P- Glycoprotein- A Unique Transporter Pump

 

Manisha D. Patel1*, Jigna S. Shah1 and Parloop A. Bhatt2

1Shri Sarvajanik Pharmacy College, Mehsana.

2L. M. College of Pharmacy, Ahmedabad.

ABSTRACT:

P-Glycoprotein (P-gp)/MDR1 is the 170-kDa ABC drug transporter protein. It is a member of the ABC(ATP Binding Cassette)super family.It is involved in limiting the harmful exposure of toxins, drugs, and xenobiotics to the body by extruding them into the gastrointestinal tract, bile and urine. Drugs or substrates can cross into the cell membrane by simple diffusion, filtration, or by specialized transport. The first step in drug efflux is drug recognition by P-gp followed by ATP-binding and subsequent hydrolysis. Finally, the generated energy is utilized to efflux substrate outside the cell membrane through central pore. P-gp acts as a rate limiting step during various stages of pharmacokinetic of drug but mainly on the absorption. Different drugs have different impact on P-gp expression. Some drugs are substrate of p-gp while some are inducers or inhibitors of P-gp. Altered P-gp/MDR1 activity due to induction and/or inhibition can cause drug–drug interactions with altered drug pharmacokinetics and response of drug. P-gpmayshow gender basis differences and so drug effect in individuals.

 

KEYWORDS:

 

1. INTRODUCTION:

1.1 The P-glycoprotein gene family:

The absorption of drugs from the intestine is an important factor in determining their bioavailability. There are two types of transporters: efflux and influx. Efflux transporters include P-gp. These pump out drugs from the enterocytes into the lumen, thus decreasing their oral bioavailability. Influx transporters such as Organic anion transport proteinOATP enable enterocytes to uptake drugs from the lumen increasing their oral bioavailability.1

P-Glycoprotein (P-gp)/MDR1,the 170-kDa ABC drug transporter protein, a member of the ABC superfamily, is expressed as a result of transcription of the ABCB1/MDR1 gene.2 P-gp in higher mammals forms a small gene family, with two isoforms expressed in humans, and three isoformsin rodents. The Class I and III isoforms (human MDR1/ABCB1, mouse mdr1/Abcb1a and mdr3/Abcb1) are drug transporters, while the Class II isoforms (human MDR2/3/ABCB4, mouse mdr2/Abcb4) carry out export of phosphatidylcholine (PC) into the bile.3 Its intracellular localization, the P-gp transporter can limit cellular uptake of drugs from the blood circulation into the brain and placenta, kidney and from the gastrointestinal lumen into the enterocytes.4

 

1.2 Different binding site on P-gp:

Evidence suggests that P-gp/MDR1 has multiple binding sites divided evenly into two categories: transport and regulation.5,6 The N- and C-terminal halves of P-gp/ MDR1 contain binding sites, and these two sites may generate a single region in the overall protein structure.7The presence of multiple drug binding sites on P-gp/MDR1 could provide an explanation for the wide range of compounds known to interact with this protein. Previous studies have determined that there are two major substrate binding sites on P-gp/MDR1, namely at the TMD (Trans Membrane Domain) sites 5 and 6 and TMD sites 11 and 12.8[Figure 1.]


 

FIGURE 1: Structure of P-glycoprotein10

 

All binding sites of P-gp/MDR1 appear to be able to switch between high- and low-affinity conformations, along with modulators affecting their action from transporting sites. This might be caused by stimuli such as substrate binding and/or ATP hydrolysis.Conformational changes in P-gp/MDR1 have been demonstrated using 2H/H-exchange kinetics, proteolytic accessibility, and changes in antibody epitope recognition.9 There is a strong need to elucidate the molecular mechanism of substrate recognition, binding, and transport by P-gp/MDR1. This information might enable the development of novel drugs that bypass recognition by P-gp/MDR1. In addition, new drugs could be designed and generated that bind tightly to P-gp/MDR1 and inactivate it.10

 

1.3 ATP-binding domains:

Both NBDs (Nucleotide Binding Domain) are essential for proper functioning of P-gp/MDR1 and the activity of P-gp/MDR1 is entirely dependent on the presence of ATP.11 The ATP-binding domains act as ATPase, which converts ATP to ADP to provide the energy required for P-gp/MDR1 to pump substrates across membranes, often against steep concentration gradients.12

 

2.      Mechanism of action:

Drugs or substrates can cross into the cell membrane by simple diffusion, filtration, or by specialized transport, and the first step in drug efflux is drug recognition by P-gp followed by ATP-binding and subsequent hydrolysis. Finally, the generated energy is utilized to efflux substrate outside the cell membrane through central pore.10 The details of various steps are as follows.

 

2.1 Drugs/substrate recognition:

The major drug binding sites reside in or near TM6, TM12, TM1, TM4, TM10, and TM11.13,14 Amino acids in TM1 are involved in the formation of a binding pocket that plays a role in determining the suitable substrate/ drug size for P-gp, whereas Gly residues in TM2 and TM3 are important in determining substrate specificity. Hence, these transmembrane domains help in large to recognize substrates/drugs.15

 

2.2 ATP-binding and subsequent hydrolysis:

ATP binding and hydrolysis to the conformational changes that most probably alter the drug binding affinity and/or the accessibility of drug-binding sites.16

The global changes in P-gp conformation upon ATP binding, ATP hydrolysis,two ATP molecules are hydrolyzed for the transport of every substrate molecule and demonstrated two distinct roles for ATP hydrolysis in a single turnover of the catalytic cycle of P-gp, one in the transport of substrate and the other in effecting conformational changes to reset the pump for the next catalytic cycle.17

 

2.3 Efflux of substrate/drug through central pore:

The data reveal a major reorganization of the TM domains throughout the entire depth of the membrane on binding of nucleotide.18 Recently, it has been proposed that drug substrates first diffuse from the lipid bilayer into the drug-binding pocket through “gates” formed by TM segments at either end of the drug-binding pocket and then effluxes the substrate through the central pore of the transporter to outside the membrane.19

 

3.      Effect of p-glycoprotein on pharmacokinetic:

The contribution of P-gp in limiting intestinal absorption is determined by (i) affinity of drugs towards P-gp, (ii) the passive permeability of the drug molecules across the enterocytes, (iii) expression levels of P-gp and variability in expression levels along the gut, and (iv) physiological variables that influence the solubility and passive transport along the gut.20 [Figure 2.]

 

FIGURE 2: Drug Pumped Out from Intestinal Cells into Lumen of Gut[1]

 

Intestinal P glycoprotein is well known to limit the absorptionof xenobiotics and is believed to act as a cytotoxic defense mechanism.21 Oral administration is the most popular route for drug administration since dosing is convenient and non-invasive and many drugs are well absorbed by the gastrointestinal tract.

 

There are two principal routes by which compounds may cross the intestinal epithelium: paracellular or transcellular. A number of small hydrophilic, ionised drugs are absorbed via the paracellular pathway.22 However, absorption via this route is generally low since intercellular tight junctions restrict free transepithelial movement between epithelial cells. The transcellular absorption of hydrophilic drugs may be facilitated via specific carrier-mediated pathways by means of utilizing the same route of absorption followed by nutrients andmicronutrients. Many orally administered drugs are lipophilic and undergo passive transcellular absorption.23 Drugs that cross the apical membrane may be substrates for apical efflux transporters, which extrude compounds back into the lumen.24,25 These apical efflux transporters are principally ABC proteins such as P-gp and MRP2, and are ideally situated to act as the first line of defense by limiting the absorption of potentially toxic ‘foreign’ compounds.26

 

4.      Impact of drugs on p-glycoprotein:

4.1 Substrate of p-gp:

P-gp/MDR1 has an important role in drug resistance and a drug’s pharmacokinetics, a number of studies have been undertaken to elucidate the molecular attributes required for interaction between this protein and its small molecule substrates.10 The most P-gp/ MDR1 substrates possess two or three electron-donor groups with a fixed spatial separation of 2.5 and 4.6A ˚, respectively, with an increased number of these elements increasing the affinity for drug binding. Correspondingly, there are a high percentage of amino acids with hydrogen bonding donor side-chains in the transmembrane sequences of P-gp/MDR1 responsible for substrate recognition.27 Further studies have found that partitioning into thelipid membrane is the rate-limiting step for the interaction of a substrate with P-gp/MDR1and that dissociation of the P-gp–substrate complex is determined by the number andstrength of the hydrogen bonds formed between the substrate and P-gp/MDR1.28

 

Other studies have suggested that there might be some physicochemical characteristic features such as lipophilicity, hydrogen-bonding ability, molecular weight, and surface areathat contribute to a drug’s binding ability to P-gp/MDR1.29,30These compounds generally have a relatively highconcentration of electronegative groups such as oxygen, nitrogen and groups with anorbitalwithin an unsaturated system.Identification of molecular properties required for the recognition of compounds by P-gp/MDR1 as substrates is useful in rationally directing lead optimization towards the desiredP-gp/MDR1 interaction (e.g. efflux, inhibition or no interaction).31 Given that most anti-human immunodeficiency virus (HIV) drugs areP-gp/MDR1 substrates, modification of their chemical structures leading to less recognitionby P-gp/MDR1 as substrates will lead to improved penetration to the central nervous system(CNS) and, thus, a better antiviral profile can be achieved.10List of substrate are given in [Table 1.]

 

4.2 Inducers:

P-gp/MDR1 is induced not only by a number of chemical compounds, but also by physical stress, such as X-irradiation, ultraviolet light irradiation, and heat shock.27 P-gp/MDR1 induction by drugs is most clinically relevant in two areas of practice: the P-gp-mediated drug–drug interactions leading to altered drug absorption and oral bioavailability and the development of MDR of cancer cells to chemotherapeutic agents.32 The extent and clinical consequence of P-gp/MDR1 induction depend on factors associated with the inducer, patient and co-administered drug. P-gp/MDR1 over-expression and consequent MDR causes a major problem in cancer chemotherapy.33 List of inducers are given in [Table 1.]

 

4.3 Inhibitor:

The potential therapeutic uses of these P-gp/MDR1 inhibitors is that co-administration with existing chemotherapy drugs (providing co-administration does not cause other interactions) will help to negate cancer cell MDR caused by P-gp/MDR1.34 Most P-gp/MDR1 binding inhibitors share some common chemical features, such as aromatic ring structures, a tertiary or secondary amino group and high lipophilicity30 although P-gp/MDR1 substrates as a whole have varying classes of inhibitory action. Some P-gp/MDR1 inhibitors are also referred to as chemosensitzers due to their ability to reduce chemotherapy drug resistance.35 The main ways in which an inhibitor of P-gp/MDR1 can exert its activity are either by being a very high-affinity substrate for P-gp/MDR1 and binding non-competitively (thus not allowing other drugs to bind), or by being efficient inhibitors of ATP hydrolysis either at the ATP binding site or by inhibiting protein kinase C which is involved with ATP coupling to P-gp/MDR1.36List of inhibitors are given in [Table 1.]

 

Table 1: List of Substrate, Inhibitors and Inducers of P-gp[10]

Substrate

Inhibitor

Inducers

Actinomycin

Daunorubicin

Digoxin

Diltiazem

Domperidone

Indonavir

Irinotecan

Ketoconalzole

Losartan

Phynobarbital

Phenytoin

Rifampin

Ritonavir

Tetracycline

Topotecan

Quinidine

Vincristine

Verapamil

Amiodarone

Astemizole

Atorvastatin

Bromocriptine

Carvedilol

Cyclosporine

Diltiazem

Dipyridamole

Disulfiram

Erythromycine

Fluoxetine

Itraconazole

Progesterone

Quinidine

Reserpine

Terfinadine

 

 

Amiodarone

Colchicine

Diltiazem

Insulin

Methotrexate

Midazolam

Morphine

Nelfinavir

Nifedipin

Phenothiazine

Phenytoin

Probenacid

Reserpine

Rifampicin

Ritonavir

St John’s wort

Verapamil

Yohimbine

 

5.      Gender and Age Related Differences in P-Glycoprotein Expression:

Some authors suggest thatactivity is one-third to one-half lower in livers obtainedfrom females compared with males,37 however inother in vitro study there were no difference in the expressionof P-gp.38 It is not easy to establish the role ofP-gp in the gender-related differences in the oral pharmacokineticsof drugs for the above mentioned that CYP3A4and P-gp share substrates.39,40Since several factors may contribute to gender differencesin the absorption of drugs, the final result will dependon the characteristics of the compound and the extentof gender differences of each of the factors described.41

P-glycoprotein and CYP3A4 act in concert to reduce absorption of xenobiotics along the gastrointestinal tractand increase drug elimination from the liver.In addition, p-glycoprotein increases fecal elimination by extruding drug into bile.42Genetic deficiencies in p-glycoprotein reduce drug excretion into bile and increase drug half-life of p-glycoprotein–dependent drugs.43Intracellular levels of p-glycoprotein substrate drugs are increased when p-glycoprotein is blocked or geneticallyreduced42,44-46Reduced p-glycoprotein predisposes to greater competitive intracellular drug interactionsfor CYP3A4.44 CYP3A4 in humans is generally the same in females and males, but hepatic p-glycoprotein is 2.4-fold lower in females.44,47Reduction inp-glycoprotein in females relative to males will reduceelimination and prolong drug half-life. Reduction in glycoprotein will lead to accumulate of these glycoproteinsubstrate drugs in brain, heart, liver, and gastrointestinal tract.45,46,48,49The end result is a greater risk for myelosuppression and gastrointestinal toxicity in females and prolonged drug elimination.50 In summary, sex differences in p-glycoprotein will influence intracellular levels of p-glycoprotein substrate drugs; increase competitive drug interactions and delay drug elimination.51 As a result; females will experience greater drug toxicity with p-glycoprotein substrate drugs.52 Females have been shown to be a risk factor for clinically relevant adverse drug reactions with a 1.5 to 1.7-fold greater risk of developing an adverse drug reaction compared to male patients.53

 

The effect of age on P-Glycoprotein expression and functionin the Fischer-344 ratwasexamined in five different tissues. An age-related increase inP-gp expression was evident in the liver and lymphocytes,whereas a reduction was observed in the kidney. In intestinalcells and endothelial cells of the BBB, there was no apparentchange with age. Of all tissues examined, P-gp expression inthe intestine and the BBB displayed the greatest variability.54The digoxin pharmacokinetics were investigated in the elderly healthy subjects and compared with those of the younger patients. It showed no statistically significant differences in comparison with the younger age group. Even statistical differences in the digoxin pharmacokinetics between young healthy women were not seen.55Some study in humans suggests that cerebral P-gp function decreases with age and it may cause neurodegenerative disease.56

 

6.      CONCLUSION:

Literature studies clearly indicate the importance of understanding P-gp/MDR1 in depth. P-gp forms a functional barrier whichrestrictsaccess of various pharmacologic agents, xenobiotic excretion and cause cancer cell resistance, a thorough understanding of its mechanisms, structure, andfunction can lead to a greater understanding of drug therapy in a number of differentareas due to the great number of known substrates, inducers, and inhibitors. Currently inthe area of pharmacy practice, knowledge of P-gp/MDR1 and its potential to impactdrug dosing regimens is greatly under-appreciated. The ability of transport proteins including P-gp/MDR1, BCRP, and MRPs to reduce oral bioavailability and alter tissue distribution has obvious implications for drug design. Gender and age basis differences are also seen in the expression of P-gp.Several factors may contribute to gender differencesin the absorption of drugs with P-gp. But females are more prone to adverse drug reaction than males. Age related differences are seen in expression of p-gp is differ in different tissues.

 

7.      REFERENCES:

1)       Chen J, Raymond K.The role of CYP3A4 and p-glycoprotein infood-drug and herb-drug interactions, Australian pharmacist 2006, 25(9), 732-738.

2)       Croop JM. P-glycoprotein structure and evolutionary homologies.Cytotechnology 1993,12:1-32.

3)       Lincke CR, Broeks A, Plasterk RH, Borst P. The expression of two P-glycoprotein (P-gp) genes in transgenic Caenorhabditiselegans is confined to intestinal cells. EMBO J1993, 12, 1615-1620.

4)       Lin JH.Drug–drug interaction mediated by inhibition and induction ofP-glycoprotein.Advanced Drug Delivery Reviews2003, 55, 53–81.

5)       Martin C, Berridge G, Higgins CF, Mistry P, Charlton P, Callaghan R. Communication between multiple drug binding sites on P-glycoprotein. Molecular Pharmacology2000a.58, 624–632.

6)       Shilling RA, Venter H, Velamakanni S, Bapna A, Woebking B, Shahi S, Van Veen HW. New light on multidrug binding by an ATP-binding-cassette transporter. Trends in Pharmacology Sciences 2006, 27,195–203.

7)       Loo TW, Bartlett MC, Clarke DM. Simultaneous binding of two different drugs in the binding pocket of the human multidrug resistance P-glycoprotein. J Biol.Chem. 2003a, 278, 39706–39710.

8)       Wang RB, Kuo CL, Lien LL, Lien EJ. Structure–activity relationship: Analyses of p-glycoprotein substrates and inhibitors. JClin. Pharm. Ther.2003, 28, 203–228.

9)       Sonveaux N, Vigano C, Shapiro AB, Ling V, Ruysschaert JM. Ligand-mediated tertiary structure changes of reconstituted P-glycoprotein. J Biol. Chem.1999, 274, 17649–17654.

10)    Zhou SF.Structure, function and regulation of P-glycoprotein andits clinical relevance in drug disposition, Xenobiotica 2008. 38(7–8): 802–832.

11)    Romsicki Y, Sharom F. The ATPase and ATP-binding functions of P-glycoprotein modulation by interaction with defined phospholipids. Eur J Biochem1998, 256, 170–178.

12)    Buxbaum E. Co-operating ATP sites in the multiple drug resistance transporter Mdr1. Eur J Biochem1999,265, 54–63.

13)    Ambudkar SV, Dey S, Hrycyna. Biochemical, cellular and pharmacological aspects of multidrug transporter. Annu Rev PharmacolToxicol 1999, 39,361–98.

14)    Ueda K, Taguchi Y, Morishima M. How does P-glycoprotein recognize its substrates?Semin Cancer Biol 1997.8, 151–9.

15)    Tandon V, Kapoor B.Bano G, Gupta S, Gillani Z, Gupta S, Kour D.P- glycoprotein: pharmacological relevance. Indian J Pharmacol 2006, 38(1),13-24.

16)    Van Veen HW, Higgins CF,Konings WN. Molecular basis of multidrug transport by ATP-binding cassette transporters: a proposed two-cylinder engine model, J. Mol. Microbiol. Biotechnol 2001, 3, 185-192.

17)    Sauna ZE, Ambudkar SV. Evidence for a requirement for ATP hydrolysis at two distinct steps during a single turnover of the catalytic cycle of human P-glycoprotein. ProcNatlAcadSci 2000, 97, 2515–20.

18)    Mark FR, Alhaji BK, Richard C, Christopher FH, Robert CF. Three-dimensional structures of the mammalian multidrug resistance P-glycoprotein demonstrates major conformational changes in the transmembrane domains upon nucleotide binding. J BiolChem 2003,278, 8294–99

19)    Loo TW, Clarke DM. Do drug substrates enter the common drug-binding pocket of P-glycoprotein through “gates”? BiochemBiophys Res Commun 2005,329,419–22.

20)    Varma MVS, PerumalOP,Panchagnula R. Functional role of P-glycoprotein in limiting peroral drug absorption: optimizing drug delivery.Current Opinion in Chemical Biology 2006, 10, 367–373.

21)    Cortes-Selva F, Munoz-Martinez F, Ilias A, Jimenez AI, Varadi A, Gamarro F, et al. Functional expression of a multidrug P-glycoprotein transporter of Leishmania. BiochemBiophys Res Commun 2005,329,502–7.

22)    Hayashi M, Tomita M, Awzu S.Transcellular and paracellular contribution to transport processes in the colorectal route. Adv. Drug Deliv. Rev. 1997, 28, 191–204.

23)    Hunter J, Hirst BH. Intestinal secretion of drugs. The role of P-glycoprotein and related drug efflux systems in limiting oral dru absorption. Adv. Drug Del. Rev.1997, 25, 129–157.

24)    Evers R, Kool M, Van Deemter L, Janssen H, Calafat J, Oomen LCJM, Paulusma CC, Oude Elferink, RPJ, Baas F, Schinkel AH, Borst P. Drug export activity of the human canalicularmultispecific organic anion transporter in polarised kidney MDCK cells expressing cMOAT (MRP2) cDNA. J. Clin. Invest. 1998, 101, 1310–1319.

25)    Fromm MF, Kauffman HM, Fritz P, Burk O, Kroemer HK, Warzok RW, Eichelbaum,M, Siegmund W, Schrenk D. The effect of rifampin treatment on intestinal expression of human MRP transporters. Am. J. Pathol. 2000, 157, 1575–1580.

26)    Watkins PB. Drug metabolism by cytochromes P450 in the liver and small bowel. Gastrointest. Pharmacol. 1992, 21, 511–526.

27)    Seelig A. A general pattern for substrate recognition by P-glycoprotein. Eur. J. Biochem.1998,251,252–261.

28)    Seelig A, Landwojtowicz E. Structure–activity relationship of P-glycoprotein substrates and modifiers. Eur. J.Pharm. Sci.2000,12,31–40.

29)    Bain L, McLachlan J, LeBlanc G. Structure–activity relationships for xenobiotic transport substrates and inhibitory ligands of P-glycoprotein. Environmental Health Perspectives1997, 105,812–818.

30)    Wang RB, Kuo CL, Lien LL, Lien EJ. Structure–activity relationship: Analyses of p-glycoprotein substrates and inhibitors. J. Clin. Pharm. Thera.2003, 28,203–228.

31)    Raub TJ. P-glycoprotein recognition of substrates and circumvention through rational drug design. Mol.Pharmacol.2006, 3,3–25.

32)    Machado CG, Calado RT,Garcia AB, Falcăo RP.Age-related changes of the multidrug resistance P-glycoprotein function in normal human peripheral blood T lymphocytes, Br J. Med. Boil. Research 2003,36, 1653-1657.

33)    Lesliea EM, Deeleyb RG, Coleb PC.Multidrug resistance proteins: role of P-glycoprotein, MRP1, MRP2, and BCRP (ABCG2) in tissue defense. Toxicol. Appl. Pharmacol. 2005,204, 216– 237.

34)    Dean M.The human ATP-binding cassette (ABC) transporter superfamily. National Center for Biotechnology Information (NCBI) 2002, National library of Medicine, Bethesda, MD, USA.

35)    Shiraga K, Sakaguchi K, Senoh T, Ohta T.Modulation of doxorubicin sensitivity by cyclosporine A in hepatocellular carcinoma cells and their doxorubicin-resistant sublines. J. Gastroenterol. Hepatol.2001, 16,460–466.

36)    ChaudharyP, Roninson I. Induction of multidrug resistance in human cells by transient exposure to different chemotherapeutic drugs. J. Natl. Caner. Inst.1993,85,632–639.

37)    Schuetz EG,FuruyaKN,Schuetz JD. “Interindividual Variation in Expression of P-Glycoprotein in Normal Human Liver and Secondary Hepatic Neoplasms,” Journal of Pharmacology and ExperimentalTherapeutics 1995, 275(2), 1011-1018.

38)    Wolbold R, Klein K, Burk O, et al. “Sex is a Major Determinant of CYP3A4 Expression in Human Liver,” Hepatology, 2003, 8(4), 978-988.

39)    Rabbaa L, Dautrey S, Colas-Linhart N, et al. “Intestinal Elimination of Ofloxacin Enantiomers in the Rat: Evidence of a Carrier-Mediated Process,” AntimicrobialAgents and Chemotherapy1996, 40(9), 2126-2130.

40)    Aminimanizani A, Beringer P, Jelliffe R.Comparative Pharmacokinetics and Pharmacodynamics of the Newer FluoroquinoloneAntibacterials.Clinical Pharmacokinetics 2001, 40(3), 169-187.

41)    Carrasco-Portugal MC, Flores-Murrieta FJ. Gender Differences in the Pharmacokinetics of Oral Drugs.Pharmacology and Pharmacy, 2011, 2, 31-41.

42)    Benet LZ, Cummins CL. The drug efflux: Metabolism alliance— Biochemical aspects. Adv Drug Deliv Rev2001, 50, S3-S11.

43)    Van Asperen J,Schinkel AH, Beijnen JH, et al. Altered pharmacokinetics of vinblastine in MDR lA P-glycoprotein deficient mice. J Natl Cancer Inst 1996, 88, 994-999.

44)    Benet LZ, Cummins CL, Wu CY. Unmasking the dynamic interplay between efflux transporters and metabolic enzymes. Int J Pharm2004, 277, 3-9.

45)    VanAsperen J, van Tellingen O, Tijssen F, et al. Increased accumulation of doxorubicin and doxorubicinol in cardiac tissue of mice lacking mdr la P-glycoprotein. Br J Cancer1999,79, 108-113.

46)    VanAsperen J, van Tellingen O, Schinkel AH, et al: Comparative pharmacokinetics of vinblastine after a 96-hour continuous infusion in wildtype mice and mice lacking mdr la P-glycoprotein. J PharmacolExpTher1999,289, 329-333.

47)    Schuetz EG, Furuya KN, Scheutz JD.Interindividual variation in expression of P-Glycoprotein in normal human liver and secondary hepatic neoplasms. J PharmacolExpTher1995, 275, 1011-1018.

48)    Cummins CL, Wu C, Benet LZ. Sex-related differences in the clearance of cytochrome P450 3A4 substrates may be caused by P-glycoprotein. ClinPharmacolTher 2002,72, 474-489.

49)    Yokogawa K, Takahashi M, Tamai I, et al. P-glycoprotein-dependent disposition kinetics of tacrolimus: Studies in mdr la knockout mice. Pharm Res 1999, 16, 1213-1218.

50)    Meyers PA, Schwartz CL, Krailo M, et al. Gender Differences in p-Glycoprotein: Drug Toxicity and Response,JClinOncol 2005, 23, 2004-2011.

51)    Chiou WL, Chung SM, Wu, Ta C: Potential role of P-Glycoprotein in affecting hepatic metabolism of drugs. Pharm Res 2000, 17, 903-905.

52)    Singh S, Parulekar W, Murray N, et al: Influence of sex on toxicity and treatment outcome in small-cell lung cancer. J ClinOncol 2005, 23, 850-856.

53)    Kando JC, Yonkers KA, Cole JO. “Gender as a Risk Factor for Adverse Events to Medications,” Drugs1995, 50(1), 1-6.

54)    Warrington JS, Greenblatt DJ, and Moltke LL. The Effect of Age on P-Glycoprotein Expression and Function in the Fischer-344 Rat. The journal of pharmacology and experimental therapeutics 2004, 309(2):730–736.

55)    Larsen  UL, Olesen LH, Nyvold CG, Eriksen J, Jakobsen P, Řstergaard M, Autrup H and Andersen V. Human intestinal P-glycoprotein activity estimated by the model substrate digoxin. Scand J Clin Lab Invest 2007, 67, 123–134.

56)    Bauer M, Karch R, Neumann F, Abrahiml A, Kletter K, Mueller M, Langer O. Decreased blood-brain barrier P-glycoprotein function with aging. J Nucl Med. 2009, 50 (2), 606.

.

Received on 17.06.2011

Accepted on 29.06.2011     

© A&V Publication all right reserved

Research J. Pharmacology and Pharmacodynamics. 3(5): Sept –Oct. 2011, 241-245