

216 views

Category:

Documents


0 download

TRANSCRIPT

  • 7/29/2019 J. Antimicrob. Chemother. 2008 Sousa 872 8 (1)

    1/7

    Pharmacokinetics and pharmacodynamics of drug interactionsinvolving rifampicin, rifabutin and antimalarial drugs

    Marta Sousa1,2, Anton Pozniak1 and Marta Boffito1*

    1St Stephens Centre, Chelsea and Westminster Hospital, 369 Fulham Road, London SW10 9NH, UK;2Centro Hospitalar de Vila Nova de Gaia, Portugal

    Malaria and tuberculosis (TB) are two major global diseases mostly affecting the developing countries.

    Their treatment is often complex because of the drugs used, multidrug resistance, drug interactions

    and logistic problems such as drug availability and access. Patients are treated for TB for a minimum

    of 6 months and may concomitantly develop and be treated for malaria, especially during the rainy

    season. Rifampicin, a standard component of combination regimens for treating TB, is a potent

    inducer of hepatic cytochrome and other metabolic enzymes and is able to influence the pharmaco-

    kinetics of many drugs. Rifabutin, another rifamycin used less frequently than rifampicin, can alsointeract with drugs metabolized through the hepatic cytochromes. The mechanisms of any interaction

    of rifamycins with drugs used in malaria are not well defined. To complicate matters, acute malaria

    also plays a role in the pharmacokinetics and pharmacodynamics of drugs (i.e. quinine). The aim of

    this paper is to review known and potential drugdrug interactions between rifampicin, rifabutin and

    antimalarial drugs.

    Keywords: treatment of malaria, tuberculosis, pharmacology of antimalarial agents

    Introduction

    Malaria and tuberculosis (TB) are endemic diseases in manycountries with limited resources and are frequently found at the

    same time. Together, they kill more than 3 million people each

    year.1

    These diseases have a synergic negative effect on public

    health and are considered to be great barriers to economic devel-

    opment. The relationship of these diseases with HIV and the

    problems of drug resistance are major causes of concern, and

    they adversely affect morbidity and mortality.1

    The co-existence of these two diseases leads to various con-

    cerns about their treatment. For several reasons, simultaneous

    malaria and TB treatment poses many challenges. One of these

    is the potential for drug drug interactions between the most

    commonly used drugs. Rifampicin, a standard component of

    combination regimens for treating TB, is a potent inducer of

    hepatic metabolism and is able to influence the pharmaco-kinetics of several drugs. Rifabutin, another rifamycin used less

    frequently than rifampicin, can also interact with drugs meta-

    bolized through the hepatic cytochromes. The drugdrug inter-

    actions of rifamycins and most drugs used in malaria are not

    well defined. Moreover, acute malaria itself can influence the

    pharmacokinetics and pharmacodynamics of different drugs by

    influencing liver metabolism, binding to plasma proteins and

    excretion by mechanisms that remain unclear.2

    The aim of this paper is to review known and potential

    drug drug interactions between rifampicin, rifabutin and anti-

    malarial agents.

    Pharmacokinetics, pharmacodynamics and drug

    interactions

    Pharmacokinetics studies the absorption, distribution, meta-

    bolism and excretion of a drug, and pharmacodynamics studies

    the relationship between the drug and its receptors, its mechan-

    ism of action and therapeutic effect. Both can play a role in

    drugdrug interactions.

    Many drugs share the same metabolic pathways or target the

    same receptors. This leads to the occurrence of pharmacokinetic

    and pharmacodynamic differences, respectively. Drug druginteractions occur mainly during drug absorption, distribution

    (plasma drug binding protein), biotransformation and excretion.

    Most drugs are given by the oral route. Subsequently, absorp-

    tion is mostly through the intestinal mucosa (duodenum). Drug

    bioavailability is dependent on an enterocyte P-glycoprotein,

    which can actively pump back drugs into the intestinal lumen,

    and on enterocyte cytochrome P450 (CYP450) enzymes, meta-

    bolizing the drug before it reaches the systemic circulation.3

    . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .

    *Corresponding author. Tel: 44-20-8846-6507; Fax: 44-20-8746-5628; E-mail: [email protected]

    Journal of Antimicrobial Chemotherapy (2008) 62, 872 878

    doi:10.1093/jac/dkn330

    Advance Access publication 18 August 2008

    . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .

    872

    # The Author 2008. Published by Oxford University Press on behalf of the British Society for Antimicrobial Chemotherapy. All rights reserved.For Permissions, please e-mail: [email protected]

  • 7/29/2019 J. Antimicrob. Chemother. 2008 Sousa 872 8 (1)

    2/7

    Induction or inhibition of P-glycoprotein and enterocyte

    CYP450 can thus influence drug bioavailability. After being

    absorbed, drugs generally bind to plasma transport proteins

    (i.e. albumin, a-1-acid-glycoprotein). The level of binding

    protein plays a fundamental role in distribution and various

    drugs can affect protein binding. When the drug passes through

    the liver, it is metabolized to active and/or non-active meta-

    bolites. This is called biotransformation and this has a great

    influence on therapeutic efficacy. Drug biotransformation isdependent on several phases. Phase I consists of hydrolysis, oxi-

    dation and reduction, which is mediated mostly by CYP450.4,5

    Phase II consists of conjugation and is mediated by enzymes

    such as uridine diphosphoglucuronosyl transferase, N-acetyl

    transferase and glutathione S-transferase. These enzymes play an

    important role in the detoxification and/or excretion rate of

    xenobiotics.5,6

    Phase III is mediated by drug plasma membrane

    transporters (influx and efflux), such as P-glycoprotein, multi-

    drug resistance protein (MRP) and organic anion transport

    protein 2 (OATP2), that are localized in the liver on the endo-

    thelium and epithelium membrane, as well as in the gastrointes-

    tinal tract, kidney and other organs. They play an important role

    in drug pharmacokinetics and pharmacodynamics. In the liver,

    OATP1 and OATP2 are localized in sinusoidal phase membraneof hepatocytes and are responsible for the uptake of many drugs

    to be metabolized.7

    MRP2 and P-glycoprotein are localized in

    the canalicular phase membrane of the hepatocytes and are

    responsible for pumping out xenobiotics (i.e. drugs and their

    metabolites) to the biliary canals.7,8

    Since they are present in

    several types of cells, and mediate cell exposure to drugs, they

    can influence not only absorption, distribution, metabolism and

    elimination, but also drug concentrations inside the target cell

    and affect the therapeutic efficacy.5

    Acute malaria itself has been shown to influence the metabolism

    and distribution of certain drugs not, it seems, by influencing drug

    absorption but by increasing binding to a-1-glycoprotein in

    plasma.2

    Inhibition of hepatic metabolism, mainly CYP3A4

    enzymes in acute malaria, leads to a reduced clearance of quinine.9

    Rifampicin and rifabutin pharmacokinetics

    Both rifampicin and rifabutin are drugs used for the treatment of

    TB. Rifampicin is derived from rifampicin B that is produced

    by Streptomyces mediterranei and has been in use since 1965.

    Rifabutin is derived from rifamycin S and is mostly used in HIV

    co-infected patients because it has fewer drug interactions with

    antiretroviral agents (i.e. protease inhibitors) than rifampicin.

    It is also used in atypical mycobacterial infections and, in some

    cases, it has been shown to be active when there is rifamycin

    resistance.1012

    Both drugs act by inhibiting the b-subunit-dependent DNA RNA polymerase that leads to suppression

    of DNA formation by Mycobacterium tuberculosis. They are

    bactericidal and bacteriostatic and act intra- and extracellularly.

    Absorption

    Both drugs are generally well absorbed by the oral route.

    Altered absorption, however, has been observed in patients

    with AIDS, diabetes and gastrointestinal diseases. Furthermore,

    pharmacokinetic studies show a high inter-individual variability

    of rifampicin absorption characterized by slow and fast absorp-

    tion patterns.13

    Distribution

    Rifampicin is highly lipophilic, it is 80% bound to plasma

    proteins, mainly a-1-acid-glycoprotein,14

    and it is characterized

    by a plasma half-life of 2 5 h. Rifabutin is 85% bound to

    plasma proteins in a concentration-independent manner. Due totheir high lipophilicity, both demonstrate a high propensity for

    distribution and intracellular tissue uptake.

    Biotransformation

    Rifampicin is deacetylated by hepatic microsomal enzymes, and

    it is also a great CYP450 inducer, the most potent inducer of all

    drugs used in clinical practice. This leads to decreased drug con-

    centrations by an autoinduction mechanism, whereby the drug

    stimulates its own metabolism into inactive metabolites.15

    It undergoes progressive enterohepatic circulation and deacetyl-

    ation to the primary active metabolite, 25-desacetyl-rifampicin.

    Rifabutin is metabolized by CYP3A.

    16

    Of the five meta-bolites that have been identified, 25-O-desacetyl and 31-hydroxy

    are the most predominant. The former has an activity equal to

    the parent drug and contributes up to 10% of the total antimicro-

    bial activity.13,17

    Excretion

    Rifampicin is rapidly eliminated mainly in the bile. Up to 30%

    is excreted in urine. Rifabutin is mostly excreted in urine, pri-

    marily as metabolites. About 30% of the dose is excreted in the

    faeces.13,17

    Enzyme induction and inhibitionBecause of the influence on the liver metabolic activity, rifam-

    picin has been shown to be involved in several drugdrug

    interactions.

    Rifampicin is an N-acetyltransferase inhibitor and leads to a

    decrease in the acetylation ratio in fast acetylators. However, the

    most important mechanism behind rifampicin drug interactions

    is its potent inducing effect on hepatic and intestinal CYP450

    enzyme activity (CYP3A4, CYP1A2, CYP2C9, CYP2C8 and

    CYP2C18/19).5,18

    Moreover, rifampicin has an important role in hepatic drug

    uptake and gastrointestinal absorption of drugs as it is a

    P-glycoprotein,8,18,19

    an MRP,20

    a transport system inducer and

    an OATP2 inhibitor.8,18

    Based on tissue distribution of these

    membrane transporters, modulation of their activity may result

    in significant alterations in the pharmacokinetics and, poten-

    tially, the pharmacodynamics of several drugs.

    The mechanism of rifampicin induction of CYP enzymes is

    mediated by the activation of nuclear pregnane X receptor

    (PXR), a member of the nuclear receptor superfamily. A recent

    study performed in mice demonstrated that CYP3A4 is

    dysregulated in PXR-null mice, confirming that PXR transcrip-

    tionally regulates CYP3A4.21,22

    PXR binds to CYP3A promoters

    to activate reporter genes. PXR also up-regulates other Phase I,

    II and III enzyme genes,21

    such as MDR1, a gene encoding

    Review

    873

  • 7/29/2019 J. Antimicrob. Chemother. 2008 Sousa 872 8 (1)

    3/7

    P-glycoprotein. Rifampicin is a PXR ligand and activates tran-

    scription of CYP3A4 and other proteins such as P-glycoprotein.

    Another receptor, constitutive androstane receptor (CAR), is

    also involved in CYP3A4 transcriptional regulation, but rifam-

    picin has a lesser effect on CAR than on PXR.

    Rifampicin is responsible for broad changes in the pattern of

    gene expression, rather than increased expression of a small

    number of metabolic enzymes. Clinicians should be alert to the

    possibility of its multiple effects.23

    Rifabutin induces CYP3A and may reduce the plasma concen-

    trations of drugs that are mostly metabolized by this enzyme.

    However, rifabutin is a weaker enzyme inducer than rifampicin.24

    Importantly, rifabutin is also a substrate of CYP3A4. Therefore,

    its concentrations may be altered during co-administration with

    CYP3A4 inducers and inhibitors and dose adjustments may be

    required.

    Antimalarial drugs

    Malaria treatment is far from simple. Resistance to chloroquine

    is widespread, and resistance to other agents and multidrug

    resistance is emerging in many countries.

    There are 15002000 cases of imported malaria reported in

    the UK each year and 10 20 deaths. Approximately three-

    quarters of reported malaria cases in the UK are caused by

    Plasmodium falciparum.25

    In 2002, 1337 cases of malaria were

    reported in the USA; all but five were imported.26

    The success

    of malaria treatment is dependent on knowledge of the current

    malaria resistance patterns of the areas where the patient was

    infected.

    Quinine

    Quinine is one of the most commonly used drugs for malaria

    treatment worldwide. It is the first-line treatment for P. falciparumcomplicated malaria and is a treatment option for P. falciparum

    uncomplicated malaria in chloroquine-resistant regions.2527

    Quinine seems to act against Plasmodium spp. by inhibiting haem

    polymerase that leads to toxic haem levels.27

    Quinine metabolism

    is interesting because acute malaria influences absorption, the

    binding-protein fraction, CYP450 activity and excretion of

    quinine. Pukrittayakamee et al.9

    studied patients with severe

    malaria to assess the factors that could influence the decreased

    clearance of quinine and concluded that the most important factor

    was the impairment of CYP3A function.

    Biotransformation. In acute malaria, 78% to 95% of quinine is

    bound to a-1-acid-glycoprotein.2

    It is metabolized almost

    exclusively via CYP450, mostly by CYP3A4,28

    and CYP2C19.The metabolism of quinine is not completely understood but

    CYP1A2 and CYP2D6 may also have a role.29,30

    One of its

    metabolites is 3-hydroxyquinine, which is important as it has

    10% of the activity of the parent drug.29

    Excretion. Quinine is mostly eliminated via the hepatic route,

    and 20% of it is excreted unchanged in the urine.

    Enzyme induction and inhibition. Quinine inhibits the activity of

    CYP2D631

    and inhibits P-glycoprotein32

    and biliary excretion.

    Therefore, it has an important role in the elimination of several

    drugs.

    Drug interactions. There is evidence that rifampicin may have

    antimalarial activity in humans.33,34

    The antimalarial mechanism

    of rifampicin may be similar to that observed in TB. The target

    of rifampicin may be the Plasmodium organellar circular DNA

    molecule that encodes the b-subunit of a prokaryocyte-like RNA

    polymerase.29,35

    Both quinine and rifampicin influence several steps in drug

    metabolism and distribution.

    Enterocyte P-glycoprotein activity is inhibited by quinine and

    induced by rifampicin.3

    Rifampicin also induces enterocyte

    CYP3A. Overall, in theory, this could lead to a decrease in bio-

    availability of quinine.

    In plasma, both rifampicin and quinine are bound to

    a-1-acid-glycoprotein. Acute malaria, TB14

    and rifampicin

    administration36

    increase a-1-acid-glycoprotein levels, and this

    could influence quinine distribution, metabolism and efficacy.

    The OATP system is responsible for quinine hepatic uptake

    and rifampicin is an OATP inhibitor7,8,18

    and could therefore

    theoretically influence quinine concentrations. Moreover, by

    inhibiting P-glycoprotein, quinine inhibits biliary excretion7 andmay theoretically alter rifampicin half-life.

    Several in vivo studies have been performed to investigate the

    interactions between quinine and rifampicin. Wanwimolruk

    et al.37

    studied the effect of 1 week pre-treatment with rifam-

    picin (and isoniazid) on quinine pharmacokinetics in healthy

    volunteers. They showed that the mean clearance of quinine was

    significantly greater (0.87 versus 0.14 L/h/kg) and the mean

    elimination half-life of quinine was shorter with rifampicin

    pre-treatment than when quinine was given alone (5.5 versus

    11.1 h).

    Pukrittayakamee et al.29

    studied the effect of combined

    therapy (rifampicin and quinine) in patients with acute uncom-

    plicated P. falciparum malaria. Interestingly, they observed that

    the parasite clearance was faster with the combination. Thiscould be because of the rifampicin antimalarial effect rather

    than a drug interaction as rifampicin can take several days to

    induce CYP3A4 activity that is responsible for quinine meta-

    bolism. After the second day of rifampicin treatment, quinine

    pre-dose plasma concentrations decreased progressively through-

    out the treatment period, leading to a fall in quinine activity that

    may have been the reason for a malaria recrudescence rate five

    times higher in this group. The authors concluded that, although

    rifampicin could have therapeutic effects in malaria, the drugs

    should not be combined to treat malaria. In those patients who

    are already on rifampicin for the treatment of TB, quinine doses

    must be increased. However, no advice on dose-adjustment strat-

    egies has been provided and quinine therapeutic drug monitoring

    has been suggested to be useful to guide dosing.

    Cerebral malaria is the commonest cause of non-traumatic

    encephalopathy in the world. The standard treatment is with

    quinine. Quinine penetrates relatively poorly into the cerebro-

    spinal fluid (CSF) in patients with cerebral malaria, with a con-

    centration approximately 2% to 7% of that measured in plasma.

    P-glycoprotein is one of the blood brain barrier (BBB) mem-

    brane transporters and modulates central nervous system (CNS)

    drug exposure, playing an important role in preventing CNS

    drug accumulation. Reducing P-glycoprotein activity dramati-

    cally increases the penetration of many therapeutic drugs into

    Review

    874

  • 7/29/2019 J. Antimicrob. Chemother. 2008 Sousa 872 8 (1)

    4/7

    the CNS. Studies in rats have shown that brain capillary

    P-glycoprotein is transcriptionally up-regulated by the PXR, and as

    rifampicin is a PXR ligand, it increases P-glycoprotein expression

    in the BBB.38

    Therefore, the co-administration of rifampicin

    and quinine could decrease quinine concentrations in the CSF.

    However, whether this compromises drug efficacy remains

    unclear, as the clinically relevant concentrations of quinine are

    those measured in the systemic circulation, where parasitized

    erythrocytes remain.Despite being a less potent CYP3A4 inducer than rifampicin,

    rifabutin and quinine should also be cautiously co-administered

    as both drugs use the same metabolic pathways. Nevertheless,

    no data are available on the potential interaction between

    rifabutin and quinine.

    Chloroquine

    Despite the worldwide spread of P. falciparum resistance to

    chloroquine, this drug is still used as a first-line treatment for

    Plasmodium ovale, Plasmodium malariae and Plasmodium

    vivax in regions where there is no known resistance.

    The mechanism of plasmodicidal action of chloroquine is not

    completely certain. It is thought to inhibit the haem polymeraseof Plasmodium, increasing haem to toxic levels. Chloroquine is

    almost completely absorbed when given by the oral route. It

    binds to plasma proteins, and it is metabolized in the liver

    by CYP450 (mainly CYP2C8 and CYP3A4/5)39

    to desethyl-

    chloroquine, which has an activity similar to chloroquine, and

    bisdesethylchloroquine. The MRP membrane transport system is

    responsible for chloroquine cellular efflux and may be the

    mechanism implicated in the development of resistance to

    chloroquine. There are several in vitro studies suggesting that

    P. falciparum chloroquine resistance may be reversed with

    P-glycoprotein inhibitors such as roxithromycin40

    or fluoxe-

    tine.41

    Importantly, despite reporting that the observed effect

    occurred at clinically achievable concentrations, the difference

    in drug protein binding between the in vitro and in vivo systemsmay lead to remarkable differences between the two environ-

    ments. Similar findings, however, have been observed both

    in vitro and in vivo with calcium channel blockers (i.e. verapamil).

    These showed effectiveness against chloroquine-resistant

    P. falciparum by increasing chloroquine accumulation inside the

    food vacuole.42

    One study in infected mice reported that the combination of

    rifampicin and chloroquine decreased their survival rate and the

    rate of clearance of parasitaemia and increased the rate of recru-

    descence.43

    The decrease in chloroquine efficacy when admini-

    strated in combination with rifampicin may be due to the

    inducing effect of rifampicin on MRP20

    and on CYP450

    activity. Concomitant use of these drugs might have to be

    avoided; however, data in humans are lacking.

    Studies are needed to explore drugdrug interactions

    between rifabutin and chloroquine as there are no data regarding

    this interaction.

    Atovaquone and proguanil

    Atovaquone, combined with proguanil, is a treatment option for

    P. vivax infection. It is also used for malaria prophylaxis.

    Its activity against Plasmodium is due to the inhibition of mito-

    chondrial electron transport.27

    Atovaquone is 99% bound to

    plasma proteins and has a 70 h half-life. It is characterized by an

    enterohepatic circulation, is metabolized in the liver and is mostly

    excreted unchanged in the faeces. There is in vitro evidence of

    possible inhibition of CYP3A4 by atovaquone,44

    and studies that

    used atovaquone in the treatment of Toxoplasma gondii showed

    that atovaquone area under the curve (AUC) decreased remark-

    ably (50%) when combined with rifampicin but not as much

    (34%) with rifabutin.44

    The concomitant administration of atova-

    quone and rifampicin is therefore not recommended, while theclinical significance of the moderate decrease in atovaquone con-

    centrations in the presence of rifabutin remains unclear.

    Moreover, despite the lack of studies on rifabutin and atova-

    quone in malaria, there are several studies on their co-administration

    for the treatment of opportunistic infections. This combination

    seems to have a synergistic effect in the prophylaxis of

    Pneumocystis jiroveci pneumonia and T. gondii in mice.45

    Proguanil is used in combination with atovaquone and should

    not be used alone because resistance to it develops very

    quickly.27

    Seventy-five percent of the drug is bound to plasma

    proteins, and it undergoes bioactivation by CYP2C19 to the

    active metabolite, cycloguanil. Cycloguanil inhibits plasmodial

    dihydrofolate reductase and influences DNA synthesis.

    Since rifampicin is a potent inducer of CYP2C19, it is poss-ible that it could affect proguanil antimalarial activity.

    Data on this interaction are unavailable and further studies

    are needed to clarify the effect of rifampicin and rifabutin on

    proguanil pharmacokinetics and efficacy.

    Mefloquine

    Mefloquine is an antimalarial agent used for the treatment and

    prophylaxis of chloroquine-resistant P. falciparum and non-

    falciparum malaria. It is also often used in combination with

    artemisinins. Its exact mechanism of action is not known. After

    absorption, it binds almost completely to plasma proteins and is

    metabolized by CYP3A into two inactive metabolites.

    46

    It isknown that the pharmacokinetics of mefloquine is altered in

    acute malaria.47

    Just like chloroquine, mefloquine resistance seems to be

    mediated by MDR transport system activity,48 which can be

    increased by rifampicin. Furthermore, rifampicin induces meflo-

    quine metabolism,46

    decreasing its AUC by 68% and half-life

    by 63%.49

    Importantly, mefloquine metabolite AUC and clear-

    ance increased by 30% and 25%, respectively. These changes

    did not reach a statistical significance. The authors suggested

    avoiding simultaneous administration of mefloquine and rifampi-

    cin on the basis of these pharmacokinetic findings. However, no

    data are available on the clinical efficacy of this combination.

    In 1999, Bermudez et al.50

    found that mefloquine has in vitro

    and in vivo activity against Mycobacterium avium complexincluding strains with multidrug resistance. More studies are

    needed to further increase our knowledge about mefloquine

    pharmacodynamics and drug interactions with rifabutin and

    rifampicin.

    Artemisinins

    Artemisinins and their derivates (i.e. artemether and artesunate)

    are drugs recently developed to treat malaria. The advantages of

    these drugs are the antimalarial effect against P. falciparum

    Review

    875

  • 7/29/2019 J. Antimicrob. Chemother. 2008 Sousa 872 8 (1)

    5/7

    gametocytes (previously only susceptible to primaquine) and the

    lack of evidence of resistance to these agents to date.

    Artemether did not prove to be better than quinine on survi-

    val rate,51

    and artesunate is the first choice in low-transmission

    areas. These drugs should be given in combination therapy with

    other antimalarial agents (i.e. mefloquine) since better parasite

    clearance rates have been observed compared with monother-

    apy.52

    High efficacy of two artemisinin-based combinations

    (artesunate plus amodiaquine and artemether plus lumefantrine)has been shown over the past few years in areas where TB is

    endemic.53

    The clinical pharmacology of artemisinin-based combination

    therapies is highly complex. All artemisinin compounds are

    metabolized by CYP3A454

    to the active compound dihydroarte-

    misinin, also available as a drug in itself. They also seem to

    induce CYP2C19 activity and their own metabolism.54,55

    There

    are no studies looking at the concomitant administration of

    artemisinins and rifampicin or rifabutin. However, theoretically,

    drug interactions may occur because of the inducing effect of

    rifampicin and rifabutin on different CYP450 enzymes, leading

    to a more extensive time-dependent decline in drug plasma con-

    centrations and hypothetical decrease in efficacy.

    Amodiaquine and lumefantrine are metabolized by CYP2C856

    and 3A4,57

    respectively, and may be theoretically decreased

    by rifampicin or rifabutin co-administration. Pharmacokinetic/

    pharmacodynamic investigations to investigate the interaction

    between artemether/lumefantrine (Coartemw

    ) and rifampicin are

    currently being conducted at the University of Makerere, Kampala,

    Uganda (clinicaltrials.gv NCT00620438; Dr Mohammed Lamorde,

    personal communication).

    Primaquine

    Primaquine is used to provide a cure for P. vivax and P. ovale

    malaria, and it is also gametocytocidal against P. falciparum.

    The mechanism of action is unknown.27

    It is metabolized in the

    liver by CYP450 enzymes, possibly CYP3A4.58,59 Therefore,co-administration of primaquine and rifampicin or rifabutin

    may influence the pharmacokinetics of primaquine and alter

    its plasma concentration. However, there are no data on this

    potential interaction.

    Doxycycline and clindamycin

    Doxycycline is a tetracycline derivative that is used in malaria

    treatment in combination with quinine to ensure cure.25,29,60 It is

    an inhibitor of protein synthetase by disrupting messenger RNA

    and transfer RNA; 80% to 95% binds to plasma proteins and is

    metabolized in the liver by an unknown mechanism. No meta-

    bolites have been identified to date.61

    Sixty percent is excreted

    in the faeces and 40% in the urine. Hepatic inducers such as

    rifampicin, and probably rifabutin, accelerate its metabolism and

    influence its plasma exposure (doxycycline AUC and half-life

    decreased by 40% with rifampicin).62

    However, whether drug

    efficacy is compromised remains unclear.

    Clindamycin is a lincosamide antibiotic that inhibits early

    stages of protein synthesis at the ribosome level. It is used

    in malaria treatment as a valid alternative to doxycycline in

    pregnant women. Ninety percent is bound to plasma proteins.

    It is metabolized in the liver by CYP3A enzymes,63

    to several

    active and inactive metabolites that are excreted in the faeces.

    Clindamycin is a moderate inhibitor of CYP3A in vitro, and

    the potential for drug drug interactions involving clindamycin

    is low.63

    However, rifampicin induces CYP3A4 and could

    theoretically induce clindamycin metabolism and decrease its

    half-life. These drugs should be co-administered cautiously.

    Conclusions

    Malaria and TB are two diseases that are often found concomi-

    tantly in developing countries and can have a synergic negative

    effect on public health. In developed countries almost all cases

    of malaria are imported, but TB still has a high prevalence in

    some European countries, especially associated with HIV infec-

    tion. Treatment of these two diseases leads to several concerns

    about drug drug interactions and the potential for inducing drug

    resistance.

    Rifampicin, a standard component of combination regimens for

    treating TB, has a great influence on the bioavailability and the

    efficacy of several drugs, not only because of the inhibition of

    Phase I and II enzymes of hepatic metabolism, but also because of

    its effect on drug absorption and distribution. It induces almost all

    CYP450 enzymes, it inhibits N-acetyltransferases and it alters theexpression of membrane transporters. Rifampicin induces intesti-

    nal, BBB and hepatic P-glycoprotein and MRP expression, and it

    inhibits OATP2. Most of these inducing effects are due to PXR-

    increased transcription; therefore, understanding each individuals

    genotype is important to explain the several unknown mechanisms

    behind these complex drugdrug interactions. Rifabutin is mostly

    used in HIV co-infected patients because it has fewer drugdrug

    interactions with antiretroviral agents. Although rifabutin is a

    weaker CYP3A4 enzyme inducer than rifampicin, it may be

    expected to have some effect on drug metabolism as well. In

    many cases, the interactions between rifampicin or rifabutin and

    antimalarial agents are likely but not studied and confirmed.

    Moreover, the consequences of simultaneous malaria and TB

    treatment are not well known. In each case, the need for conco-

    mitant treatment should be carefully considered, since malaria is

    an acute disease and TB is a subacute disease. There are a large

    number of drug mechanisms that need to be explored concerning

    antimalarial drug activity. These may be vital to treat TB and

    malaria simultaneously where multidrug resistance in both

    diseases is common.

    Transparency declarations

    Conflicts of interest: none to declare.

    References

    1. Department for International Development. The Challenge of TB

    and Malaria Control. 2005. wwww.dfid.gov.uk/Pubs/files/tb-malaria-

    control.pdf.

    2. Krishna S, White NJ. Pharmacokinetics of quinine, chloroquine

    and amodiaquine. Clinical implications. Clin Pharmacokinet 1996; 30:

    26399.

    3. Cummins CL, Jacobsen W, Benet LZ. Unmasking the dynamic

    interplay between intestinal P-glycoprotein and CYP3A4. J Pharmacol

    Exp Ther2002; 300: 103645.

    Review

    876

  • 7/29/2019 J. Antimicrob. Chemother. 2008 Sousa 872 8 (1)

    6/7

    4. Derelanko MJ, Hollinger MA. Metabolism and toxicokinetics of

    xenobiotics. In: Derelanko MJ, Hullinger MA, eds. Handbook of

    Toxicology, 1st edn. New York: CRC Press, 1995; 53979.

    5. Brandon EFA, Raap CD, Meijerman I et al. An update on in vitro

    test methods in human hepatic drug biotransformation research: pros

    and cons. Toxicol Appl Pharmacol 2003; 189: 23346.

    6. Lu FC. Biotransformation of toxicants. Basic Toxicology:

    Fundamentals, Target Organs and Risk Assessment, 3rd edn.

    Washington, DC: Taylor and Francis, 1996; 2739.

    7. Liu L, Sandy Pang K. The roles of transporters and enzymes in

    hepatic drug processing. Drug Metab Dispos2005; 33: 19.

    8. Lau YY, Wu CY, Okochi H et al. Ex situ inhibition of hepatic

    uptake and efflux significantly changes metabolism: hepatic enzyme-

    transporter interplay. J Pharmacol Exp Ther2004; 308: 10405.

    9. Pukrittayakamee S, Looareesuwan S, Keeratithakul D et al.

    A study of the factors affecting the metabolic clearance of quinine in

    malaria. Eur J Clin Pharmacol 1997; 52: 48793.

    10. Senol G, Erbaycu A, Ozsoz A. Incidence of cross resistance

    between rifampicin and rifabutin in Mycobacterium tuberculosis strains

    in Izmir, Turkey. J Chemother2005; 17: 3804.

    11. Luna-Herrera J, Reddy MV, Gangadharam PR. In-vitro and

    intracellular activity of rifabutin on drug-susceptible and multiple

    drug-resistant (MDR) tubercle bacilli. J Antimicrob Chemother 1995;

    36: 35563.12. Sintchenko V, Chew WK, Jelfs PJ et al. Mutations in rpoB gene

    and rifabutin susceptibility of multidrug-resistant Mycobacterium tuber-

    culosisstrains isolated in Australia. Pathology 1999; 31: 25760.

    13. Peloquin CA, Jaresko GS, Yong CL et al. Population pharmaco-

    kinetic modeling of isoniazid, rifampicin, and pyrazinamide. Antimicrob

    Agents Chemother1997; 41: 26709.

    14. Johnson DA, Smith KD. The efficacy of certain anti-tuberculosis

    drugs is affected by binding to a-1-acid glycoprotein. Biomed

    Chromatogr 2006; 20: 55160.

    15. Benedetti SM, Dostert P. Induction and autoinduction properties

    of rifamycin derivatives: a review of animal and human studies. Environ

    Health Perspect1994; 102: 1015.

    16. Burman WJ, Gallicano K, Peloquin C. Comparative pharmaco-

    kinetics and pharmacodynamics of the rifamycin antibacterials. Clin

    Pharmacokinet 2001; 40: 32741.

    17. Iwainsky H. Mode of action, biotransformation and pharmaco-

    kinetics of antituberculosis drugs in animals and man. In: Bartmann K,

    ed. Antituberculosis Drugs. Berlin: Springer-Verlag, 1988; 399553.

    18. Finch CK, Chrisman CR, Baciewicz AM et al. Rifampicin and

    rifabutin drug interactions. Arch Intern Med 2002; 162: 98592.

    19. Schuetz EG, Schinkel AH, Relling MV et al. P-glycoprotein: a

    major determinant of rifampicin-inducible expression of cytochrome

    P4503A in mice and humans. Proc Natl Acad Sci USA 1996; 93:

    40015.

    20. Ehret MJ, Levin GM, Narasimhan M et al. Venlafaxine induces

    P-glycoprotein in human Caco-2 cells. Hum Psychopharmacol 2007;

    22: 4953.

    21. Chen J, Raymond K. Roles of rifampicin in drug drug inter-

    actions: underlying molecular mechanisms involving the nuclear preg-nane X receptor. Ann Clin Microbiol Antimicrobials 2006; 5: 3.

    22. Xie W, Barwick JL, Downes M. Humanized xenobiotic response

    in mice expressing nuclear receptor SXR. Nature2000; 406: 4359.

    23. Rae JM, Johnson MD, Lippman ME et al. Rifampicin is a

    selective, pleiotropic inducer of drug metabolism genes in human

    hepatocytes: studies with cDNA and oligonucleotide expression arrays.

    J Pharmacol Exp Ther2001; 299: 84957.

    24. Reinach B, de Sousa G, Dostert P et al. Comparative effects of

    rifabutin and rifampicin on cytochromes P450 and UDP-glucuronosyl-

    transferases expression in fresh and cryopreserved human hepato-

    cytes. Chem Biol Interact1999; 121: 3748.

    25. Lalloo DG, Shingadia D, Pasvol G et al. UK malaria treatment

    guidelines. J Infect2007; 54: 11121.

    26. CDC. Treatment Guidelines Treatment of Malaria (Guidelines for

    Clinicians). 2004.

    27. World Health Organization. Guidelines for the Treatment of

    Malaria. 2006.

    28. Zhang H, Coville PF, Walker RJ et al. Evidence for involvement

    of human CYP3A in the 3-hydroxylation of quinine. Br J Clin

    Pharmacol 1997; 43: 24552.29. Pukrittayakamee S, Prakongpan S, Wanwimolruk S et al .

    Adverse effect of rifampicin on quinine efficacy in uncomplicated

    falciparum malaria. Antimicrob Agents Chemother2003; 47: 150913.

    30. Mirghani RA, Hellgren U, Westerberg PA et al. The roles of

    cytochrome P4503A4 and 1A2 in the 3-hydroxylation of quinine in vivo.

    Clin Pharmacol Ther1999; 66: 45460.

    31. Branch RA, Adedoyin A, Frye RF et al. In vivo modulation of

    CYP enzymes by quinidine and rifampicin. Clin Pharmacol Ther 2000;

    68: 40111.

    32. Drescher S, Glaeser H, Murdter T et al. P-glycoprotein-mediated

    intestinal and biliary digoxin transport in humans. Clin Pharmacol Ther

    2003; 73: 22331.

    33. Pukrittayakamee S, Viravan C, Charoenlarp P et al. Antimalarial

    effects of rifampicin in Plasmodium vivax malaria. Antimicrob Agents

    Chemother1994; 38: 5114.34. Strath M, Scott-Finnigan T, Gardner M et al. Antimalarial activity

    of rifampicin in vitro and in rodent models. Trans R Soc Trop Med Hyg

    1993; 87: 2116.

    35. Gardner MJ, Williamson DH, Wilson RJM. A circular DNA in

    malaria parasites encodes an RNA polymerase like that of prokaryotes

    and chloroplasts. Med Biochem Parasitol 1991; 44: 11524.

    36. Abramson FP, Lutz MP. The kinetics of induction by rifampicin

    of a 1-acid glycoprotein and antipyrine clearance in the dog. Drug

    Metab Dispos1986; 14: 4651.

    37. Wanwimolruk S, Kang W, Coville PF et al. Marked enhancement

    by rifampicin and lack of effect of isoniazid on the elimination of

    quinine in man. Br J Clin Pharmacol 1995; 40: 8791.

    38. Bauer B, Yang X, Hartz AMS et al. In vivo activation of human

    pregnane X receptor tightens the blood-brain barrier to methadonethrough P-glycoprotein up-regulation. Mol Pharmacol 2006; 70:

    12129.

    39. Kim KA, Park JY, Lee JS et al. Cytochrome P450 2C8 and

    CYP3A4/5 are involved in chloroquine metabolism in human liver

    microsomes. Arch Pharm Res2003; 26: 6317.

    40. Min TH, Khairul MF, Low JH et al. Roxithromycin potentiates

    the effects of chloroquine and mefloquine on multidrug-resistant

    Plasmodium falciparum in vitro. Exp Parasitol2007; 115: 38792.

    41. Khairul MF, Min TH, Hou Low J et al. Fluoxetine potentiates

    chloroquine and mefloquine effect on multidrug-resistant Plasmodium

    falciparum in vitro. Jpn J Infect Dis 2006; 59: 32931.

    42. Rabinovich SA, Orlov VS, Dadasheva NR et al. Ca ion

    transport blockers as reversants of the drug resistance of malarial

    parasites. 1. The effect of verapamil on the resistance to chloroquine in

    vivo of Plasmodium berghei and in vitro of Plasmodium falciparum.Med Parazitol (Mosk)1996; 1: 1822.

    43. Hou LJ, Raju SS, Abdulah MS et al. Rifampicin antagonizes the

    effect of chloroquine on chloroquine-resistant Plasmodium berghei in

    mice. Jpn J Infect Dis 2004; 57: 198202.

    44. Thapar MM, Ashton M, Lindegardh N et al. Time-dependent

    pharmacokinetics and drug metabolism of atovaquone plus proguanil

    (Malarone) when taken as chemoprophylaxis. Eur J Clin Pharmacol

    2002; 58: 1927.

    45. Romand S, Della Bruna C, Farinotti R et al. In vitro and in vivo

    effects of rifabutin alone or combined with atovaquone against

    Toxoplasma gondii. Antimicrob Agents Chemother1996; 40: 201520.

    Review

    877

  • 7/29/2019 J. Antimicrob. Chemother. 2008 Sousa 872 8 (1)

    7/7

    46. Fontaine F, de Sousa G, Burcham PC et al. Role of cytochrome

    P450 3A in the metabolism of mefloquine in human and animal hepato-

    cytes. Life Sci 2000; 66: 2193212.

    47. Karbwang J, White NJ. Clinical pharmacokinetics of mefloquine.

    Clin Pharmacokinet1990; 19: 26479.

    48. Valderramos SG, Fidock DA. Transporters involved in resistance

    to antimalarial drugs. Trends Pharmacol Sci 2006; 27: 594 601.

    49. Ridtitid W, Wongnawa M, Mahatthanatrakul W et al. Effect of

    rifampin on plasma concentrations of mefloquine in healthy volunteers.J Pharm Pharmacol 2000; 52: 12659.

    50. Bermudez LE, Kolonoski P, Wu M et al. Mefloquine is active

    in vitro and in vivo against Mycobacterium avium complex. Antimicrob

    Agents Chemother 1999; 43: 18704.

    51. Newton PN, Angus BJ, Chierakul W et al. Randomized compari-

    son of artesunate and quinine in the treatment of severe falciparum

    malaria. Clin Infect Dis2003; 37: 716.

    52. Hassan Alin M, Bjorkman A, Landberg-Lindgren A et al. The

    effect of artemisinine, its derivatives and mefloquine against chloroquine-

    resistant strains of Plasmodium falciparum in vitro. Trans R Soc Trop

    Med Hyg1992; 86: 3657.

    53. Guthmann JP, Cohuet S, Rigutto C et al. High efficacy of

    two artemisinin-based combinations (artesunateamodiaquine and

    artemetherlumefantrine) in Caala, Central Angola. Am J Trop Med

    Hyg2006; 75: 1435.54. Giao PT, de Vries PJ. Pharmacokinetic interactions of antimalarial

    agents. Clin Pharmacokinet 2001; 40: 34373.

    55. German PI, Aweeka FT. Clinical pharmacology of artemisinin-

    based combination therapies. Clin Pharmacokinet2008; 47: 91 102.

    56. Li XQ, Bjorkman A, Andersson TB et al. Amodiaquine clearance

    and its metabolism to N-desethylamodiaquine is mediated by CYP2C8:

    a new high affinity and turnover enzyme-specific probe substrate.

    J Pharmacol Exp Ther2002; 300: 399407.

    57. Lefevre G, Thomsen MS. Clinical pharmacokinetics of artemether

    and lumefantrine (Riametw). Clin Drug Invest1999; 18: 46780.

    58. Bangchang KN, Karbwang J, Back DJ. Primaquine metabolismby human liver microsomes: effect of other antimalarial drugs.

    Biochem Pharmacol1992; 44: 58790.

    59. Constantino L, Paixao P, Moreira R et al. Metabolism of prima-

    quine by liver homogenate fractions. Evidence for monoamine oxidase

    and cytochrome P450 involvement in the oxidative deamination of pri-

    maquine to carboxyprimaquine. Exp Toxicol Pathol1999; 51: 299303.

    60. Pukrittayakamee S, White NJ. Combination therapy: making the

    best use of existing drugs. Pharm News 2001; 8: 215.

    61. Agwuh NK, MacGowan A. Pharmacokinetics and pharmaco-

    dynamics of the tetracyclines including glycylcyclines. J Antimicrob

    Chemother 2006; 58: 25665.

    62. Colmenero JD, Fernandez-Gallardo LC, Agundez JA et al.

    Possible implications of doxycycline-rifampicin interaction for treatment

    of brucellosis. Antimicrob Agents Chemother 1994; 38: 2798802.

    63. Wynalda MA, Hutzler JM, Koets MD et al. In vitro metabolism ofclindamycin in human liver and intestinal microsomes. Drug Metab

    Dispos 2003; 31: 87887.

    Review

    878