CLINICALLY SIGNIFICANT DRUG-DRUG INTERACTION IN A LARGE ANTIRETROVIRAL TREATMENT CENTRE IN LAGOS, NIGERIA

Main Article Content

IA Oreagba
SO Usman
KA Oshikoya
AA Akinyede
EO Agbaje
O Opanuga
SA Akanmu

Keywords

potential drug interaction, antiretroviral therapy, co-prescribed non-antiretroviral

Abstract



Background
An important cause of treatment failure to antiretroviral therapy (ART) is the potential interaction between the antiretroviral (ARV) drugs and concomitant drugs (CD) used for the treatment of opportunistic infections and comorbid ailments in HIV-infected patients.
Objectives
The study evaluated potential Clinically Significant Drug Interactions (CSDIs) occurring between recommended ART regimens and their CD.
Method
This study was carried out in a large HIV treatment centre supported by AIDS Preventive initiative in Nigeria (APIN) clinic in a teaching hospital in Lagos, Nigeria, caring for over 20,000 registered patients. Electronic Medical Records (EMRs) of 500 patients, who received treatment between 2005 and 2015, were selected using systematic random sampling, reviewed retrospectively, and evaluated for potential CSDIs using Liverpool HIV Pharmacology Database and other databases for drug-drug interaction check.
Results
Majority of patients, 421 (84%) prescribed CDs were at risk of CSDIs, of which 410 (82%) were moderate and frequently involved co-trimoxazole + combinations of Nucleoside Reverse Transcriptase Inhibitors (NRTIs) such as zidovudine (or stavudine) /lamivudine 386 (77.2%) and Non-nucleoside Reverse Transcriptase Inhibitors (NNRTIs) or Protease Inhibitors (PIs) + artemisinin-based combination therapies (ACTs) 296 (59.2%). Age (p=0.13), sex (p=0.32) and baseline CD4+ cell counts (p=0.20) were not significantly associated with CSDIs. The interactions, however, were significantly associated with the development of antiretroviral treatment failure (p <0.001) which occurred in nearly a third 139 (27.8%) of the patients.


Conclusion
There is a high prevalence of CSDIs between ART and CDs, most of which were categorized as moderate. Further studies are required to evaluate the pharmacokinetic and clinical relevance of these interactions.



Abstract 2177 | pdf Downloads 764

References

1. Palella FJ, Delaney KM, Moorman AC. Declining morbidity and mortality among patients with advanced human immunodeficiency virus infection. N Engl J Med 1998;338:853–60.
2. Aidsinfo. Guidelines for the use of antiretroviral agents in adults and adolescents living with HIV: Initiation of antiretroviral therapy. 2017. Available at: https://aidsinfo.nih.gov/guidelines/ html/1/adult-and-adolescent-arv/10/initiation-of-antiretroviral-therapy.
3. Pujades-Rodríguez M, Balkan SA, Martin B, Alexandra C. Treatment Failure and Mortality Factors in Patients Receiving Second-Line HIV Therapy in Resource Limited Countries. JAMA 2010;304(3):303–12.
4. Lorenzi P, Opravil M, Hirschel B, et al. Impact of drug resistance mutations on virologic response to salvage therapy. Swiss HIV Cohort Study AIDS 1999;13:17–21.
5. Kwobah C, Mwangi AW, Koech JK, Simiyu GN, Siika AM. Factors associated with first-line antiretroviral therapy failure amongst hiv-infected african patients: a case-control study. World J AIDS 2012;2:271–78.
6. Pirmohammed M. Drug-drug interactions and adverse drug reactions: separating the wheat from the chaff. Wien KlinWochenschr 2010;122:62–64.
7. Ingolf Cascorbi. Drug interactions—principles, examples and clinical consequences. Dtsch Arztebl Int 2012;109(33-34):546–56.
8. LaPorte C, Colbers E, Bertz R, et al. Pharmacokinetics of adjusted-dose lopinavir-ritonavir combined with rifampin in healthy volunteers. Antimicrob Agents Chemother 2004;48:1553–60.
9. Breen RA, Lipman MC, Johnson MA. Increased incidence of peripheral neuropathy with co-administration of stavudine and isoniazid in HIV-infected individuals. AIDS 2000;14:615.
10. Xu, C., and Desta Z. In vitro analysis and quantitative prediction of efavirenz inhibition of eight cytochrome P450 (CYP) enzymes (2013): major effects on CYPs 2B6, 2C8, 2C9 and 2C19. Drug Metab Pharmacokin 28(4):362–71.
11. Faucette SR, Zhang TC, Moore R, et al. Relative activation of human pregnane X receptor versus constitutive androstane receptor defines distinct classes of CYP2B6 and CYP3A4 inducers. J Pharmacol Experiment Ther 2007;320(1):72–80.
12. Dixit V, Hariparsad N, Li F, et al. Cytochrome P450 enzymes and transporters induced by anti-human immunodeficiency virus protease inhibitors in human hepatocytes: implications for predicting clinical drug interactions. Drug Metab Disp 2007;35(10)1853–59.
13. Sulaiman A, Akanmu, Usman SO, Oreagba IA, et al. Antiretrovirals and Co-prescribed drugs for people living with HIV/AIDS (PLWHA) in a University Teaching Hospital, South-West Nigeria. West African J Pharm 2015;26(2):103–115.
14. Oshikoya KA, Oreagba IA, Lawal S, Awodele O, et al. Potential drug-drug interactions in HIV-infected children on antiretroviral therapy in Lagos, Nigeria. HIV/AIDS Res Palliat Care 2014;6:49–59.
15. Boyd MA, Zhang X, Dorr A, et al. Lack of enzyme inducing effect of rifampicin on the pharmacokinetics of enfuvirtide. J Clin Pharmacol 2003;43:1382–91.
16. Williamsona B, Dooleyb KE, Zhanga Y, et al. Induction of influx and efflux transporters and cytochrome P450 3A4 in primary human hepatocytes by rifampin, rifabutin, and rifapentine. Antimicrob. Agents Chemother 2013;57(12):6366–69.
17. Hughes CA, Foisy M, Tseng A. Interactions between antifungal and antiretroviral agents. Expert Opin Drug Saf 2010;9(5):723–42.
18. Marzolini C, Elzi L, Gibbons S, et al. Swiss HIV Cohort Study Prevalence of comedications and impact of potential drug-drug interactions in the Swiss HIV Cohort Study. Antivir Ther 2010;15:413–423.
19. Miller CD, El-Kholi R, Faragon JJ, Lodise TP. Prevalence and risk factors for clinically significant drug interactions with antiretroviral therapy. Pharmacotherapy 2007;27:1379–86.
20. Kigen G, Kimaiyo S, Nyandiko W, et al. USAID Academic Model for Prevention Treatment of HIV/AIDS Prevalence of potential drug-drug interactions involving antiretroviral drugs in a large Kenyan cohort. PLoS One 2010;6:e16800 f30.
21. Liverpool HIV Pharmacology Group (LHPG). Available at: http://www.hiv-druginteractions.org/main.aspx?PageId=7.
22. Drugs.com. Drug interactions. Available at: https://www.drugs.com/interactions-check.php?drug_listf28
23. Raosoft® Sample Size Calculator. 2013. Available at: http://www.raosoft.com/samplesize.html.
24. Armahizer M, Kane-Gill SL, Smithburger PL, Anthes AM, Seybert AL. Comparing drug-drug interaction severity for clinician opinion to proprietary databases. Adv Pharmacoepidemiol Drug Saf 2012;1:115. F29.
25. Chatton JY, Munafo A, Chave JP, et al. Trimethoprim, alone or in combination with sulphamethoxazole, decreases the renal excretion of zidovudine and its glucuronide. Br J Clin Pharmacol 1992;34:551–54.
26. Sahai J, Gallicano K, Pakuts A, Cameron DW Effect of fluconazole on zidovudine pharmacokinetics in patients infected with human immunodeficiency virus. J Infect Dis 169(1994):1103–1107.
27. Brockmeyer NH, et al. Pharmacokinetic interaction of fluconazole and zidovudine in HIV-positive patients. Eur J Med Res 1997;2:377–383.
28. Kredo T, Mauff K, Van der Walt JS, et al. The interaction between artemether-lumefantrine and NVP-based antiretroviral therapy in HIV-1 infected patients. Antimicrob Agents Chemother 2011;55(12):5616–23.
29. von Moltke LL, Greenblatt DJ, Granda BW, et al. Inhibition of human cytochrome P450 isoforms by nonnucleoside reverse transcriptase inhibitors. J Clin Pharmacol 2001;41(1):85–91.
30. Van Agtmael MA, Cheng-Qi S, Qing JX, Mull R, van Boxtel CJ. Multiple dose pharmacokinetics of artemether in Chinese patients with uncomplicated falciparum malaria. Int J Antimicrob Agents 1999;12:151–58.
31. Moore KHP, Yuen GJ, Raasch RH, Eron JJ, Martin D, Mydlow PK, Hussey EK. Pharmacokinetics of lamivudine administered alone and with trimethoprim-sulfamethoxazole. Clin Pharmacol Ther 1996;59:550–58
32. Fehintola AF,Scarsi KK,Ma Q,et al. NVP-based antiretroviral therapy impacts artesunate and dihydroartemisinin disposition in HIV-Infected Nigerian adults. Aids Res Treat 2012;Doi:10: 1155/2012/703604
33. Scarsi KK, Fehintola FA, Ma Q, et al. Disposition of amodiaquine and desethylamodiaquine in HIV-infected Nigerian subjects on nevirapine-containing antiretroviral therapy. J Antimicrob Chemother 2014;69(5):1370–76.
34. Akiyoshi Marie T, Murase S, Miyazaki M et al. Mechanism-based inhibition profiles of erythromycin and clarithromycin with cytochrome P450 3A4 genetic variants. Drug Metab Pharmacokin 2013;28(5):411–15.
35. Ribera E, Pou L, Lopez RM, et al. Pharmacokinetic interaction between nevirapine and rifampicin in HIV-infected patients with tuberculosis. J Acquir Immune Defic Syndr 2001;15;28(5):450–53.
36. Brennan-Benson P, Lyus R, Harrison T, et al. Pharmacokinetic interactions between efavirenz and rifampicin in the treatment of HIV and tuberculosis: one size does not fit all. AIDS 2005;19:1541–43.
37. Byakika-Kibwika P, Lamorde M, Mayito J, et al. Significant Pharmacokinetic interactions between artemether/lumefantrine and efavirenz or nevirapine in HIV-infected Ugandan adults. J Antimicrob Chemother 2012;67(9):2213–21.
38. German P, Greenhouse B, Coates C, et al. Hepatotoxicity due to a drug interaction between amodiaquine plus artesunate and efavirenz. Clin Infect Dis 2007;44:889–91.
39. Cong Xu and Zeruesenay Desta. In vitro analysis and quantitative prediction of efavirenz inhibition of eight cytochrome P450 (CYP) enzymes: major effects on CYPs 2B6, 2C8, 2C9 and 2C19. Drug Metab Pharmacokinet 2013;28(4):362–71.
40. Elsharkawy AM, Schwab U, McCarron B, et al. Efavirenz induced acute liver failure requiring liver transplantation in a slow drug metaboliser. J Clin Virol 2013;58:331–3.
41. Wang P, Pradhan K, Zhong X and Ma X Isoniazid metabolism and hepatotoxicity. Acta Pharm Sin B 2016;6(5):384–92.
42. German P, Parikh S, Lawrence J, et al. Lopinavir/ritonavir affects pharmacokinetic exposure of artemether/lumefantrine in HIV-uninfected healthy volunteers. J Acquir Immune Defic Syndr 2009;51(4):424–29.
43. Yeh RF, Gaver VE, Patterson KB, et al Rezk. Lopinavir/ritonavir induces the hepatic activity of cytochrome P450 enzymes CYP2C9, CYP2C19, and CYP1A2 but inhibits the hepatic and intestinal activity of CYP3A as measured by a phenotyping drug cocktail in healthy volunteers. J Acquired Immune Def Synd 2006;42:52–60.
44. Gunness P, Aleksa K, Koren G. The effect of acyclovir on the tubular secretion of creatinine in vitro. J Transl Med 2010;8:139.
45. Wyen C, Fuhr U, Frank D, et al. Effect of an antiretroviral regimen containing ritonavir boosted lopinavir on intestinal and hepatic CYP3A, CYP2D6 and P-glycoprotein in HIV-infected patients. Clin Pharmacol Ther 2008;84(1):75–82.
46. Dixit A, Hariparsad N, Li F, et al. Cytochrome P450 enzymes and transporters induced by anti-human immunodeficiency virus protease inhibitors in human hepatocytes: implications for predicting clinical drug interactions. Drug Metab Disposit 2007;35(10)1853–59.
47. Khaliq Y, Gallicano K, Tisdale C, et al. Pharmacokinetic interaction between mefloquine and ritonavir in healthy volunteers. Br J Clin Pharmacol 2001;51:591–600
48. Krudsood S, Looareesuwan P, Wilairatama W, et al. Effect of artesunate and mefloquine in combination on the Fridericia corrected QT intervals in Plasmodium falciparum infected adults from Thailand. Trop Med Internat Health 2011;16(4):458–65.
49. Briasoulis A, Agarwal V and·Pierce WJ. QT prolongation and torsade de pointes induced by fluoroquinolones: infrequent side effects from commonly used medications. Cardiology 2011;120:103–110
50. Kharasch ED, Mitchell D, Coles R, and Blanco R. Rapid clinical induction of hepatic cytochrome P450 2B6 activity by ritonavir. Antimicrob Agents Chemother May 2008;52(5)1663–69.
51. Hunt K, Hughes CA, and Hills-Nieminen C. Protease inhibitor–associated qt interval prolongation. Ann Pharmacother 2011;45:1544–50.
52. Kounas SP, Letsas KP, Sideris A, Efraimidis M, Kardaras F. QT interval prolongation and torsades de pointes due to a coadministration of metronidazole and amiodarone. Pacing Clin Electrophysiol 2005;28:472–3.
53. Decloedt EH, McIlleron H, Smith P, et al. Pharmacokinetics of lopinavir in HIV-infected adults receiving rifampin with adjusted doses of lopinavir-ritonavir tablets. Antimicrob Agents Chemother 2011;55(7):3195–200.
54. Hossain A, Tran T, Chen T et al. Inhibition of human cytochromes P450 in vitro by ritonavir and cobicistat. J Pharm Pharmacol 2017;69(12):1786–93.
55. Giovanni Fazio, Federica Vernuccio and Giuseppe Grutta. Drugs to be avoided in patients with long QT syndrome: Focus on the anaesthesiological management. World J Cardiol 2013;5(4):87–93.
56. Mugundu GM, Hariparsad N, Desai PB. Impact of ritonavir, atazanavir and their combination on the CYP3A4 induction potential of efavirenz in primary human hepatocytes. Drug Metab Lett 2010;4(1):45–50.
57. Schmitt C, Hofmann C, Riek M, et al. Effect of saquinavir-ritonavir on cytochrome P450 3A4 activity in healthy volunteers using midazolam as a probe. Pharmacotherapy 2009;29(10):1175–81.
58. Walsky RL, Gaman EA, Obach RS. Examination of 209 drugs for inhibition of cytochrome P450 2C8. J Clin Pharmacol 2005;45(1):68–78.
59. de Maat MM, de Boer A, Koks CH, et al. Evaluation of clinical pharmacist interventions on drug interactions in outpatient pharmaceutical HIV-care. J Clin Pharm Ther 2004;29(2):121–30.
60. Evans-Jones JG, Cottle LE, Back DJ, et al. Recognition of risk for clinically significant drug interactions among HIV-infected patients receiving antiretroviral therapy. Clin Infect Dis 2010;50:1419–21.
61. Oshikoya KA, Oreagba IA, Ogunleye OO, Lawal S, Senbanjo IO. Clinically significant interactions between antiretroviral and co-prescribed drugs for HIV-infected children: profiling and comparison of two drug data-bases. Ther Clin Risk Manag 2013;9:215–21. Epub 2013 May 14.
62. Alomar MJ. Factors affecting the development of adverse drug reactions. Saudi Pharm J 2014;22(2):83–89.
63. Walmsley S. Update on antiretroviral treatment failure and the management of treatment-experienced HIV-infected patients. Available at: http://www.thebody.com/content /art42267.html.
64. Nworu CS, Akah PA, Ndu OO, et al. Pharmacokinetic evaluation of drug interactions between co-trimoxazole and zidovudine in rabbits. Internat J Trop Med 2008;3(2):30–35.
65. Moh R, Danel C, Sorho S, et al. Haematological changes in adults receiving a zidovudine-containing HAART regimen in combination with co-trimoxazole in Côte d’Ivoire. Antivir Ther 2005;10(5):615–24.
66. Kore S and Waghmare CS. Anti retroviral therapy (ART) - induced lactic acidosis: A potentially life threatening but preventable complication in HIV/AIDS patients receiving nucleoside reverse transcriptase inhibitors (NRTIs). Biomed Res-India 2012;23(4):625–27. ISSN 0970-938X
67. van Griensven J, De Naeyer L, Mushi T, et al. High prevalence of lipoatrophy among patients on stavudine-containing first-line antiretroviral therapy regimens in Rwanda. Trans R Soc Trop Med Hyg 2007;101(8):793–8.
68. Kallianpur AR, Hulgan T. Pharmacogenetics of nucleoside reverse-transcriptase inhibitor associated peripheral neuropathy. Pharmacogenomics 2009;10 (4):623–37.
69. Anafi SB, Muktar HM and Alawode DF. Commonly prescribed drugs in HIV/AIDS and patient’s socio-demographic data: a case study of University of Ilorin teaching hospital (UITH), Ilorin, Nigeria. Nigerian J Pharm Sci 2008;7(1). ISSN: 0189-823X
70. Aidsinfo. Panel on Opportunistic Infections in HIV-Infected Adults and Adolescents. Guidelines for the prevention and treatment of opportunistic infections in HIV-infected adults and adolescents: recommendations from the Centers for Disease Control and Prevention, the National Institutes of Health, and the HIV Medicine Association of the Infectious Diseases Society of America, page F-8. 2013. Available at http://aidsinfo.nih.gov/contentfiles /lvguidelines/ adult_oi.pdf.
71. Grub S, Bryson H, Goggin T, et al. The interaction of saquinavir (soft gelatin capsule) with ketoconazole, erythromycin and rifampicin: comparison of the effect in healthy volunteers and in HIV-infected patients. Eur J Clin Pharmacol 2001;57:115–21.
72. Kasirye R, Baisley K, Munderi P and Grosskurth H. Effect of cotrimoxazole prophylaxis on malaria occurrence in HIV-infected patients on antiretroviral therapy in sub-Saharan Africa. Trop Med Int Health 2015;20(5):569–80.
73. Rattanapunya S, Cressey TR, Rueangweerayut R, et al. Pharmacokinetic interactions between artesunate/mefloquine and ritonavir-boosted lopinavir in healthy Thai adults. Malar J 2015;14:400.
74. German P, Greenhouse B, Coates C, et al. Hepatotoxicity due to a drug interaction between amodiaquine plus artesunate and EFV. Clin Infect Dis 2007;44(6):889–91.
75. Scarsi KK, Fehintola FA, Ma Q, et al. Disposition of amodiaquine and desethylamodiaquine in HIV-infected Nigerian subjects on nevirapine-containing antiretroviral therapy. J Antimicrob Chemother 2014;69(5):1370–76.
76. Fehintola FA, Scarsi KK, Ma Q, et al. NVP-based antiretroviral therapy impacts artesunate and dihydroartemisinin disposition in HIV-infected Nigerian adults. AIDS Res Treat 2012; Article ID 703604, 6 pages.
77. Schmitt C, Riek M, Winters K, et al. Unexpected hepatotoxicity of rifampin and saquinavir/ritonavir in healthy male volunteers. Arch Drug Inf 2009;2(1):8–16.
78. Matteelli A, Regazzi M, Villani P, et al. Multiple-dose pharmacokinetics of EFV with and without the use of rifampicin in HIV-positive patients. Curr HIV Res 2007;5(3):349–53.
79. Calcagno A, Cusato J, D’Avolio A, Bonora S. Genetic polymorphisms affecting the pharmacokinetics of antiretroviral drugs. [Review]. Clin Pharmaco 2017;56:355–69.