Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 + 1327 word(s) 1327 2021-07-01 04:29:10 |
2 The format is correct Meta information modification 1327 2021-07-28 11:10:17 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Tziastoudi, M. Pharmacogenetics in Chronic Kidney Disease. Encyclopedia. Available online: https://encyclopedia.pub/entry/12502 (accessed on 29 March 2024).
Tziastoudi M. Pharmacogenetics in Chronic Kidney Disease. Encyclopedia. Available at: https://encyclopedia.pub/entry/12502. Accessed March 29, 2024.
Tziastoudi, Maria. "Pharmacogenetics in Chronic Kidney Disease" Encyclopedia, https://encyclopedia.pub/entry/12502 (accessed March 29, 2024).
Tziastoudi, M. (2021, July 27). Pharmacogenetics in Chronic Kidney Disease. In Encyclopedia. https://encyclopedia.pub/entry/12502
Tziastoudi, Maria. "Pharmacogenetics in Chronic Kidney Disease." Encyclopedia. Web. 27 July, 2021.
Pharmacogenetics in Chronic Kidney Disease
Edit

Chronic kidney disease (CKD) is an important global public health problem due to its high prevalence and morbidity. Although the treatment of nephrology patients has changed considerably, ineffectiveness and side effects of medications represent a major issue. Pharmacogenetics could fill this gap.

genetic association chronic kidney disease meta-analysis pharmacogenetics

1. Introduction

Chronic kidney disease (CKD) continues to constitute a global health burden. It is known that CKD elevates the risk of cardiovascular disease, kidney failure, and other complications [1][2][3]. According to the Kidney Disease Outcomes Quality Initiative (KDOQI) classification, CKD is defined as kidney damage or glomerular filtration rate (GFR) < 60 mL/min/1.73 m2 for 3 months or more, irrespective of the cause [4]. Although significant progress has been made in the treatment of nephrology patients with both conservative therapies and dialysis or transplantation, the emergence of drug-related problems such as ineffectiveness and side effects represents a major issue [5]. Pharmacogenetics could fill this gap [6].
Over the last 30 years, new drugs have been introduced to treat major kidney diseases, slow down the progression of CKD, and reduce the development of clinical complications associated with dialysis and kidney transplantation [7]. The use of different combinations of potent immunosuppressive drugs in transplant patients (calcineurin inhibitors, mammalian target of rapamycin inhibitors (mTORs), corticosteroids) have significantly improved the treatment of various renal disorders, and the short- and long-term pharmacological management of renal graft recipients [8].
In general, currently approved immunosuppressive drugs for maintenance therapy include calcineurin inhibitors (cyclosporine (CsA), tacrolimus (TAC)), mTOR inhibitors (sirolimus (SIR), everolimus), antiproliferatives (azathioprine (AZA) and mycophenolic acid (MPA)) and biologic drugs (belatacept) [9]. Differences between individuals regarding the efficacy and safety of immunosuppressive treatment are determined to some extent by genetic factors. For example, a common nonfunctional splicing variant, CYP3A5*3 (rs776746), determines TAC doses [10]. More specifically, patients with the CYP3A5*3/*3 genotype require less TAC to reach target concentrations compared with cytochrome P450 family 3 subfamily A member 5 (CYP3A5) CYP3A5*1 allele carriers [11]. Tacrolimus pharmacokinetic and pharmacodynamic variability is also attributed to ATP binding cassette subfamily B member 1 (ABCB1) variants: 1236C > T (rs1128503), 2677G > T/A (rs2032582), and 3435C > T (rs1045642) [12][13]. In addition, another example of the implication of pharmacogenetics in nephrology constitutes the thiopurine S-methyltransferase (TPMT) gene [14]. Many lines of evidence have reported that genetic variants located in the TPMT gene affect AZA metabolism and patients with low activity (10% prevalence) or absent activity (0.3% prevalence) are at risk of myelosuppression [15][16]. Among 20 variant alleles (TPMT *2-*18) identified to date, mutant alleles TPMT*2 and TPMT*3 explain more than 95% of defective gene activity [8][17].

2. Pharmacogenetic Studies in Patients with Chronic Kidney Disease

With regard to the ABCB1 gene and the three polymorphisms harbored in it, the ABCB1 1236 C > T polymorphism was statistically significant in the studies with prednisolone (PRE) and mycophenolate (MMF). The ABCB1 2677 G > T polymorphism was also statistically significant in the analyses for PRE, whereas the ABCB1 3435 C > T polymorphism was statistically significant in the analyses for MMF and cyclosporine (CsA).

Regarding the genes encoding interleukins, the IL-10 -592 C > A polymorphism in all genetic models and --819 C > T in the dominant and the additive model in the CsA analyses were statistically significant. Another statistically significant polymorphism was the ITPA 94 C > A polymorphism in the recessive model in azathioprine (AZA) analyses. In addition, a statistically significant polymorphism was the MIF -173 G > C polymorphism in PRE analyses in all genetic models. Statistically significant results were also obtained for the TNF-308 G > A polymorphism in the recessive and additive models in PRE analyses.
Regarding heterogeneity control, statistically significant heterogeneity was observed among the studies regarding the CYP2C19*2 polymorphism in the main analysis for cyclophosphamide (CYC): for the TPMT 1 vs. polymorphism, 3C, MIF -173 G > C, Il-6 C-174G for PRE; for TPMT 1 vs. polymorphisms, 3C, ABCB1 1236 C > T, 2677 G > T, for CsA; for TPMT 1 vs. polymorphism 3C for AZA. For tacrolimus (TAC), a statistically significant heterogeneity was observed for polymorphisms ABCB1 2677 G > T and 3435C > T. Due to the statistically significant heterogeneity, the above results should be interpreted with caution, the majority of which are non-statistically significant.
Variants ABCB1 (1236 C > T, 2677 G > T, 3435 C > T), IL-10 (-592 C > A, -819 C > T), ITPA (94 C > A), MIF (-173 G > C), and TNF (-308 G > A) gave significant results, suggesting the contribution of these loci to different responses to treatment in patients with CKD.
However, only TPMT has been included in the table of pharmacogenetics biomarkers in drug labeling of the U.S. Food and Drug administration (FDA) for the treatment of AZA [18]. More specifically, homozygous TPMT-deficient patients experience severe myelosuppression. For the other variants, the results are not so robust.
Regarding calcineurin inhibitors, the effects of ABCB1 3435C > T, 1236C > T, and 2677G > T/A SNPs on the pharmacokinetics of CsA and TAC remain uncertain, with conflicting results. Genetic linkage between these three genotypes suggests that the pharmacokinetic effects are complex and unrelated to any ABCB1 polymorphism. In contrast, it is possible that these polymorphisms may exert a small but combined effect. Any effect is likely to be in addition to the effects of CYP3A5 6986A > G SNP [12].
With regard to the CYP3A5 6986A > G variant, eight studies [19][20][21][22][23][24][25][26] included patients under treatment with pulse CYC, steroids, calcineurin inhibitors, and AZA/SIR. In contrast to CsA, a strong relationship between the CYP3A5 6986A > G SNP and TAC pharmacokinetics was demonstrated in kidney, heart, and liver transplant recipients, as well as in healthy volunteers [12]. Several recent studies have reported an approximate halving of the TAC C0/dose and doubling of the tacrolimus dose requirements in CYP3A5 expressers compared to that in CYP3A5 non-expressers [27][25][28][29][30][31][32].
However, studies with a small number of patients may be responsible for many conflicting results to date. The low frequency of some alleles, such as CYP3A4*1B allele, may not have been sufficient in many cases to detect a difference. In addition, the influence of ethnicity may play a role, as mutated genotypes are often more common in specific ethnic groups. However, even in the same ethnic group, for example in Caucasians, the frequencies of the studied polymorphisms differ. For instance, Caucasians present a minor allelic frequency around 50% regarding the ABCB1 1236C > T polymorphism, whereas the studied TPMT allele frequency polymorphisms range from 0.2–5.5% in Caucasians. Although the genotype itself, rather than the underlying ethnicity, should theoretically detect any differences, it is possible that indeterminate genetic differences (for example, co-inherited SNPs) among Africans, Caucasians, and Asians contribute to significant variables. In addition, the associations presented in these meta-analyses resulted from pooling a relatively small number of studies and patients with large heterogeneity between studies. Furthermore, the impact of effect modifiers such as age and the pre-treatment cytogenetic and molecular genetic findings was not considered as the individual studies did not provide the relevant data. Indeed, we have not included the analyses of interactions of age and comorbidity in the meta-analysis because these details were not included in the available data. It would be very interesting if future pharmacogenetic studies included this type of data in the analysis. The present systematic review and meta-analysis included studies that varied in terms of treatment and primary cause of CKD, as well as racial descent. Thus, the results should be interpreted with caution. Future studies with more homogenous studies will shed light on the pharmacogenetics in CKD. Thus, lack of significant association in the remaining gene variants does not exclude the possibility of an association.
Last but not least, epigenetic changes in drug metabolizing enzymes, nuclear receptors, and transporters are associated with individual drug responses and acquired multidrug resistance [33]. Consequently, pharmacoepigenetics could provide an explanation for why patients with the same genotype respond differently to therapy with a specific medication. Unrelated to epigenetics, inflammation can significantly influence the extent of CYP suppression, thus contributing to intra- and interindividual variability to drug exposure [34].

References

  1. Levey, A.S.; Atkins, R.; Coresh, J.; Cohen, E.P.; Collins, A.J.; Eckardt, K.-U.; Nahas, M.E.; Jaber, B.L.; Jadoul, M.; Levin, A.; et al. Chronic Kidney Disease as a Global Public Health Problem: Approaches and Initiatives—a Position Statement from Kidney Disease Improving Global Outcomes. Kidney Int. 2007, 72, 247–259.
  2. Coresh, J.; Selvin, E.; Stevens, L.A.; Manzi, J.; Kusek, J.W.; Eggers, P.; Van Lente, F.; Levey, A.S. Prevalence of Chronic Kidney Disease in the United States. JAMA 2007, 298, 2038–2047.
  3. Sarnak, M.J.; Levey, A.S.; Schoolwerth, A.C.; Coresh, J.; Culleton, B.; Hamm, L.L.; McCullough, P.A.; Kasiske, B.L.; Kelepouris, E.; Klag, M.J.; et al. Kidney Disease as a Risk Factor for Development of Cardiovascular Disease: A Statement from the American Heart Association Councils on Kidney in Cardiovascular Disease, High Blood Pressure Research, Clinical Cardiology, and Epidemiology and Prevention. Circulation 2003, 42, 1050–1065.
  4. Moe, S.; Drüeke, T.; Cunningham, J.; Goodman, W.; Martin, K.; Olgaard, K.; Ott, S.; Sprague, S.; Lameire, N.; Eknoyan, G. Definition, Evaluation, and Classification of Renal Osteodystrophy: A Position Statement from Kidney Disease: Improving Global Outcomes (KDIGO). Kidney Int. 2006, 69, 1945–1953.
  5. Adams, S.M.; Crisamore, K.R.; Empey, P.E. Clinical Pharmacogenomics: Applications in Nephrology. Clin. J. Am. Soc. Nephrol. Clin. J. Am. Soc. Nephrol. 2018, 13, 1561–1571.
  6. Roden, D.M.; McLeod, H.L.; Relling, M.V.; Williams, M.S.; Mensah, G.A.; Peterson, J.F.; Van Driest, S.L. Pharmacogenomics. Lancet Lond. Engl. 2019, 394, 521–532.
  7. Halloran, P.F. Immunosuppressive Drugs for Kidney Transplantation. N. Engl. J. Med. 2004, 351, 2715–2729.
  8. Zaza, G.; Granata, S.; Sallustio, F.; Grandaliano, G.; Schena, F.P. Pharmacogenomics: A New Paradigm to Personalize Treatments in Nephrology Patients. Clin. Exp. Immunol. 2010, 159, 268–280.
  9. Kurzawski, M.; Droździk, M. Pharmacogenetics in Solid Organ Transplantation: Genes Involved in Mechanism of Action and Pharmacokinetics of Immunosuppressive Drugs. Pharmacogenomics 2013, 14, 1099–1118.
  10. Brazeau, D.A.; Attwood, K.; Meaney, C.J.; Wilding, G.E.; Consiglio, J.D.; Chang, S.S.; Gundroo, A.; Venuto, R.C.; Cooper, L.; Tornatore, K.M. Beyond Single Nucleotide Polymorphisms: CYP3A5∗3∗6∗7 Composite and ABCB1 Haplotype Associations to Tacrolimus Pharmacokinetics in Black and White Renal Transplant Recipients. Front. Genet. 2020, 11, 889.
  11. Hesselink, D.A.; van Schaik, R.H.N.; van der Heiden, I.P.; van der Werf, M.; Gregoor, P.J.H.S.; Lindemans, J.; Weimar, W.; van Gelder, T. Genetic Polymorphisms of the CYP3A4, CYP3A5, and MDR-1 Genes and Pharmacokinetics of the Calcineurin Inhibitors Cyclosporine and Tacrolimus. Clin. Pharmacol. Ther. 2003, 74, 245–254.
  12. Staatz, C.E.; Goodman, L.K.; Tett, S.E. Effect of CYP3A and ABCB1 Single Nucleotide Polymorphisms on the Pharmacokinetics and Pharmacodynamics of Calcineurin Inhibitors: Part II. Clin. Pharmacokinet. 2010, 49, 207–221.
  13. Hesselink, D.A.; Bouamar, R.; Elens, L.; van Schaik, R.H.N.; van Gelder, T. The Role of Pharmacogenetics in the Disposition of and Response to Tacrolimus in Solid Organ Transplantation. Clin. Pharmacokinet. 2014, 53, 123–139.
  14. van Gelder, T.; van Schaik, R.H.; Hesselink, D.A. Pharmacogenetics and immunosuppressive drugs in solid organ transplantation. Nat. Rev. Nephrol. 2014, 10, 725–731.
  15. Evans, W.E. Pharmacogenetics of Thiopurine S-Methyltransferase and Thiopurine Therapy. Ther. Drug Monit. 2004, 26, 186–191.
  16. Budhiraja, P.; Popovtzer, M. Azathioprine-Related Myelosuppression in a Patient Homozygous for TPMT*3A. Nat. Rev. Nephrol. 2011, 7, 478–484.
  17. Yates, C.R.; Krynetski, E.Y.; Loennechen, T.; Fessing, M.Y.; Tai, H.L.; Pui, C.H.; Relling, M.V.; Evans, W.E. Molecular Diagnosis of Thiopurine S-Methyltransferase Deficiency: Genetic Basis for Azathioprine and Mercaptopurine Intolerance. Ann. Intern. Med. 1997, 126, 608–614.
  18. Relling, M.V.; Gardner, E.E.; Sandborn, W.J.; Schmiegelow, K.; Pui, C.-H.; Yee, S.W.; Stein, C.M.; Carrillo, M.; Evans, W.E.; Klein, T.E. Clinical Pharmacogenetics Implementation Consortium Guidelines for Thiopurine Methyltransferase Genotype and Thiopurine Dosing. Clin. Pharmacol. Ther. 2011, 89, 387–391.
  19. Takada, K.; Arefayene, M.; Desta, Z.; Yarboro, C.H.; Boumpas, D.T.; Balow, J.E.; Flockhart, D.A.; Illei, G.G. Cytochrome P450 Pharmacogenetics as a Predictor of Toxicity and Clinical Response to Pulse Cyclophosphamide in Lupus Nephritis. Arthritis Rheum. 2004, 50, 2202–2210.
  20. Chiou, Y.-H.; Wang, L.-Y.; Wang, T.-H.; Huang, S. Genetic Polymorphisms Influence the Steroid Treatment of Children with Idiopathic Nephrotic Syndrome. Pediatr. Nephrol. 2012, 27, 1511–1517.
  21. Santoro, A.; Felipe, C.R.; Tedesco-Silva, H.; Medina-Pestana, J.O.; Struchiner, C.J.; Ojopi, E.B.; Suarez-Kurtz, G. Pharmacogenetics of Calcineurin Inhibitors in Brazilian Renal Transplant Patients. Pharmacogenomics 2011, 12, 1293–1303.
  22. Kuypers, D.R.J.; Naesens, M.; de Jonge, H.; Lerut, E.; Verbeke, K.; Vanrenterghem, Y. Tacrolimus Dose Requirements and CYP3A5 Genotype and the Development of Calcineurin Inhibitor-Associated Nephrotoxicity in Renal Allograft Recipients. Ther. Drug Monit. 2010, 32, 394–404.
  23. Quteineh, L.; Verstuyft, C.; Furlan, V.; Durrbach, A.; Letierce, A.; Ferlicot, S.; Taburet, A.-M.; Charpentier, B.; Becquemont, L. Influence of CYP3A5 Genetic Polymorphism on Tacrolimus Daily Dose Requirements and Acute Rejection in Renal Graft Recipients. Basic Clin. Pharmacol. Toxicol. 2008, 103, 546–552.
  24. Qiu, X.-Y.; Jiao, Z.; Zhang, M.; Zhong, L.-J.; Liang, H.-Q.; Ma, C.-L.; Zhang, L.; Zhong, M.-K. Association of MDR1, CYP3A4*18B, and CYP3A5*3 Polymorphisms with Cyclosporine Pharmacokinetics in Chinese Renal Transplant Recipients. Eur. J. Clin. Pharmacol. 2008, 64, 1069–1084.
  25. Kumaraswami, K.; Katkam, S.K.; Aggarwal, A.; Sharma, A.; Manthri, R.; Kutala, V.K.; Rajasekhar, L. Epistatic Interactions among CYP2C19*2, CYP3A4 and GSTP1 on the Cyclophosphamide Therapy in Lupus Nephritis Patients. Pharmacogenomics 2017, 18, 1401–1411.
  26. Moussa, A.; Mabrouk, S.; Hamdouni, H.; Ajmi, M.; Tfifha, M.; Omezzine, A.; Abroug, S.; Bouslama, A. MDR-1 and CYP3A5 Polymorphisms in Pediatric Idiopathic Nephrotic Syndrome: Impact on Susceptibility and Response to Steroids (Preliminary Results). Clin. Lab. 2017, 63, 1233–1242.
  27. Kurzawski, M.; Dziewanowski, K.; Gawrońska-Szklarz, B.; Domański, L.; Droździk, M. The Impact of Thiopurine S-Methyltransferase Polymorphism on Azathioprine-Induced Myelotoxicity in Renal Transplant Recipients. Ther. Drug Monit. 2005, 27, 435–441.
  28. Ferraresso, M.; Tirelli, A.; Ghio, L.; Grillo, P.; Martina, V.; Torresani, E.; Edefonti, A. Influence of the CYP3A5 Genotype on Tacrolimus Pharmacokinetics and Pharmacodynamics in Young Kidney Transplant Recipients. Pediatr. Transplant. 2007, 11, 296–300.
  29. Uesugi, M.; Masuda, S.; Katsura, T.; Oike, F.; Takada, Y.; Inui, K. Effect of Intestinal CYP3A5 on Postoperative Tacrolimus Trough Levels in Living-Donor Liver Transplant Recipients. Pharmacogenet. Genomics 2006, 16, 119–127.
  30. Op den Buijsch, R.A.M.; Christiaans, M.H.L.; Stolk, L.M.L.; de Vries, J.E.; Cheung, C.Y.; Undre, N.A.; van Hooff, J.P.; van Dieijen-Visser, M.P.; Bekers, O. Tacrolimus Pharmacokinetics and Pharmacogenetics: Influence of Adenosine Triphosphate-Binding Cassette B1 (ABCB1) and Cytochrome (CYP) 3A Polymorphisms. Fundam. Clin. Pharmacol. 2007, 21, 427–435.
  31. Macphee, I.A.M.; Fredericks, S.; Mohamed, M.; Moreton, M.; Carter, N.D.; Johnston, A.; Goldberg, L.; Holt, D.W. Tacrolimus Pharmacogenetics: The CYP3A5*1 Allele Predicts Low Dose-Normalized Tacrolimus Blood Concentrations in Whites and South Asians. Transplantation 2005, 79, 499–502.
  32. Zhang, X.; Liu, Z.; Zheng, J.; Chen, Z.; Tang, Z.; Chen, J.; Li, L. Influence of CYP3A5 and MDR1 Polymorphisms on Tacrolimus Concentration in the Early Stage after Renal Transplantation. Clin. Transplant. 2005, 19, 638–643.
  33. Majchrzak-Celińska, A.; Baer-Dubowska, W. Pharmacoepigenetics: An Element of Personalized Therapy? Expert Opin. Drug Metab. Toxicol. 2017, 13, 387–398.
  34. Stanke-Labesque, F.; Gautier-Veyret, E.; Chhun, S.; Guilhaumou, R. Inflammation Is a Major Regulator of Drug Metabolizing Enzymes and Transporters: Consequences for the Personalization of Drug Treatment. Pharmacol. Ther. 2020, 215, 107627.
More
Information
Contributor MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register :
View Times: 449
Revisions: 2 times (View History)
Update Date: 28 Jul 2021
1000/1000