Journal Information
Vol. 34. Issue. 1.January 2014
Pages 0-138
Vol. 34. Issue. 1.January 2014
Pages 0-138
Full text access
Pharmacogenetics of tacrolimus: from bench to bedside?
Farmacogenética del tacrolimus: ¿del laboratorio al paciente?
Visits
15947
Beatriz Taviraa, Carmen Díaz-Corteb, Diego Coronelc, Francisco Ortegad, Eliecer Cotoe
a Servicio de Genética Molecular, Hospital Universitario Central de Asturias, Oviedo,
b Servicio de Nefrología, Hospital Universitario Central de Asturias. Departamento de Medicina, Universidad de Oviedo,
c Servicio de Nefrología, Hospital Universitario Central de Asturias, Oviedo,
d Servicio de Nefrología, Hospital Universitario Central de Asturias. Fundación Renal Iñigo Álvarez de Toledo, Madrid,
e Servicio de Genética Molecular, Hospital Universitario Central de Asturias, Oviedo. Departamento de Medicina, Universidad de Oviedo. Fundación Renal Iñigo Álvarez de Toledo, Madrid,
This item has received
Article information
Abstract
Full Text
Bibliography
Download PDF
Statistics
Figures (2)

El tacrolimus (Tac) es un inmunosupresor ampliamente usado para prevenir el rechazo en el trasplante renal. Los pacientes reciben una dosis inicial estándar y se miden los niveles sanguíneos, con ajuste de la dosis hasta alcanzar una concentración dentro del rango aceptado. Existe una gran variabilidad interindividual en las dosis necesarias para alcanzar ese nivel diana en sangre, y muchos pacientes requieren varias modificaciones de la dosis hasta alcanzarlo. Uno de los principales determinantes de estas diferencias es un polimorfismo del gen CYP3A5 que determina que alrededor del 80 % de los caucásicos sean metabolizadores lentos y requieran dosis menores que los metabolizadores rápidos. Se ha propuesto que los pacientes trasplantados reciban dosis iniciales de Tac con base en el genotipo CYP3A5. Para que este procedimiento fuese aceptado por los clínicos, deberían demostrarse sus ventajas frente al procedimiento actual, más allá de un menor tiempo para alcanzar la dosis óptima. Por ejemplo, menor tasa de nefrotoxicidad y rechazo o menor coste por necesitar, entre otros, menos modificaciones de la dosis de Tac y menos terapia de inducción con anticuerpos.

Palabras clave:
Trasplante renal
Palabras clave:
Farmacogenética
Palabras clave:
Citocromo P450
Palabras clave:
Tacrolimus
Palabras clave:
Farmacocinética

Tacrolimus (FK-506) is an immunosuppressant widely used to prevent kidney transplant rejection. Patients receive an initial standard dose and tacrolimus levels are measured in blood. If necessary, the dose is adjusted to reach a blood concentration within the accepted range. There is great interindividual variability in the dose required to achieve the target blood level, and many patients require multiple modifications of the dose to reach the range. One of the main determinants of these differences is a CYP3A5 gene polymorphism that determines that about 80% of Caucasians are poor metabolisers and require lower doses compared to the extensive metabolisers. It has been proposed that transplanted patients could receive an initial Tacrolimus dose based on the CYP3A5 genotype. This could reduce the time to achieve the optimal blood level, reducing the number of dose modifications. However, to be accepted by clinicians and translated to the clinical practice this adapted dose procedure should give additional advantages such as a significant reduction of the rates of nephrotoxicity and rejection, or a lower cost due to less dose modifications of Tacrolimus and less antibody induction therapy.

Keywords:
renal transplant
Keywords:
Pharmacogenetics
Keywords:
Cytochrome P450
Keywords:
Tacrolimus
Keywords:
Pharmacokinetics
Full Text

INTRODUCTION

 

The goal of immunosuppressive therapy is to prevent graft rejection. For this purpose drugs acting by different routes on the alloimmune response are administered, to achieve the highest degree of immunosuppression while minimising toxicity and other adverse effects, especially the development of tumours and infections.1 The recommended immunosuppressive therapy is an initial ‘induction’ therapy with antibodies (monoclonal or polyclonal), directed at preventing early rejection, and reduced doses of other immunosuppressives, especially calcineurin inhibitors, and maintenance immunosuppression with three drugs: Calcineurin inhibitors, such as tacrolimus or cyclosporine A, an inhibitor of purine synthesis (mycophenolate mofetil [MMF]) and steroids.2 Other immunosuppressive agents such as mTOR (mammalian rapamycin receptor) inhibitors (sirolimus and everolimus), although not usually used from the beginning, make it possible to use different combinations of immunosuppressants over time.

For most drugs each patient receives an initial dose based on variables such as weight and age. If drug blood levels can be measured, the dose may be adjusted to a blood value within an acceptable range. With regards to immunosuppressive drugs, a dose that is too low could cause organ rejection, and a dose that is too high could cause toxicity. Gene variations (mutations and polymorphisms) that encode proteins involved in drug metabolism may condition blood concentration of the pharmacologically active ingredient. Proteins encoded by these pharmacogenetically relevant genes may act at intestinal absorption level (thus affecting the amount of drug absorbed into the bloodstream) or liver, resulting in inactive or therapeutically active molecules. Finally, they can modify the drug or its metabolites to facilitate their elimination. For most genes involved in these processes there are polymorphisms that determine more or less active forms of proteins, so that, based on the corresponding genotypes, it would be possible to include each patient in a group with a better/worse therapeutic response. In this review we analyse the pharmacogenetics of tacrolimus, discussing the pros and cons of choosing a dose based on the patient's genotype.

 

TACROLIMUS: MECHANISM OF ACTION, PHARMACOKINETICS AND PHARMACODYNAMICS

 

Tacrolimus (FK-506) is an inhibitor of calcineurin, a phosphoprotein that promotes transcription of genes involved in growth and differentiation of CD4+ T lymphocytes. When tacrolimus binds to its intracellular target, there is inhibition of transcription of genes such as interleukin-2.3This immunosuppressant is characterised by its pharmacokinetic variability (interindividual) and its reduced therapeutic window.4 Therefore, continuous dose monitoring is required during the first weeks after transplantation in order to achieve adequate blood levels to prevent rejection (too low) or nephrotoxicity (too high).5,6 Although rapidly absorbed (in the intestine), its oral bioavailability is very low: of the total dose the patient receives only 25% would reach the blood stream. Maximum blood concentration is achieved within one to three hours after administration. Administration, either in two divided doses (Prograf ®, Astellas ®) or a single daily dose in the case of an extended release form (Advagraf ®) should be administered within 24 hours after transplantation, although depending on the donor’s features there may be differences between the centres, both in administration regimes and doses, as also combinations with other immunosuppressants. In the case of renal transplantation, the Symphony study recommends an initial oral dose of tacrolimus in the range of 0.10-0.30 mg/kg/day, depending on donor and recipient characteristics and associated immunosuppressive medication.7

In the intestine, tacrolimus is a substrate for P-glycoprotein (Pgp) (also known as MDR-1 -multidrug resistance 1-), encoded by the ABCB1gene.8-10This protein is found in the membrane of enterocytes and regulates the passage of substances from the interior of the cell into the extracellular space based on an adenosine triphosphate (ATP) mechanism. Once in the blood stream, tacrolimus reaches the liver where it is metabolised by CYP3A family cytochrome P450 reductases, primarily CYP3A5 and CYP3A4.11 Finally, its metabolites are excreted in urine and bile. The narrow therapeutic margin and large pharmacokinetic and pharmacodynamic variability between individuals make it necessary to measure tacrolimus blood levels and those of its metabolites to adjust the dose to achieve an appropriate circulating concentration.12 There are various measuring methods based on the use of antibodies (ELISA) or mass spectrometry. Monitoring trough levels of tacrolimus (C0) is critical during the first days after transplant and is performed immediately before each administration. In the case of renal transplant, the Symphony study recommends that C0 levels in whole blood values should be between 5 and 20ng/ml during the first weeks after transplant, and between 3 and 12ng/ml when the patient changes to maintenance therapy.7 Monitoring and dose adjustment should be performed when immunosuppressive regimen changes are introduced, or changes in the dosage form, or after administration of other drugs that could interfere with absorption and/or metabolism (such as clopidogrel and simvastatin). Dose modifications are also necessary when nephrotoxicity is suspected.13,14

 

PHARMACOGENETICS OF TACROLIMUS

 

The main determinant of interindividual variability in the dose of tacrolimus is the activity of cytochrome P450-3A5, encoded by CYP3A5 gene. Of all the polymorphisms of this gene, a change in a single nucleotide (SNP), known as CYP3A5*3 (SNP rs776746), is the main regulator of the optimum dose.15-19 This variant is in intron 3 of the CYP3A5 gene and affects pre-mRNA processing, so that there is not a perfect splicing between exons 3 and 4. The resulting mRNA will have an abnormal sequence, which is unstable and will be eliminated by the cell, so that it fails to synthesise protein.20 21 Consequently, carriers of two copies (homozygous) of the CYP3A5*3 allele do not have the protein (non-expressers), in contrast to carriers of at least one copy of the normal allele (designated CYP3A5*1).22-26 In summary, CYP3A5*3 would make it possible to classify patients according to phenotype as “slow metabolisers” (homozygous CYP3A5*3*3), “intermediate metabolisers” or “fast metabolisers” (heterozygous CYP3A5*1*3 and homozygous CYP3A5*1*1, respectively). The latter would require higher doses to achieve target levels of tacrolimus (Table 1).24-26 Other gene variants might be involved in the metabolism of tacrolimus, although the results obtained to date are not as conclusive as those seen with CYP3A5*3.27-30 Differences in allele frequencies of CYP3A5 gene between ethnic groups could explain the increased dose requirement in African Americans: while about 80 % of Caucasians are slow metabolisers (homozygous for the CYP3A5*3 allele), most black subjects are homozygous CYP3A5 *1*1 (fast metabolisers ).31.32These genetic differences could also explain the increased risk of rejection and nephrotoxicity among Afro-Americans.33

About 40 % of tacrolimus administered to a patient is metabolised by P450-3A4. Variants of gene CYP3A4 have been found that could affect the activity of this cytochrome and, therefore, basal drug levels/doses. In the case of tacrolimus CYP3A4*1B polymorphism (SNP rs2740574) in the promoter region of the gene (-392 A> G), perhaps resulting in higher protein levels, has been associated with a requirement for greater drug doses.22,34,35 In addition, some variants have recently been described that could affect tacrolimus metabolism, such as CYP3A4*22 polymorphism.36,37 However, other studies do not support a significant effect of this variant of CYP3A4 on doses.38,39 When assessing these discrepancies it is necessary to be aware that most carriers of CYP3A4*22 allele in turn are homozygous for CYP3A5*3 allele, which makes it difficult to quantify its effect on fast metabolisers for CYP3A5.39

ABCB1 gene encodes PgP, a protein that is expressed in many cell types and tissues and regulates intestinal absorption of many drugs. A relationship has been described between C3435T polymorphism (SNP rs1045642) within exon 26 of the gene and intestinal expression of this glycoprotein. Thus, this (and other polymorphisms) could condition dosage requirements for several drugs.40, 41 In the case of tacrolimus several studies found a significant relationship between dose requirement and C3435T genotype polymorphism.17, 28.42 However, this effect has not been confirmed by others, nor does it affect the dosage level in our patients.17,22,28,42-44 The expression of PgP in renal cells could condition the amount of tacrolimus reaching their interior and, therefore, toxicity levels. In this respect, a relationship has been described between donor C3435T genotype and the risk of nephrotoxicity.45 Although this study would open up a new pharmacogenetic pathway in renal transplant, it is based on a small number of patients, and should be confirmed in other populations.

 

CLINICAL APPLICATION OF TACROLIMUS PHARMACOGENETICS

 

The initial dose of tacrolimus was determined by variables such as patient age, weight or race.7,46,47 Some authors have proposed algorithms based on different variables to calculate the initial dose, including CYP3A5 genotype, which should be determined before transplant. In theory, the inclusion of genotype could reduce the number of dose adjustments and the time necessary to reach the desired blood level. For several drugs, the choice of a dose based on genotype is already a recommendation from the Food and Drug Administration and other agencies. In the case of tacrolimus, for the medical community to accept pharmacogenetics as a prescription criterion, patient genotype should provide significant advantages with regards to current procedures: for example, reducing the percentage of rejection or other adverse events, fewer modification of post-transplant doses; savings by reducing pharmaceutical expenditure or hospital stay, etc.   However, so far no studies have clearly demonstrated the advantages of giving a variable initial dose by genotype compared to the current method of a predetermined dose.

To demonstrate these advantages, a group of patients would have to be treated using an initial dose based on genotype to compare the results with a group of similar characteristics (age, sex, immunosuppressive scheme) to which tacrolimus is administered following traditional guidelines. This approach has been used in a study performed on 236 renal transplant patients divided into two groups: 120 patients (control group) received 0.2 mg/kg/day and 116 patients (adapted dose group) received a dose according to CYP3A5 genotype: 0.15 mg/kg/day in the case of slow metabolisers (homozygous for CYP3A5*3*3, n = 90) and 0.30 mg/kg/day in the case of fast metabolisers (CYP3A5*1 carriers, n = 26).48 However, patients began to receive tacrolimus as from post-transplant day 7 (not from the first day), and were kept under low induction therapy with basiliximab (Simulect®; Novartis, Basel, Switzerland) or anti-thymocyte globulin (Thymoglobulin®; Genzyme, Cambridge, MA) during that week. The two groups were compared based on the percentage of cases with a C0 value in the 10-15ng/l range after six oral doses of tacrolimus, time to achieve that range, number of dose modifications to achieve that range, number dialysis sessions to organ function, incidence of acute rejection (biopsy-proven ), loss of organ and death.48 The study concluded that after three days of treatment with tacrolimus there were a greater percentage of patients achieving target value C0 (43.2% vs. 29.1%, P=.03) in the adapted dose group, and these patients also required fewer dose modifications. However, the incidence of acute rejection or renal function values were similar between groups (Table 2). The sample size would be sufficient to achieve a statistical power of 80%.

Since measuring levels and adjusting the daily dose would make it possible for most patients to achieve target blood levels within the first two weeks after transplant, nephrologists may see little use in dose by genotype in the absence of other advantages. Since the study performed by Thervet et al. included patients treated with induction therapy and high doses of MMF, it is not surprising that there were no differences in the rate of acute rejection and other clinical variables. Therefore, it would be necessary to evaluate the role of dosage according to genotype in patients without induction therapy and administering tacrolimus immediately after transplant, instead of waiting for a week.49

 

CONCLUSIONS

 

While the role of CYP3A5 genotype as a determinant of the dose of tacrolimus to administer is indisputable, the usefulness of giving an initial dose based on genotype may depend on greater benefits than the mere reduction in the number of dose modifications. More trials are necessary to demonstrate that administration of adapted doses reduces nephrotoxicity and rejection, or saves money due to lower requirements for induction therapy or shortens hospitalisation times.

 

KEY CONCEPTS

 

  • Interindividual differences in the dose of tacrolimus are determined by a polymorphism of CYP3A5 gene.
  • CYP3A5 subjects, slow metabolisers, (80 % of Caucasians ) require lower doses.
  • Administration of an initial dose of tacrolimus based on CYP3A5 genotype could reduce the time and number of dose modifications necessary to achieve the target blood levels.
  • However, it is still necessary to determine whether this would reduce (among other variables) toxicity and rejection rates, the need for induction therapy or the expense of hospitalisation, among others advantages.
  •  

    ACKNOWLEDGEMENTS

     

    Study financed by the Renal Research Network of the Carlos III Institute of Health (RD12/0021/0012; RD12/0021/0018).

    Table 1. Daily dose of tacrolimus (median and range) according to CYP3A5 genotype in three series of kidney transplants

    Table 2. Summary of the study performed by Thervet et al.48

    Bibliography
    [1]
    Halloran F. Immunosuppressive drugs for kidney transplantation. N Engl J Med 2004;351(26):2715-29. [Pubmed]
    [2]
    Kasiske BL, Zeier MG, Craig JC, Ekberg H, Garvey CA, Green MD, et al. KDIGO clinical practice guideline for the care of kidney transplant recipients. Am J Transplant 2009;9 Suppl 3:S1-155.
    [3]
    Liu J, Farmer JD, Lane WS, Friedman J, Weissman I, Schreiber SL. Calcineurin is a common target of cyclophilin-cyclosporin A and FKBP-FK506 complexes. Cell 1991;66(4):807-15. [Pubmed]
    [4]
    Venkataramanan R, Swaminathan A, Prasad T, Jain A, Zuckerman S, Warty V, et al. Clinical pharmacokinetics of tacrolimus. Clin Pharmacokinet 1995;29(6):404-30. [Pubmed]
    [5]
    Staatz C, Taylor P, Tett S. Low tacrolimus concentrations and increased risk of early acute rejection in adult renal transplantation. Nephrol Dial Transplant 2001;16(9):1905-9. [Pubmed]
    [6]
    Mayer AD, Dmitrewski J, Squifflet JP, Besse T, Grabensee B, Klein B, et al. Multicenter randomized trial comparing tacrolimus (FK506) and cyclosporine in the prevention of renal allograft rejection: a report of the European Tacrolimusrolimus Multicenter Renal Study Group. Transplantation 1997;64(3):436-43.
    [7]
    Ekberg H, Tedesco-Silva H, Demirbas A, Vítko S, Nashan B, Gürkan A; ELITE-Symphony Study. Reduced exposure to calcineurin inhibitors in renal transplantation. N Engl J Med 2007;357(25):2562-75. [Pubmed]
    [8]
    Ueda K, Cornwell MM, Gottesman MM, Pastan I, Roninson IB, Ling V, et al. The mdr1 gene, responsible for multidrug-resistance, codes for P-glycoprotein. Biochem Biophys Res Commun 1986;141(3):956-62. [Pubmed]
    [9]
    Saeki T, Ueda K, Tanigawara Y, Hori R, Komano T. Human P-glycoprotein transports cyclosporin A and FK506. J Biol Chem 1993;268(9):6077-80. [Pubmed]
    [10]
    Hebert MF. Contributions of hepatic and intestinal metabolism and P-glycoprotein to cyclosporine and tacrolimus oral drug delivery. Adv Drug Deliv Rev 1997;27(2-3):201-14.
    [11]
    Shiraga T, Matsuda H, Nagase K, Iwasaki K, Noda K, Yamazaki H, et al. Metabolism of FK506, a potent immunosuppressive agent, by cytochrome P450 3A enzymes in rat, dog and human liver microsomes. Biochem Pharmacol 1994;47(4):727-35. [Pubmed]
    [12]
    Scholten EM, Cremers SC, Schoemaker RC, Rowshani AT, van Kan EJ, den Hartigh J, et al. AUC-guided dosing of tacrolimus prevents progressive systemic overexposure in renal transplant recipients. Kidney Int 2005;67(6):2440-7. [Pubmed]
    [13]
    Ekberg H, Tedesco-Silva H, Demirbas A, Vítko S, Nashan B, Gürkan A, et al. Reduced exposure to calcineurin inhibitors in renal transplantation. N Engl J Med 2007;357(25):2562-75. [Pubmed]
    [14]
    Stratta P, Quaglia M, Cena T, Antoniotti R, Fenoglio R, Menegotto A, et al. The interactions of age, sex, body mass index, genetics, and steroid weight-based doses on tacrolimus dosing requirement after adult kidney transplantation. Eur J Clin Pharmacol 2012;68(5):671-80. [Pubmed]
    [15]
    Dai Y, Hebert MF, Isoherranen N, Davis CL, Marsh C, Shen DD, et al. Effect of CYP3A5 polymorphism on tacrolimus metabolic clearance in vitro. Drug Metab Dispos 2006;34(5):836-47. [Pubmed]
    [16]
    Jacobson PA, Oetting WS, Brearley AM, Leduc R, Guan W, Schladt D, et al. Novel polymorphisms associated with tacrolimus trough concentrations: results from a multicenter kidney transplant consortium. Transplantation 2011;91(3):300-8. [Pubmed]
    [17]
    Macphee IA, Fredericks S, Tai T, Syrris P, Carter ND, Johnston A, et al. Tacrolimus pharmacogenetics: polymorphisms associated with expression of cytochrome p4503A5 and P-glycoprotein correlate with dose requirement. Transplantation 2002;74(11):1486-9. [Pubmed]
    [18]
    Thervet E, Anglicheau D, King B, Schlageter MH, Cassinat B, Beaune P. Impact of cytochrome p450 3A5 genetic polymorphism on tacrolimus doses and concentration-to-dose ratio in renal transplant recipients. Transplantation 2003;76:1233-5. [Pubmed]
    [19]
    Haufroid V, Wallemacq P, VanKerckhove V, Elens L, De Meyer M, Eddoure DC, et al. CYP3A5 and ABCB1 polymorphisms and tacrolimus pharmacokinetics in renal transplant candidates: guidelines from an experimental study. Am J Transplant 2006;6(11):2706-13. [Pubmed]
    [20]
    Kuehl P, Zhang J, Lin Y, Lamba J, Assem M, Schuetz J, et al. Sequence diversity in CYP3A promoters and characterization of the genetic basis of polymorphic CYP3A5 expression. Nat Genet 2001;27(4):383-91. [Pubmed]
    [21]
    Hustert E, Haberl M, Burk O, Wolbold R, He YQ, Klein K, et al. The genetic determinants of the CYP3A5 polymorphism. Pharmacogenetics 2001;11(9):773-9. [Pubmed]
    [22]
    Tavira B, Coto E, Diaz-Corte C, Ortega F, Arias M, Torres A, et al. Pharmacogenetics of tacrolimus after renal transplantation: analysis of polymorphisms in genes encoding 16 drug metabolizing enzymes. Clin Chem Lab Med 2011;49(5):825-33. [Pubmed]
    [23]
    23.Tsuchiya N, Satoh S, Tada H, Li Z, Ohyama C, Sato K, et al. Influence of CYP3A5 and MDR1 (ABCB1) polymorphisms on the pharmacokinetics of tacrolimus in renal transplant recipients. Transplantation 2004;78(8):1182-7. [Pubmed]
    [24]
    Quteineh L, Verstuyft C, Furlan V, Durrbach A, Letierce A, Ferlicot S, et al. Influence of CYP3A5 genetic polymorphism on tacrolimus daily dose requirements and acute rejection in renal graft recipients. Basic Clin Pharmacol Toxicol 2008;103(6):546-52. [Pubmed]
    [25]
    Satoh S, Saito M, Inoue T, Kagaya H, Miura M, Inoue K, et al. CYP3A5 *1 allele associated with tacrolimus trough concentrations but not subclinical acute rejection or chronic allograft nephropathy in Japanese renal transplant recipients. Eur J Clin Pharmacol 2009;65(5):473-81. [Pubmed]
    [26]
    MacPhee IA, Holt DW. A pharmacogenetic strategy for immunosuppression based on the CYP3A5 genotype. Transplantation 2008;85(2):163-5. [Pubmed]
    [27]
    Miura M, Satoh S, Kagaya H, Saito M, Numakura K, Tsuchiya N, et al. Impact of the CYP3A4*1G polymorphism and its combination with CYP3A5 genotypes on tacrolimus pharmacokinetics in renal transplant patients. Pharmacogenomics 2011;12(7):977-84. [Pubmed]
    [28]
    Roy JN, Barama A, Poirier C, Vinet B, Roger M. Cyp3A4, Cyp3A5, and MDR-1 genetic influences on tacrolimus pharmacokinetics in renal transplant recipients. Pharmacogenet Genomics 2006;16(9):659-65. [Pubmed]
    [29]
    Min SI, Kim SY, Ahn SH, Min SK, Kim SH, Kim YS, et al. CYP3A5 *1 allele: impacts on early acute rejection and graft function in tacrolimus-based renal transplant recipients. Transplantation 2010;90:1394-400. [Pubmed]
    [30]
    Hesselink DA, van Schaik RH, van der Heiden IP, van der Werf M, Gregoor PJ, Lindemans J, et al. Genetic polymorphisms of the CYP3A4, CYP3A5, and MDR-1 genes and pharmacokinetics of the calcineurin inhibitors cyclosporine and tacrolimus. Clin Pharmacol Ther 2003;74(3):245-54. [Pubmed]
    [31]
    Dirks NL, Huth B, Yates CR, Meibohm B. Pharmacokinetics of immunosuppressants: a perspective on ethnic differences. Int J Clin Pharmacol Ther 2004;42(12):701-18. [Pubmed]
    [32]
    Neylan JF. Racial differences in renal transplantation after immunosuppression with tacrolimus versus cyclosporine. FK506 Kidney Transplant Study Group. Transplantation 1998;65(4):515-23.
    [33]
    Padiyar A, Augustine JJ, Bodziak KA, Aeder M, Schulak JA, Hricik DF. Influence of African-American ethnicity on acute rejection after early steroid withdrawal in primary kidney transplant recipients. Transplant Proc 2010;42(5):1643-7. [Pubmed]
    [34]
    Kuypers DR, de Jonge H, Naesens M, Lerut E, Verbeke K, Vanrenterghem Y. CYP3A5 and CYP3A4 but not MDR1 single-nucleotide polymorphisms determine long-term tacrolimus disposition and drug-related nephrotoxicity in renal recipients. Clin Pharmacol Ther 2007;82(6):711-25. [Pubmed]
    [35]
    Amirimani B, Ning B, Deitz AC, Weber BL, Kadlubar FF, Rebbeck TR. Increased transcriptional activity of the CYP3A4*1B promoter variant. Environ Mol Mutagen 2003;42(4):299-305. [Pubmed]
    [36]
    Elens L, van Schaik RH, Panin N, de Meyer M, Wallemacq P, Lison D, et al. Effect of a new functional CYP3A4 polymorphism on calcineurin inhibitors' dose requirements and trough blood levels in stable renal transplant patients. Pharmacogenomics 2011;12(10):1383-96. [Pubmed]
    [37]
    Elens L, Bouamar R, Hesselink DA, Haufroid V, van der Heiden IP, van Gelder T, et al. A new functional CYP3A4 intron 6 polymorphism significantly affects tacrolimus pharmacokinetics in kidney transplant recipients. Clin Chem 2011;57(11):1574-83. [Pubmed]
    [38]
    Tavira B, Coto E, Diaz-Corte C, Alvarez V, Lopez-Larrea C, Ortega F. A search for new CYP3A4 variants as determinants of tacrolimus dose requirements in renal-transplanted patients. Pharmacogenet Genomics 2013;23(8):445-8. [Pubmed]
    [39]
    Santoro AB, Struchiner CJ, Felipe CR, Tedesco-Silva H, Medina-Pestana JO, Suarez-Kurtz G. CYP3A5 genotype, but not CYP3A4*1b, CYP3A4*22, or hematocrit, predicts tacrolimus dose requirements in Brazilian renal transplant patients. Clin Pharmacol Ther 2013;94(2):201-2. [Pubmed]
    [40]
    Hoffmeyer S, Burk O, von Richter O, Arnold HP, Brockmoller V, Johne A, et al. Functional polymorphisms of the human multidrug-resistance gene: multiple sequence variations and correlation of one allele with P-glycoprotein expression and activity in vivo. Proc Natl Acad Sci U S A 2000;97(7):3473-8. [Pubmed]
    [41]
    Zimprich F, Sunder-Plassmann R, Stogmann E, Gleiss A, Dal-Bianco A, Zimprich A, et al. Association of an ABCB1 gene haplotype with pharmacoresistance in temporal lobe epilepsy. Neurology 2004;63(6):1087-9. [Pubmed]
    [42]
    Op den Buijsch RA, Christiaans MH, Stolk LM, de Vries JE, Cheung CY, Undre NA, et al. Tacrolimus pharmacokinetics and pharmacogenetics: influence of adenosine triphosphate-binding cassette B1 (ABCB1) and cytochrome (CYP) 3A polymorphisms. Fundam Clin Pharmacol 2007;21(4):427-35. [Pubmed]
    [43]
    Fredericks S, Moreton M, Reboux S, Carter ND, Goldberg L, Holt DW, et al. Multidrug resistance gene-1 (MDR-1) haplotypes have a minor influence on tacrolimus dose requirements. Transplantation 2006;82(5):705-8. [Pubmed]
    [44]
    Ciftci HS, Ayna TK, Caliskan YK, Guney I, Bakkaloglu H, Nane I et al. Effect of MDR1 polymorphisms on the blood concentrations of tacrolimus in Turkish renal transplant patients. Transplant Proc 2013;45(3):895-900. [Pubmed]
    [45]
    Naesens M, Lerut E, de Jonge H, Van Damme B, Vanrenterghem Y, Kuypers DR. Donor age and renal P-glycoprotein expression associate with chronic histological damage in renal allografts. J Am Soc Nephrol 2009;20(11):2468-80. [Pubmed]
    [46]
    Wang P, Mao Y, Razo J, Zhou X, Wong ST, Patel S, et al. Using genetic and clinical factors to predict tacrolimus dose in renal transplant recipients. Pharmacogenomics 2010;11(10):1389-402. [Pubmed]
    [47]
    Passey C, Birnbaum AK, Brundage RC, Schladt DP, Oetting WS, Leduc RE, et al. Validation of tacrolimus equation to predict troughs using genetic and clinical factors. Pharmacogenomics 2012;13(10):1141-7. [Pubmed]
    [48]
    Thervet E, Loriot MA, Barbier S, Buchler M, Ficheux M, Choukroun G, et al. Optimization of initial tacrolimus dose using pharmacogenetic testing. Clin Pharmacol Ther 2010;87(6):721-6. [Pubmed]
    [49]
    van Gelder T, Hesselink DA. Dosing tacrolimus based on CYP3A5 genotype: will it improve clinical outcome? Clin Pharmacol Ther 2010;87(6):640-1. [Pubmed]
    Download PDF
    Idiomas
    Nefrología (English Edition)
    Article options
    Tools
    es en

    ¿Es usted profesional sanitario apto para prescribir o dispensar medicamentos?

    Are you a health professional able to prescribe or dispense drugs?