Review. Therapeutic Drug Monitoring of Antiretroviral Therapy

Similar documents
PHARMACOKINETICS OF ANTIRETROVIRAL AND ANTI-HCV AGENTS

Anumber of clinical trials have demonstrated

104 MMWR December 17, 2004

Combination of protease inhibitors for the treatment of HIV-1-infected patients: a review of pharmacokinetics and clinical experience

Terapia antirretroviral inicial y de rescate: Utilidad actual y futura de nuevos medicamentos

Concomitant antiretroviral therapy : Avifanz must be given in combination with other antiretroviral medications.

Global Pharmaceutical Research and Development, Abbott Laboratories, Abbott Park, Illinois 60064

Many highly active antiretroviral therapy PROCEEDINGS

HIV Treatment Update. Awewura Kwara, MD, MPH&TM Associate Professor of Medicine and Infectious Diseases Brown University

Perspective Resistance and Replication Capacity Assays: Clinical Utility and Interpretation

Efavirenz plasma levels can predict treatment failure and central nervous system side effects in HIV-1-infected patients

Drug-Drug Interactions that Matter

Kimberly Adkison, 1 Lesley Kahl, 1 Elizabeth Blair, 1 Kostas Angelis, 2 Herta Crauwels, 3 Maria Nascimento, 1 Michael Aboud 1

THERAPEUTIC DRUG MONITORING (TDM) Table 2. Dose Adjustment. Patient Drug (mg) Symptoms C trough -fold increase compared to MEC WT

Treatment of HIV-1 in Adults and Adolescents: Part 2

National AIDS Treatment Advocacy Project

Guidelines for the Use of Antiretroviral Agents in HIV-1-Infected Adults and Adolescents

Clinical Commissioning Policy: Use of cobicistat (Tybost ) as a booster in treatment of HIV positive adults and adolescents

Management of patients with antiretroviral treatment failure: guidelines comparison

Special Contribution Questions to and Answers from the International AIDS Society USA Resistance Testing Guidelines Panel

2 nd Line Treatment and Resistance. Dr Rohit Talwani & Dr Dave Riedel 12 th June 2012

Recommendations for Use of Antiretroviral Drugs in Pregnant HIV-1-Infected Women for Maternal Health and

0.14 ( 0.053%) UNAIDS 10% (94) ( ) (73-94/6 ) 8,920

Principles of Antiretroviral Therapy

Central Nervous System Penetration of ARVs: Does it Matter?

PART VI: SUMMARY OF THE RISK MANAGEMENT PLAN

TB/HIV Co-Infection. Tuberculosis and HIV

Liver Toxicity in Epidemiological Cohorts

Supplemental Digital Content 1. Combination antiretroviral therapy regimens utilized in each study

PART VI: SUMMARY OF THE RISK MANAGEMENT PLAN

Introduction to HIV Drug Resistance. Kevin L. Ard, MD, MPH Massachusetts General Hospital Harvard Medical School

Reverse transcriptase and protease inhibitor resistant mutations in art treatment naïve and treated hiv-1 infected children in India A Short Review

Pharmacokinetics and therapeutic drug monitoring of antiretrovirals in pregnant women

TB Intensive Tyler, Texas December 2-4, Tuberculosis and HIV Co-Infection. Lisa Y. Armitige, MD, PhD. December 4, 2008.

Management of NRTI Resistance

UvA-DARE (Digital Academic Repository)

Pharmacology Update Alice Tseng, Pharm.D., FCSHP Vancouver May 11, 2005


Second-Line Therapy NORTHWEST AIDS EDUCATION AND TRAINING CENTER

Second and third line paediatric ART strategies

Once-a-Day Highly Active Antiretroviral Therapy: A Systematic Review

Mechanisms of rifamycin-antiretroviral drug interactions

Pharmacotherapy of Comorbities: Cancer Patients with HIV/AIDS

PAEDIATRIC HIV INFECTION. Dr Ashendri Pillay Paediatric Infectious Diseases Specialist

Prevalence of drug-drug interactions of antiretroviral agents in the private health care sector in South Africa. S Afr Med J 2008; 98:

The legally binding text is the original French version TRANSPARENCY COMMITTEE OPINION. 14 December 2011

VIKING STUDIES Efficacy and safety of dolutegravir in treatment-experienced subjects

Nevirapine 200mg Tablet WHOPAR part 3 May 2005 Updated: May 2016 PACKAGE LEAFLET

Antiretroviral Therapy During Pregnancy and Delivery: 2015 Update

HIV associated CNS disease in the era of HAART

GSK Medicine: Study Number: Title: Rationale: Phase: Study Period: Study Design: Centres: Indication: Treatment: Objectives:

Somnuek Sungkanuparph, M.D.

ABC/3TC/ZDV ABC PBO/3TC/ZDV

EFFECT OF FIRST LINE THERAPY INCLUDING EFAVIRENZ AND TWO NUCLEOSIDE REVERSE TRANSCRIPTASE INHIBITORS IN HIV-INFECTED CHILDREN

Continuing Education for Pharmacy Technicians

Pharmacological determinants of long-term treatment success

Distribution and Effectiveness of Antiretrovirals in the Central Nervous System

Pediatric Patient Information:

Practical Scenarios Calculating doses for newborns. Karen Buckberry

PDF hosted at the Radboud Repository of the Radboud University Nijmegen

Jennifer R King, Amit Khatri, Roger Trinh, Bifeng Ding, Jiuhong Zha and Rajeev Menon AbbVie Inc.

Scottish Medicines Consortium

HIV basics. Katya Calvo Medical Director of Antimicrobial Stewardship

EXPLORING THE IMPACT OF SEX DISCREPANCIES IN HIV TREATMENT AND CURE

Quick Reference Guide to Antiretrovirals. Guide to Antiretroviral Agents

HIV medications HIV medication and schedule plan

Lê MP, 1,2 Cournil A, 3 Kouanfack C, 4 Lem S, 5 Le Gac S, 5 Delaporte E, 3 Peytavin G, 1,2 for the NAMSAL study group

Frailty and age are independently associated with patterns of HIV antiretroviral use in a clinical setting. Giovanni Guaraldi

HIV THERAPY STRATEGIES FOR THIRD LINE. issues to consider when faced with few drug options

Pharmacological considerations on the use of ARVs in pregnancy

The US Food and Drug Administration has

Etravirine, a Next-Generation Nonnucleoside Reverse- Transcriptase Inhbitor

Pharmacologic Characteristics and Delivery Options for Integrase Inhibitors

Recommended dosing for pediatric patients (6 months to 12 years of age) 1. Dose based on lopinavir component* 1.25 ml ml

TDM. Measurement techniques used to determine cyclosporine level include:

Each tablet contains:

ORIGINAL RESEARCH. Introduction

Clinical Study Report AI Final 28 Feb Volume: Page:

Cover Page. The handle holds various files of this Leiden University dissertation.

Pharmacology of generics. Dario Cattaneo Unit of Clinical Pharmacology Luigi Sacco University Hospital, Milano, ITALY

The management of HIV-1 protease inhibitor pharmacokinetic interactions

Indinavir, Efavirenz, and Abacavir Pharmacokinetics in Human Immunodeficiency Virus-Infected Subjects

Antiretroviral Dosing in Renal Impairment

Register for notification of guideline updates at

WHAT S NEW IN THE 2015 PERINATAL HIV GUIDELINES?

Structured Treatment Interruption in HIV Positive Patients. Leah Jackson, BScPhm Pharmacy Resident HIV Rotation January 23, 2007

Protease Inhibitor Therapy: Boosted and Double-Boosted Options to the Fore

THE HIV LIFE CYCLE. Understanding How Antiretroviral Medications Work

Fluconazole dimenhydrinate, diphenhydramine. Raltegravir or dolutegravir with antacids

British HIV Association Guidelines for the Management of Hepatitis Viruses in Adults Infected with HIV 2013 Appendix 2

This graph displays the natural history of the HIV disease. During acute infection there is high levels of HIV RNA in plasma, and CD4 s counts

Comprehensive Guideline Summary

HIV replication and selection of resistance: basic principles

HIV and the Central Nervous System Impact of Drug Distribution Scott L. Letendre, MD. Professor of Medicine University of California, San Diego

Supplementary information

CURRICULUM VITAE. Miguel Goicoechea, M.D.

Drug Resistance: Part 2 Application to clinical practice

Median (Min Max) CVC 100 mg + EFV placebo + TDF/FTC (N=8) CVC 200 mg + EFV placebo + TDF/FTC (N=10) LLOQ=5.00 ng/ml Time (h)

A Genetic Test to Screen for Abacavir Hypersensitivity Reactions

Improving accessibility to antiretroviral drugs: A south-south collaboration

Transcription:

AIDS PATIENT CARE and STDs Volume 18, Number 1, 2004 Mary Ann Liebert, Inc. Review Therapeutic Drug Monitoring of Antiretroviral Therapy NATELLA Y. RAKHMANINA, M.D., 1,4 JOHN N. VAN DEN ANKER, M.D., Ph.D., 2,4 6 and STEVEN J. SOLDIN, Ph.D. 3 5,7 ABSTRACT The concept of managing pharmacotherapy based on plasma drug concentrations has been used for decades in a variety of clinical settings. The interest in therapeutic drug monitoring (TDM) of antiretroviral drugs has grown significantly since highly active antiretroviral therapy (HAART) became a standard of care in clinical practice. A primary characteristic of TDM of antiretroviral drugs is that multiple agents are concomitantly used in HAART regimens. Inadequate drug concentrations may lead to evolution of drug resistance mutations and endanger present and future treatment options. A number of clinical trials have demonstrated that drug serum concentrations are an important factor in response to therapy for HIV, but whether TDM will become a tool for the routine management of HIV infection remains to be determined. This review includes an illustrative case report of measuring concentrations of antiretroviral drugs in a pediatric patient. CONCEPTS OF THERAPEUTIC DRUG MANAGEMENT IN HIV THERAPEUTIC DRUG MONITORING (TDM) permits timely dosage adjustments based on individual plasma concentrations to optimize the therapeutic effect of the medication and prevent exposure to toxic or subtherapeutic concentrations of the drug. Criteria for successful TDM include: well-defined therapeutic range, strong correlation between drug concentration and therapeutic effect, high interpatient and low intrapatient variability in plasma concentrations and failure of the long-term outcome with inadequate target levels. 1 A reliable, cost-effective method should be available for the determination of drug concentrations. 2 While several features of antiretroviral therapy (ART) fit the criteria for TDM, a number of questions need to be answered before largescale introduction of TDM can be justified. For the majority of antiretroviral drugs the therapeutic window is narrow, with toxicities ranging from nausea and vomiting to pancreatitis, nephrolithiasis, and neurologic effects. 2 4 Some of these toxicities might be prevented with the 1 Special Immunology Program, 2 Division of Pediatric Clinical Pharmacology, and 3 Department of Laboratory Medicine, Children s National Medical Center, Washington, D.C. Departments of 4 Pediatrics, 5 Pathology and 6 Pharmacology, The George Washington University School of Medicine, Washington, D.C. 7 Department of Pharmacology, Georgetown University and Georgetown Clinical Research Center, Washington, D.C. 7

8 RAKHMANINA ET AL. appropriate use of TDM. 5,6 Concentrations of antiretroviral agents vary greatly among individual patients who are treated with the same dose, often more than 10-fold. 7,8 The cause of such significant interpatient pharmacokinetic variability is multifactorial and includes differences in metabolism, bioavailability, complex drug-drug and food-drug interactions. Several studies have demonstrated a lack of adequate viral suppression in the absence of therapeutic drug levels. 9 11 The pharmacokinetic (PK) data from the Viradapt study have shown a significant correlation between suboptimal drug concentrations and the risk of virologic failure. 12 This is of particular importance because preliminary studies from centers that offer TDM have been reporting that a substantial proportion of patients may have subtherapeutic levels of antiretroviral agents. 8,13,14 Adverse events, nonadherence, and drug interactions are frequently responsible for subtherapeutic drug levels. A recent study by De Maat et al., 15 however, demonstrated a high number of subtherapeutic drug concentrations without an identifiable cause. Inadequate concentrations of antiretroviral agents may increase the potential of drug resistance and treatment failure. A narrow therapeutic index of many antiretroviral agents suggests the possibility of using TDM for concentration-targeted dosing. Applying TDM to enhance treatment outcome is much more complex and difficult to achieve. There are no studies currently available that have compared peak plasma concentrations (C max ), the area under the serum concentration versus time curves (AUC), and trough plasma concentrations (C min ) as predictors of treatment efficacy. The AUC, a measure of systemic exposure, may be an important target because it is more likely to reflect exposure in body compartments. Studies of virologic efficacy have found a significant correlation both with C min and AUC. The ratio of C min to inhibitory concentration at 50% (IC 50 ), defined as inhibitory quotient (IQ), is used by many researchers and is believed to be the parameter most likely to predict efficacy. TDM of antiretrovirals is also being used to monitor patient adherence to the regimen. 16 Poor adherence has been identified as a leading cause of treatment failure by many authors. 17,18 Nonadherence is a serious problem and is reported in 33% to 69% of this patient population. 4,19 Common reasons for missing doses include clinical toxicity, forgetfulness, sleeping through the time of prescribed dose, and being away from home. TDM can help identify nonadherence, although a drug concentration only reflects the last few drug doses taken by the patient. It is crucial that any TDM results are incorporated with careful adherence assessment to facilitate interpretation of the results and adherence counseling should be established as part of clinical care in the centers practicing TDM. TDM OF DIFFERENT ANTIRETROVIRAL AGENTS Adequate drug concentrations may not reach the virus because of poor compliance, poor absorption, increased metabolism and/or elimination, and drug-drug and drug-food interactions. The most important and most common pharmacokinetic drug interactions involve inhibition of metabolism, induction of metabolism, altered drug absorption, inhibition of renal secretion, and displacement from plasma protein and/or binding sites. The nucleoside reverse transcriptase inhibitors (NRTI) are active as intracellular triphosphates, and there is little evidence to suggest that their measurement would be helpful other than to assess adherence to the drug regimen. Several studies have established a relationship between plasma concentrations of NRTI and virologic and immunologic outcomes. 20,21 The usefulness of NRTI plasma concentration in predicting the intracellular levels of their triphosphate metabolites remains unclear. The relationship between the intracellular drug levels and outcome parameters for zidovudine and lamivudine has been shown to be significant, 20 but the methodology of intracellular triphosphate metabolites is expensive and labor-intensive and is limited to highly specialized centers. For the protease inhibitors (PIs) and to lesser extent for non-nucleoside reverse transcriptase inhibitors (NNRTIs), the relationship between

THERAPEUTIC DRUG MONITORING 9 plasma drug concentrations and their efficacy and toxicity has been identified. 9,11,12,22 24 Large interindividual and intraindividual differences in drug disposition have been observed with the use of many antiretroviral agents, which makes dose based prediction of plasma concentrations unreliable. 7,25,26 The patients taking NNRTIs are at the greatest risk for developing resistance and treatment-limiting toxicity. These drugs are metabolized by CYP 450 2B6, 1A2, 2A6, 2C and 2D6 (efavirenz, nevirapine, nelfinavir, and ritonavir) and by 3A, primarily 3A4 (indinavir, delavirdine, ritonavir, nevirapine, saquinavir, and nelfinavir) creating a solid base for multiple drug-drug interactions. 27,28 Many drugs used in the treatment of infections associated with HIV-1 disease (ketoconazole, fluconazole, rifampin, rifabutin, methadone) as well as a herbal supplement (St. John s wort) have shown significant interactions with antiretroviral drugs leading to toxicity-related complications and subtherapeutic concentrations. The interactions between PIs and NNRTIs, and among different PIs, especially in salvage regimens, have become clinical issues with the growing resistance pattern of the HIV. The application of TDM in the patients who are using two and more P-450 interacting agents seems therefore indicated. The PIs are strongly protein bound, and especially to a-1-acid glycoprotein (AAG), an acute phase reactant. 29,30 In general there is a lack of data on the effect of varying concentrations of the binding protein on the free drug concentrations and its antiviral activity. This is particularly true for pediatric patients, in whom concentrations of albumin and/or AAG may vary substantially. Currently, studies are being conducted on the value of free drug concentration measurements and its relationship with outcome measurements. Recently, randomized, prospective clinical trials evaluating the role of TDM in the management of HIV-1 infected patients have shown promising results. 4 The ATHENA study conducted in The Netherlands included two clinical trials using TDM in treatment-naïve patients who started indinavir- or nelfinavir-based regimens. TDM of nelfinavir improved viral load effects but did not reduce toxicity, while dose adjustment for indinavir reduced toxicity but did not improve antiviral effect. 13,14 TDM did prevent either virologic failure (presumably by preventing development of resistance) or treatment discontinuation because of concentrationrelated toxicities. These findings are even more impressive, taking into consideration that only 20% of the physicians responded to the dosing recommendations of the intervention group. This protocol, however, did not incorporate resistance data in the dosage adjustment calculation. Another study conducted in France (PharmAdapt) failed to find a significant benefit of TDM versus standard care. 31 Application of routine TDM in other patients groups (treatment-experienced patients) or for different drugs (NNRTIs, other PIs, combination of PIs) has still to be proven. 32 Additional clinical trials with improved design are needed to investigate if routine TDM as standard of care for the treatment of HIV infection is warranted. Several important limitations to the application of TDM for ART should be recognized, including uncertainty about the best PK predictor of response and insufficient validation of target concentrations for individual PIs and NNRTIs. The availability of a large TDM database may facilitate the establishment of expected concentration ranges for a variety of antiretroviral drugs. The concept of inhibitory quotient, which integrates drug concentrations and resistance testing, shows promise in a number of retrospective analyses. 13,14,33 A logistical problem remains with regard to its feasibility, and theoretical issues such as protein binding, variability, and the appropriate time of sampling continue to be debated. Available technology allows to set target inhibitory concentrations for a particular virus isolate based on genotypic and phenotypic sensitivity, however it is expensive and requires highly specialized assistance. 34 Ongoing research in the field of the HIV pharmacology is necessary for the future improvement of ART. CURRENT APPLICATION OF TDM IN CLINICAL PRACTICE The major reasons for TDM include individualizing therapy, preventing drug toxicity, and

verifying patient compliance. Poor adherence and pharmacokinetic factors may contribute to highly active antiretroviral therapy (HAART) failure. 34 Other factors such as degree of underlying resistance, drug tolerability and treatment history also play a role in determining virologic outcome. Today TDM has been proven to be most useful in selected patients groups: children, pregnant women, patients with renal and hepatic dysfunction and for the measurement of drug-drug interactions. 8,32,34 39,42,43 The recommendations of antiretroviral drugs for the treatment of HIV-infected pregnant women are subject to special considerations, including potential changes in dosing requirement and potential effects on the fetus or newborn. The pharmacokinetics of antiretroviral agents have not been adequately studied in pregnant women. The pharmacokinetic disposition of antiretroviral drugs may be profoundly altered during pregnancy as a result of dramatic physiologic, hormonal, and metabolic changes, which may lead to subtherapeutic or toxic systemic and intracellular drug levels in pregnant women. Available information is limited to the compounds currently used to decrease the risk of vertical transmission of HIV infection to the fetus. 35 37 Patients with renal or hepatic insufficiency are at increased risk of toxic concentrations for antiretroviral medications. The interindividual variability in plasma concentration has been reported to be higher in patients with liver dysfunction and examples of the application of TDM for HAART drugs in patients with liver impairment have been published. 38,39 Rapid changes in drug metabolism and distribution among pediatric patients, especially infants, can significantly alter their pharmacokinetic profiles. As with many other medications the extrapolation of the adult PK data and its application in pediatrics has been proven to have poor predictive value. 40 42 As children grow, drug pharmacokinetics are affected by changes in absorption, liver metabolizing capacity, plasma protein concentrations, body compartments, and renal function. Brundgade et al. 43 have shown that the dose based on pharmacokinetic data in older children may not be applicable to younger children. Most researchers agree that HIV-infected children may 10 be a target population for TDM application due to the unpredictability of plasma concentrations based on the administered dose. 34 Many clinicians feel drug concentrations may be of greatest utility shortly after initiating any new regimen, to ensure concentrations are adequate. Others see TDM only in the settings of previously failed therapy and in conjunction with resistance testing. 44 TDM for antiretroviral agents in HIV is available in a number of large reference centers in the United States and in Europe. The data from United Kingdom reported that the most common reasons for using TDM in HIV were: the administration of nonrecommended doses, suspected treatment failure, pharmacologic enhancement, suspected drug-drug or drug-food interaction, clinical symptoms of toxicity, change in therapy, and pediatric treatment. 8 The panel of experts in the field of pharmacology of antiretroviral agents held in Perugia in 2000 has agreed that TDM may represent a practical tool to improve the outcome of patients receiving HAART. The current limitations of routine clinical use of TDM include the insufficiency of data, lack of the assays standardization, and need for an expert data interpretation. The panel recommended further the conduct of large randomized trials to assess the clinical utility of TDM in the management of HIV-1 infection, assay standardization and educational programs for the caregivers and patients. 4 METHODOLOGY RAKHMANINA ET AL. The analytical criteria for TDM require a drug assay with high specificity, sensitivity, small sample volume, reasonable cost, and rapid turnaround time. 45 Methods for antiretroviral drug quantitation include high-performance chromatography and tandem-mass spectrometry (MS)/MS. 2,46 49 The tandem- MS/MS procedure developed at our laboratory is simple and allows for the quantitation of 16 HIV medications simultaneously in less than 5 minutes. 50 This procedure provides a better sensitivity and specificity than the high-performance liquid chromatography methods, which also require larger sample volumes, and are far more labor intensive. 51

THERAPEUTIC DRUG MONITORING 11 The ideal sample is drawn at steady state just before the next dose (steady-state trough sample). Steady state is reached after an interval of five drug half-lives have elapsed, which for most of these drugs is less than 48 hours. Exceptions to this are nevirapine and saquinavir, both of which have longer half-lives and should not be monitored until 4 days after initiation of the drug regimen. Most of the PIs have in vitro 95% inhibitory concentrations of approximately 100 ng/ml. At the Children s National Medical Center (CNMC) the lower limit for the therapeutic range for PIs has been 150 ng/ml. For the upper limit we recommend concentrations less than 6000 ng/ml, except for lopinavir, which we recommend should be maintained at concentrations less than 1200 ng/ml. These values were derived from the literature and are tentative therapeutic ranges. 7,9,11,12,25, 52,53 For the NNRTIs we recommend tentative therapeutic ranges between 1200 and 7000 ng/ml. External proficiency testing for the AIDS drugs is available from the International Quality Control Program for Therapeutic drug Monitoring in HIV infection (University Medical Center Nijmegen, The Netherlands). Currently, more than 65 laboratories worldwide are enrolled in this program. Drug standards can be obtained from the National Institutes of Health (NIH) AIDS Reagent Reference Program. CASE REPORT The following is a short illustration of the usefulness of TDM in pediatric practice at CNMC during a complex multiple drug regimen treatment of one of our patients. The patient was an African American girl, 9 years of age, with HIV/AIDS infection in category C3 per Centers for Disease Control (CDC) classification of pediatric HIV infection, a history of poor adherence with medications, encephalopathy, and severe failure to thrive with wasting. Her CD4 counts have dropped down to 3% (22 cells/mm 3 ) and her viral load was at 47,871 copies/ml. Because of profound immunosuppression she has developed disseminated Mycobacterium avium infection (MAI) with significant gastrointestinal disease, confirmed by small bowel biopsy. Her gut involvement was so severe that she was unable to tolerate any enteral intake including HAART therapy during a period of 9 weeks. Her MAI therapy included rifabutin, ethambutol, and amikacin via the intravenous route along with total parenteral nutrition (TPN), trimethoprim-sulphamethoxazole, metoclopramide, ranitidine, ondansetrone and methadone. Unfortunately, despite amikacin serum concentrations in the therapeutic range, she developed moderate sensorineural hearing loss bilaterally. Her MAI regimen was changed to a combination of rifabutin, ethambutol, and azithromycin. After a 9-week interruption of her HIV treatment, she was able to tolerate medications through her gastrojejunal tube and was restarted on HAART regimen consisting of didanosine, zidovudine, and kaletra (lopinavir/ ritonavir). Her new HAART regimen was chosen based on the results of the resistance profile by phenotype (PhenoSense, ViroLogic, South San Francisco, CA), which showed susceptibility to all antiretroviral drugs, except for nevirapine and nelfinavir. Because of known drug-drug interaction between lopinavir/ ritonavir and rifabutin (with lopinavir and ritonavir induced suppression of rifabutin metabolism and increase of rifabutin levels by 570% with lopinavir and by 350% with ritonavir administration) 34 the dose of rifabutin was decreased accordingly. There was also a concern of her gastrointestinal tract ability to absorb antiretroviral agents in view of severe MAI infiltration of the gastric and intestinal walls. To confirm adequate drug concentrations of her antiretroviral agents and to avoid unnecessary toxicity in combination therapy with drug-drug interactions the levels of her antiretroviral medications were obtained. It is important to mention that TDM was conducted in an inpatient setting where the administration of HAART and other medication was performed by medical personnel and assured 100% adherence with the regimen. The levels of the medications were determined at our laboratory using liquid chromatography tandem MS/MS. The trough levels were 18 ng/ml for ddi; 50 ng/ml for zidovudine; 189 ng/ml for ritonavir (reference range of 150 6000 ng/ml) and 10,700 ng/ml for lopinavir (with reference

range 150 12,000 ng/ml). The conclusion was made that the gastrointestinal absorption of HAART in our patient was adequate and the level of medications were in therapeutic and not in the toxic range, and no adjustments of the medications doses and regimen were warranted. Within 16 weeks of restarting HAART regimen she has increased her CD4 counts to 20% (179 cells/mm 3 ) and her viral load was undetectable at less than 400 copies per milliliter. This clinical case illustrates that TDM for antiretrovirals can be useful to monitor the therapeutic window of antiretroviral therapy with the complex treatment involving multiple drugs and toxicity could be avoided. 12 CONCLUSION Optimal care in HIV requires individualized management and ongoing attention to relevant scientific and clinical information in the field. 54 A growing body of literature supports the concept of TDM in HIV, but it is important that it is incorporated with other interventions such as resistance testing, adherence monitoring, and patient counseling to be an effective tool in patient management. Prospective randomized and double-blinded clinical trials are necessary to establish the role of concentrationtargeted therapy of HIV in routine clinical care. Properly applied TDM program can already benefit many HIV-infected patients on antiretroviral therapy, especially those at risk for subtherapeutic or toxic drug concentrations. REFERENCES 1. Acosta EP, Gerber JG. Adult Pharmacology Committee of the AIDS Clinical Trials Group. Position paper on therapeutic drug monitoring of antiretroviral agents. AIDS Res Hum Retroviruses 2002;18:825 834. 2. Soldin OP, Elin RJ, Soldin SJ. Therapeutic drug monitoring in human immunodeficiency virus/acquired immunodeficiency syndrome. Quo vadis. Arch Pathol Lab Med 2003;127:102 104. 3. Back DJ, Khoo SH, Gibbons SE, et al. Therapeutic drug monitoring of antiretrovirals in human immunodeficiency virus infection. Ther Drug Monit 2000;22: 122 126. 4. van Heeswijk RPG. Critical issues in therapeutic drug monitoring of antiretroviral drugs. Ther Drug Monit 2002;30:313 318. RAKHMANINA ET AL. 5. Casado JL, MorenoA, Sabido R, et al. A clinical study of the combination of 100 mg ritonavir plus 800 mg indinavir as salvage therapy: Influence of increased plasma drug levels in the rate of response. HIV Clin Trials 2000;1:13 19. 6. Lamotte C, Peytavin G, Perre P, et al. Increasing adverse events with indinavir dosages and plasma concentrations in four different ritonavir-indinavir containing regimens in HIV-infected patients [Abstract 738]. Eighth Conference on Retroviruses and Opportunistic Infections. Chicago, 2001. 7. Marzolini C, Telenti, Decosterd LA, et al. Efavirenz plasma levels can predict treatment failure and central nervous system side effects in HIV-infected patients. AIDS 2001;15:71 75. 8. Gibbons ES, Reynolds HE, TIJA JF et al. Therapeutic drug monitoring in the management of subjects on the protease inhibitors nelfinavir and saquinavir: results of the Roche UK TDM service [Abstract P259]. 5th International Congress on Drug therapy in HIV infection. Glasgow, 2000. 9. Hoetelmans RMW, Reijers MHE, Weverling GJ, et al. The effect of plasma drug concentrations on HIV-1 clearance rate during quadruple drug therapy. AIDS 1998;12:F111 F115. 10. Lebergeber B, Egger M, Opravil M, et al. Clinical progression and virologic failure of highly active antiretroviral therapy in HIV-1 patients: A prospective cohort stuffy. Lancet 1999;353:863 868. 11. Veldkamp AI, Weverling GJ, Lange JM, et al. High exposure to nevirapine in plasma is associated with improved virological response in HIV-1 infected individuals. AIDS 2001;15:1089 1095. 12. Durant J, Clevengergh P, Garaffo R, et al. Importance of protease inhibitors plasma levels in HIV-infected patients treated with genotypic-guided therapy: pharmacologic data from the Viradept study. AIDS 2000; 14:1333 1339. 13. Burger DM, Hugen PWH, Droste J, et al. Therapeutic drug monitoring of indinavir in treatment-naïve patients improves therapeutic outcome after one year: results from ATHENA [Abstract 6.2a]. 2nd International Workshop on Clinical Pharmacology of HIV Therapy. Noordwijk, 2001. 14. Burger DM, Hugen PWH, Droste J, et al. Therapeutic drug monitoring of nelfinavir 1250 BID in treatmentnaïve patients improves therapeutic outcome after one year: Results from ATHENA [Abstract 6.2b]. 2nd International Workshop on Clinical Pharmacology of HIV Therapy. Noordwijk, 2001. 15. De Maat MM, Huitema AD, Mulder JW, et al. Subtherapeutic antiretroviral plasma concentrations in routine clinical outpatient HIV care. Ther Drug Monit 2003;25:367 373. 16. Hugen PW, Burger DM, Aarnouste RE, Baede PA, et al. Therapeutic drug monitoring of HIV-protease inhibitors to assess non-compliance. Ther Drug Monit 2002;24:579 587. 17. Descamps D, Flandre P, Calvez V, et al. Mechanisms of virologic failure in previously untreated HIV-in-

THERAPEUTIC DRUG MONITORING 13 fected patients from a trial of induction-maintenance therapy. Trilege (Agence Nationale de Recherches sur le SIDA 072 Study Team). JAMA 2000;283:205 211. 18. Austin DJ, Ferguson NM, Fraser C, et al. Understanding antiretroviral treatment failure: the role of pharmacokinetic/dynamics and patient non-compliance in the emergence of resistance [Abstract 212]. 7th European Conference on Clinical Aspects and Treatment of HIV Infection. Lisbon, 1999. 19. Dasgupta A, Okhuysen PC. Pharmacokinetic and other drug interactions in patients with AIDS. Ther Drug Monit 2001;3:591 605. 20. Fletcher CV, Kawle SP, Kakuda TN, et al. Zidovudine triphosphate and lamivudine-triphopshate concentration-response relationship in HIV-infected persons. AIDS 2000;14:2137 2144. 21. Fletcher CV, Acosta EP, Henry K, et al. Concentrationcontrolled zidovudine therapy. Clin Pharmac Ther 1998;64:331 338. 22. Mallon PW, Ray J, Cooper DA. Effect of therapeutic drug monitoring on outcome in antiretroviral experienced HIV-infected individuals. J Clin Virol 2003; 26:223 227. 23. Van Heeswijk RP, Veldkamp AL, Mulder JW, et al. The steady-state pharmacokinetics of nevirapine during once daily and twice daily dosing of HIV-infected individuals. AIDS 2000;14:F77 82. 24. Joshi AS, Barrett JS, Fiske WD,t al. Population pharmacokinetics of efavirenz in phase II studies and relationship with efficacy [Abstract 1201]. 39th Interscience Conference on Antimicrobial Agents and Chemotherapy. San Francisco, 1999. 25. Fletcher CV, Anderson PL, Kakuda RTN, et al. Concentration-controlled compared with conventional antiretroviral therapy for HIV infection. AIDS 2002; 16:551 560. 26. Langmann P, Zilly M, Weissbrich B, et al. Therapeutic drug monitoring of indinavir in HIV-infected patients undergoing HAART. Infections 2002;30:13 16. 27. Decker CJ, Laitinen LM, Bridson GW, et al. Metabolism of amprenavir in liver microsomes: Role of CYP3A4 inhibition on drug interactions. J Pharm Sci 1998;87:803 807. 28. Barry C, Mulchany F, Merry C, et al. Pharmacokinetics and potential interactions amongst antiretroviral agents used to treat patients with HIV infection. Clin Pharmacokinet 1999;36:289 304. 29. Barry M, Gibbons S, Back D, et al. Protease inhibitors in patients with HIV disease. Clinically important pharmacologic considerations. Clin Pharmacokinet 1997;32:194 209. 30. Zhang XQ, Schooley RT, Gerber JG. The effect of increasing alpha 1-acid glycoprotein concentration on the antiviral efficacy of human immunodeficiency virus protease inhibitors. J Infect Dis 1999;180: 852 856. 31. Clevenbergh P, Garaffo R, Durant J, et al. PharmAdapt: a prospective multicenter randomized controlled trial to evaluate the usefulness of protease inhibitor therapeutic drug monitoring: 12 weeks result [Abstract 6.9]. 2nd International Workshop on Clinical Pharmacology of HIV Therapy. Noordwijk, 2001. 32. Aarnouste R, Schapiro J, Boucher C, et al. Therapeutic drug monitoring: An aid to optimizing response to antiretroviral drugs? Drugs 2003;63:741 753. 33. Piscitelli SC. The role of therapeutic drug monitoring in the management of HIV-infected patient. Curr Infect Dis Rep 2002;4:353 358. 34. Back D, Gatti G, Fletcher C, et al. Therapeutic drug monitoring in HIV infection: current status and future directions. AIDS 2002;16:S5 37. 35. Rodman JH, Flynn PM, Robbins B, et al. Systemic pharmacokinetics and cellular pharmacology of zidovudine in human immunodeficiency virus type 1- infected women and newborn infants. J Infect Dis 1999;180:1844 1850. 36. Mirochnick M, Fenton T, Gagnier P, et al. Pharmacokinetics of nevirapine in human immune deficiency virus type 1-infected pregnant women and their neonates. Pediatric AIDS Clinical Trials Group Protocol 250 Team. J Infect Dis 1998;178:368 374. 37. Moodley J, Moodley D, Pillay K, et al. Pharmacokinetics and antiretroviral activity of lamivudine alone or when coadministered with zidovudine in human immune deficiency virus type 1-infected pregnant women and their offspring. J Infect Dis 1998;178: 1327 1333. 38. Landman R, Peytavin G, Lamotte C, et al. Therapeutic drug monitoring of nelfinavir in a prospective study (LIVIR IMEA 014) in HIV-HCV co-infected patient with chronic liver disease [Abstract 6.4]. 2nd International Workshop on Clinical Pharmacology of HIV Therapy. Noordwijk, 2001. 39. Khaliq Y, Gallicano K, Seguin I, et al. Single and multiple dose pharmacokinetics of nelfinavir and CYP2C19 activity in human immunodeficiency virusinfected patients with chronic liver disease. Br J Clin Pharmacol 2000;50:108 115. 40. Fletcher CV, Brundage RC, Remmel Rp, et al. Pharmacologic characteristics of indinavir, didanosine, and stavudine in human immunodeficiency virusinfected children receiving combination therapy. Antimicrob Agents Chemother 2000;44:1029 1034. 41. Soldin OP, Soldin SJ. Review: Therapeutic drug monitoring in pediatrics. Ther Drug Monit 2002;24:1 8. 42. Gatti G, Vigano A, Sala N, et al. Indinavir pharmacokinetics and pharmacodynamics in children with human immunodeficiency virus infection. Antimocrob Agents Chemother 2000;44:752 755. 43. Brundage RC, Fletcher CV, Fenton T, et al. Efavirenz and nelfinavir pharmacokinetics in HIV infected children under 2 years of age [Abstract 719]. Seventh Conference on Retroviruses and Opportunistic Infections. San Francisco, 2000. 44. Flexner CW. Advances in HIV pharmacology: protein binding, pharmacogenomics, and the therapeutic drug monitoring. Top HIV Med 2003;11:40 44. 45. Gerber JG, Acosta EP. The potential role of therapeutic drug monitoring in the treatment of HIV infection. Top HIV Med 2002;10:27 32.

46. Titier K, Lagrange F, Pehourcq F, et al. High-performance liquid chromatographic method for the simultaneous determination of the six HIV-protease inhibitors and two non-nucleoside reverse transcriptase inhibitors in human plasma. Ther Drug Monit 2002; 24:417 424. 47. Villani P, Feoggio M, Gianelli L, et al. Antiretrovirals: simultaneous determination of five protease inhibitors and the nonnucleoside transcriptase inhibitors in human plasma by a rapid high-performance liquid chromatography-mass spectrometry assay. Ther Drug Monit 2002;24:380 388. 48. Volosov A, Alexander C, Ting L, Soldin SJ. Simple rapid method for quantification of antiretrovirals by liquid chromatography-tandem mass-spectrometry. Clin Biochem 2002;35:99 103. 49. Crommentuyn KM, Rosing H, Nan-Offeringa LG, et al. Rapid quantification of HIV protease inhibitors in human plasma by high-performance liquid chromatography coupled with electrospray ionization tandem mass spectrometry. J Mass Spectrom 2003; 38:157 166. 50. Ghoshal AK, Soldin SJ. Improved method for concurrent quantification of antiretrovirals by liquid chromatography-tandem mass spectrometry. Ther Drug Monit 2003;25:541 543. 14 RAKHMANINA ET AL. 51. Moyer TP, Tmesgen Z, Enger R, et al. Drug monitoring of antiretroviral therapy for HIV-1 infection: Method validation and results of the pilot study. Clin Chem 1999;45:1465 1476. 52. Anderson PL, Fletcher CV. Clinic pharmacologic considerations for HIV-1 protease inhibitors. Curr Infect Dis Rep 2001;3:381 387. 53. Gatti G, Di Biagio A, Casazza R, et al. The relationship between ritonavir plasma levels and side effects: implication for therapeutic drug monitoring. AIDS 1999;13:2083 2089. 54. Carpenter CC, Cooper DA, Fishl MA, et al. Antiretroviral therapy in adults: Updated recommendations of the international AIDS Society-USA Panel. JAMA 2000;283:381 390. Address reprint requests to: Natella Y. Rakhmanina, M.D. Special Immunology Program Children s National Medical Center 111 Michigan Avenue, NW Washington, D.C. 20010-2970 E-mail: nrakhmanina@cnmc.org