Abstract
There are many factors that can affect the pharmacokinetics (PK) of drugs. Pathophysiological changes from disease states can alter the mechanisms that control the PK of antiretrovirals (ARVs), direct-acting antivirals (DAAs) and addiction treatment medications. Drug-drug interaction pathways of certain ARVs and DAAs can be very complex, with agents being substrates, inhibitors or inducers of multiple metabolic and transporter pathways. Buprenorphine and methadone may be used in HIV and hepatitis C virus (HCV) - infected patients, and may also be affected by drug interactions. Current research is focused on novel PK analyses, which aim to describe the PK of agents within the organs that host the infection of interest, such as within hepatocytes during treatment for HCV. Modeling techniques allow for the prediction of drug PK in specific organs and the plasma compartment. This review will provide a summary of these areas while exploring PK considerations for ARVs, DAAs, and addiction treatment medications.
Introduction
Multiple factors can contribute to pharmacokinetic (PK) drug interactions. If drugs are administered orally, several possible mechanisms can modulate PK from within the gut, such as gastrointestinal absorption (e.g., chelation, changes in pH, motility alterations), membrane transporters, and metabolism in the gut wall. When drug is transferred via the portal vein to the liver, hepatic processes that affect PK can occur, consisting of additional uptake and efflux membrane transporters, metabolism, elimination in the bile, and enterohepatic cycling.
Pathophysiological changes from disease states can alter the mechanisms that control the PK of antiretrovirals (ARVs), direct-acting antivirals (DAAs) and addiction treatment medications. When drug-drug interaction studies are conducted in healthy volunteers this assumes that the PK profiles seen in these healthy adults will be comparable to patients with the disease state of interest. 1 However, this assumption has certain limitations. HIV and hepatitis C virus (HCV) infections affect the small intestine and liver, 2,3 which are major sites involved in drug absorption, transport, metabolism and elimination. 4 Possible pathophysiological changes in transporter and metabolic function from HIV and HCV are summarized in Table 1.
Table 1.
HCV-infected, liver cirrhosis compared to non-infected, non-cirrhosis group.
Patients with cirrhosis, compared to those with hepatic angioma.
Trend toward downregulation. Increase ↑; decrease ↓
P-gp; P-glycoprotein. CYP3A4; Cytochrome P450 3A4. MRP2; Multidrug resistance-associated protein 2. OATP2B1; Organic Anion Transporting Polypeptide 2B1. BCRP; Breast Cancer Resistance Protein. CYP2D6; Cytochrome P450 2D6. OATP1B1/3; Organic Anion Transporting Polypeptide 1B1 and 1B3.
The possibility that individuals with disease may exhibit altered PK parameters has been discussed. 9 For example, HIV-infected patients demonstrate decreased relative bioavailability of efavirenz as compared to healthy subjects. 10 It has also been reported that subjects co-infected with HIV/HCV with or without cirrhosis had significantly lower oral nelfinavir clearance than HIV monoinfected subjects. 11 In a study of HIV+ adult males, and controls with comparable weight, BMI, and estimated creatinine clearance, significantly lower mean plasma fluconazole clearance was found in the HIV+ subjects with the lowest CD4+T cell count, versus HIV+ subjects with higher CD4+T cell counts and controls. 12
Drug interactions in HAART era and today
Highly active antiretroviral therapy (HAART) was first introduced in the 1990s and has revolutionized the treatment of HIV, greatly improving the morbidity and mortality associated with HIV infection. However, combining these medications with other agents also came with an increased risk for drug interactions, based on the different PK effects each drug had on the others. In addition, HAART required a large pill burden, and certain drugs have specific administration requirements, such as food or pH dependencies. These concerns have been somewhat reduced as single daily medications have become available in co-formulated combinations.
Currently with the advancement in HCV therapies and increased rate of HIV/HCV co-infection being simultaneously treated, new drug-drug interactions have become a concern. 13 The effects may manifest in changes in systemic or tissue exposure of the direct-acting antiviral (DAA) or ARV. Potentially this could cause increases in exposure, resulting in adverse or toxic effects, or decreases in efficacy. In addition, these interactions could extend to concomitant medications commonly used in this patient population, such as antihypertensives, statins and opioid maintenance therapy. 1,14
Factors Influencing Intrahepatic Drug Concentrations
Currently research interest has been extended beyond measuring only plasma concentrations, since it has been realized that this may not be representative of the drug that is present at the site of action, or tissue of interest. 15,16 Membrane transporters are important for hepatic uptake and efflux, potentially affecting plasma or hepatic drug concentration. 15 The question arises about whether to target plasma or liver concentrations for deciding DAA dosing, and the importance of understanding information gained from hepatic drug sampling in addition to plasma pharmacokinetics. 17
Clinical Pharmacology of New HCV DAAs
The interaction pathways of some current DAAs can be rather complex, with agents being substrates, inhibitors or inducers of multiple metabolic and transporter pathways. 13 In addition, DAAs with different mechanisms of action are given in combination to increased efficacy and decrease the chance of resistant virus emerging. Importantly, these combination of DAAs has allowed for the removal of interferon from HCV regimens,18 thus limiting the toxicity and burden of injections associated with this agent. When ARVs and DAAs are combined to treat HIV/HCV co-infected populations, these patients could take at least 5 different medications to treat these two infections. Notably, these patients may also have multiple co-morbidities that require additional pharmacotherapy. Therefore, the net effect from these individual drug interaction pathways result in the pharmacologic/toxic effect that is observed within an individual.
Complexities with Substance Abuse
In a study examining the influence of active substance related disorder (SRD) in HIV subjects receiving combination antiretroviral therapy, 19 subjects were divided into those with SRD and those without.19 Pharmacokinetic evaluation was performed by sampling ARV trough concentrations and additional samples after an observed dose. It was noted that those subjects with SRD were more likely to have efavirenz or protease inhibitor trough concentrations below the desired range (p=0.048).19 At the time of therapeutic drug monitoring, patients with active SRD had a significantly lower percentage of those with HIV-1 RNA < 75 copies/mL compared to those without active SRD (p=0.044), however, in a multivariate linear regression model known substance use was not significantly associated with CD4+ cell count and HIV RNA <75 copies/mL at study entry.19
Drug Interactions with Addiction Treatment Medications
Buprenorphine and methadone are addiction treatment medications recommended to treat opioid addiction in HIV and HCV infected patients. In-vitro, buprenorphine and methadone are both substrates of cytochrome P450 3A4, with methadone also primarily being metabolized by CYP2B6. 20,21 Thus, these agents may be subject to potential drug interactions with ARV or DAA regimens that modulate these metabolic enzymes. Caution is warranted and understanding possible interactions is necessary when these drugs are co-administered. The investigation of buprenorphine and methadone drug interactions with ARVs have been extensively performed and reported in the literature. 22–25
Pharmacokinetic Analysis Approaches
Traditional PK analysis consists of the estimation of PK parameters from intensive drug concentration sampling over time. Population pharmacokinetic modeling is a method that can utilize not only intensive data, but also data from sparse sampling strategies 26 that cannot be used for traditional PK analysis. 26 Population pharmacokinetics is used to evaluate the pharmacokinetic variability seen in individuals within a population.27 However, a limitation to population pharmacokinetic modeling is that this method of analysis cannot be used to anticipate new drug-drug interactions in conditions where multiple medications are given.
Another modeling approach called physiologically-based pharmacokinetic (PBPK) modeling estimates parameters based on physiology, with models containing multiple compartments that correspond to organs within the body that are connected using physiologic blood flow rates. 28 PBPK models can estimate traditional PK parameters, 28 while also accounting for pathophysiological changes that occur during a disease state of interest to predict the pharmacokinetic effect. 28 Also, in-vitro data can be extrapolated to in-vivo and used to inform these models. 29 These PBPK models can incorporate information from various sources, and predict the PK of drug(s) in various situations, populations or disease states. 28
Pharmacogenomics
Utilizing pharmacogenomics to tailor patients pharmacotherapy is a growing field, but is still in its infancy. 30 Looking at differences in gene expression can be examined as a covariate to explain differences in treatment response or adverse outcomes. In a study examining the effect of single nucleotide polymorphisms (SNPs) for the gene encoding the delta-opioid receptor, the association of the genetic effect with buprenorphine treatment outcome in men and women was examined separately. An association between certain SNPs and buprenorphine treatment outcome was found for women, depending on the genotype there was a significantly worse outcome, however, validation is required in an independent study. 31 In a study of 366 Asian subjects who were on maintenance methadone treatment, researchers examined a matrix of genetic variants from CYP2C19, CYP2B6, and CYP3A4 polymorphisms. It was found that the methadone dose was significantly influenced by allelic variants associated with the CYP2C19 gene. 32
Conclusion
The pharmacokinetic interactions of ARV, DAAs and addiction therapy are complex, potentially leading to alterations in drug concentrations and treatment outcomes with certain agents. Due to the high potential of drug interactions, patient’s medications should be closely examined prior to HCV treatment, including attention to addiction treatment medications. Re-evaluation of the patient’s medication list should be performed after the completion of DAA therapy.
Liver sampling strategies may advance virologic and pharmacologic studies as well as identify drug interactions. It is important to be cognizant of possible tissue uptake/efflux and where the site of action of the drug is, and to remember that plasma pharmacokinetics may not be representative of drug concentration at the tissue of interest.
Lastly, a variety of modeling and analysis tools are available to help answer the question of interest. The choice of which tool to use should be based on the study design, type of data that will be collected, and the question that is to be answered.
Acknowledgments
Research reported in this publication was supported by the National Institutes of Health under the following Award Numbers: Dr. Gene D. Morse was supported by UM1AI106701, HHSN272201500006C and D43TW010313; Dr. Qing Ma was supported by K08MH098794; Dr. Cindy J. Bednasz was supported by UM1AI068636 and an award from the University of Rochester Center for Human Experimental Therapeutics; Dr. Charles S. Venuto was supported by K23 AI108355. The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health.
Footnotes
The authors report no conflict of interest and have no financial disclosures to report in regards to this article.
References
- 1.Menon R, Badri P, Das U, et al. Drug-drug interactions with direct-acting antiviral combination therapy of ABT-450/r, ombitasvir, and dasabuvir. 54th Interscience Conference on Antimicrobial Agents and Chemotherapy; 5–9 september 2014; Washington, D.C. [on 11/20/2015]. Accessed at http://www.natap.org/2014/ICAAC/ICAAC_19.htm. [Google Scholar]
- 2.Gaulke CA, Porter M, Han YH, et al. Intestinal epithelial barrier disruption through altered mucosal microRNA expression in human immunodeficiency virus and simian immunodeficiency virus infections. Journal of Virology. 2014;88:6268–6280. doi: 10.1128/JVI.00097-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Ogasawara K, Terada T, Katsura T, et al. Hepatitis C virus-related cirrhosis is a major determinant of the expression levels of hepatic drug transporters. Drug Metabolism and Pharmacokinetics. 2010;25:190–199. doi: 10.2133/dmpk.25.190. [DOI] [PubMed] [Google Scholar]
- 4.U.S. Food and Drug Administration. Drug Development and Drug Interactions: Table of Substrates, Inhibitors and Inducers. [Date Accessed 11/5/14]. [Google Scholar]
- 5.Kis O, Sankaran-Walters S, Hoque MT, Walmsley SL, Dandekar S, Bendayan R. HIV-1 Alters Intestinal Expression of Drug Transporters and Metabolic Enzymes: Implications for Antiretroviral Drug Disposition. Antimicrob Agents Chemother. 2016;60:2771–81. doi: 10.1128/AAC.02278-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Kikuchi R, McCown M, Olson P, et al. Effect of hepatitis C virus infection on the mRNA expression of drug transporters and cytochrome p450 enzymes in chimeric mice with humanized liver. Drug Metab Dispos. 2010;38:1954–1961. doi: 10.1124/dmd.109.031732. [DOI] [PubMed] [Google Scholar]
- 7.Ogasawara K, Terada T, Katsura T, et al. Hepatitis C virus-related cirrhosis is a major determinant of the expression levels of hepatic drug transporters. Drug Metab Pharmacokinet. 2010;25:190–199. doi: 10.2133/dmpk.25.190. [DOI] [PubMed] [Google Scholar]
- 8.Yang LQ, Li SJ, Cao YF, et al. Different alterations of cytochrome P450 3A4 isoform and its gene expression in livers of patients with chronic liver diseases. World J Gastroenterol. 2003;9:359–363. doi: 10.3748/wjg.v9.i2.359. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Wilkinson GR. The effects of diet, aging and disease-states on presystemic elimination and oral drug bioavailability in humans. Advanced Drug Delivery Reviews. 1997;27:129–159. doi: 10.1016/s0169-409x(97)00040-9. [DOI] [PubMed] [Google Scholar]
- 10.Mukonzo JK, Nanzigu S, Rekic D, et al. HIV/AIDS patients display lower relative bioavailability of efavirenz than healthy subjects. Clinical Pharmacokinetics. 2011;50:531–540. doi: 10.2165/11592660-000000000-00000. [DOI] [PubMed] [Google Scholar]
- 11.Regazzi M, Maserati R, Villani P, et al. Clinical pharmacokinetics of nelfinavir and its metabolite M8 in human immunodeficiency virus (HIV)-positive and HIV-hepatitis C virus-coinfected subjects. Antimicrobial Agents and Chemotherapy. 2005;49:643–649. doi: 10.1128/AAC.49.2.643-649.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Tett S, Moore S, Ray J. Pharmacokinetics and bioavailability of fluconazole in two groups of males with human immunodeficiency virus (HIV) infection compared with those in a group of males without HIV infection. Antimicrobial Agents and Chemotherapy. 1995;39:1835–1841. doi: 10.1128/aac.39.8.1835. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Bednasz CJ, Sawyer JR, Martinez A, et al. Recent advances in management of the HIV/HCV coinfected patient. Future Virology. 2015;10:981–997. doi: 10.2217/fvl.15.64. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.European Association for Study of Liver. EASL Recommendations on Treatment of Hepatitis C 2015. Journal of Hepatology. 2015;63:199–236. doi: 10.1016/j.jhep.2015.03.025. [DOI] [PubMed] [Google Scholar]
- 15.Watanabe T, Kusuhara H, Maeda K, Shitara Y, Sugiyama Y. Physiologically based pharmacokinetic modeling to predict transporter-mediated clearance and distribution of pravastatin in humans. The Journal of Pharmacology and Experimental Therapeutics. 2009;328:652–662. doi: 10.1124/jpet.108.146647. [DOI] [PubMed] [Google Scholar]
- 16.Rose RH, Neuhoff S, Abduljalil K, Chetty M, Rostami-Hodjegan A, Jamei M. Application of a Physiologically Based Pharmacokinetic Model to Predict OATP1B1-Related Variability in Pharmacodynamics of Rosuvastatin. CPT: Pharmacometrics & Systems Pharmacology. 2014;3:e124. doi: 10.1038/psp.2014.24. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Wright DH, Caro L, Cerra M, et al. Liver-to-plasma vaniprevir (MK-7009) concentration ratios in HCV-infected patients. Antivir Ther. 2015;20:843–8. doi: 10.3851/IMP2958. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.AASLD/IDSA/IAS–USA. [Accessed 11/20/16];Recommendations for testing, managing, and treating hepatitis C. http://www.hcvguidelines.org.
- 19.Ma Q, Zingman BS, Luque AE, et al. Therapeutic drug monitoring of protease inhibitors and efavirenz in HIV-infected individuals with active substance-related disorders. Therapeutic drug monitoring. 2011;33:309–314. doi: 10.1097/FTD.0b013e31821d3adb. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Kobayashi K, Yamamoto T, Chiba K, Tani M, Shimada N, Ishizaki T, et al. Human buprenorphine N-dealkylation is catalyzed by cytochrome P450 3A4. Drug Metab Dispos. 1998;26:818–21. [PubMed] [Google Scholar]
- 21.Chang Y, Fang WB, Lin SN, Moody DE. Stereo-selective metabolism of methadone by human liver microsomes and cDNA-expressed cytochrome P450s: a reconciliation. Basic Clin Pharmacol Toxicol. 2011;108:55–62. doi: 10.1111/j.1742-7843.2010.00628.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Gruber VA, McCance-Katz EF. Methadone, buprenorphine, and street drug interactions with antiretroviral medications. Current HIV/AIDS reports. 2010;7:152–160. doi: 10.1007/s11904-010-0048-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.McCance-Katz EF, Moody DE, Morse GD, et al. Interaction between buprenorphine and atazanavir or atazanavir/ritonavir. Drug and alcohol dependence. 2007;91:269–278. doi: 10.1016/j.drugalcdep.2007.06.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.McCance-Katz EF. Treatment of opioid dependence and coinfection with HIV and hepatitis C virus in opioid-dependent patients: the importance of drug interactions between opioids and antiretroviral agents. Clin Infect Dis. 2005;41(Suppl 1):S89–95. doi: 10.1086/429503. [DOI] [PubMed] [Google Scholar]
- 25.McCance-Katz EF, Rainey PM, Friedland G, Jatlow P. The protease inhibitor lopinavir-ritonavir may produce opiate withdrawal in methadone-maintained patients. Clin Infect Dis. 2003;37:476–482. doi: 10.1086/376907. [DOI] [PubMed] [Google Scholar]
- 26.U.S. Department of Health and Human Services. Food and Drug Administration Center for Drug Evaluation and Research (CDER). Center for Biologics Evaluation and Research (CBER) Population Pharmacokinetics. Feburary. 1999. Guidance for Industry. [Google Scholar]
- 27.Aarons L. Population pharmacokinetics: theory and practice. Br J Clin Pharmacol. 1991;32:669–670. [PMC free article] [PubMed] [Google Scholar]
- 28.Jones H, Rowland-Yeo K. Basic concepts in physiologically based pharmacokinetic modeling in drug discovery and development. CPT: Pharmacometrics & Systems Pharmacology. 2013;2:e63. doi: 10.1038/psp.2013.41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Jamei M. An Introduction to: In Vitro - In Vivo Extrapolation (IVIVE) The University of Greenwich; Oct 29, 2009. [Accessed on 11/20/15]. http://www.slideshare.net/mjamei/an-introduction-toin-vitroin-vivoextrapolation-ivive. [Google Scholar]
- 30.U.S. National Library of Medicine. [Accessed on 11/11/15];Genetics Home Reference. http://ghr.nlm.nih.gov/handbook/genomicresearch/pharmacogenomics.
- 31.Clarke TK, Crist RC, Ang A, et al. Genetic variation in OPRD1 and the response to treatment for opioid dependence with buprenorphine in European-American females. The Pharmacogenomics Journal. 2014;14:303–308. doi: 10.1038/tpj.2013.30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Tsai HJ, Wang SC, Liu SW, et al. Assessment of CYP450 genetic variability effect on methadone dose and tolerance. Pharmacogenomics. 2014;15:977–986. doi: 10.2217/pgs.14.19. [DOI] [PubMed] [Google Scholar]
