1
|
Chaira T, Subramani C, Barman TK. ADME, Pharmacokinetic Scaling, Pharmacodynamic and Prediction of Human Dose and Regimen of Novel Antiviral Drugs. Pharmaceutics 2023; 15:pharmaceutics15041212. [PMID: 37111697 PMCID: PMC10146820 DOI: 10.3390/pharmaceutics15041212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 03/29/2023] [Accepted: 04/07/2023] [Indexed: 04/29/2023] Open
Abstract
The search for new drugs is an extremely time-consuming and expensive endeavour. Much of that time and money go into generating predictive human pharmacokinetic profiles from preclinical efficacy and safety animal data. These pharmacokinetic profiles are used to prioritize or minimize the attrition at later stages of the drug discovery process. In the area of antiviral drug research, these pharmacokinetic profiles are equally important for the optimization, estimation of half-life, determination of effective dose, and dosing regimen, in humans. In this article we have highlighted three important aspects of these profiles. First, the impact of plasma protein binding on two primary pharmacokinetic parameters-volume of distribution and clearance. Second, interdependence of primary parameters on unbound fraction of the drug. Third, the ability to extrapolate human pharmacokinetic parameters and concentration time profiles from animal profiles.
Collapse
Affiliation(s)
- Tridib Chaira
- Department of Pharmacology, SGT University, Gurugram 122505, Haryana, India
| | - Chandru Subramani
- Department of Pathology, Galveston National Laboratory, University of Texas Medical Branch, Galveston, TX 77550, USA
| | - Tarani Kanta Barman
- Department of Pathology, Galveston National Laboratory, University of Texas Medical Branch, Galveston, TX 77550, USA
| |
Collapse
|
2
|
Boffito M, Jackson A, Pozniak A, Giraudon M, Kulkarni R, Abelardo MC, Patel IH, Morcos PN. Effect of a modified saquinavir/ritonavir dosing regimen with lower dose lead-in phase on QTc interval, pharmacokinetics, antiviral activity and safety in treatment-naïve HIV-1-infected patients. Drugs R D 2015; 15:141-53. [PMID: 25742730 PMCID: PMC4359187 DOI: 10.1007/s40268-015-0087-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
Background Saquinavir/ritonavir (1000/100 mg twice daily [BID]) is associated with dose- and exposure-dependent prolongation of the QT interval. The QT risk is considered higher during the first week of therapy, when saquinavir peak exposure has been observed. A modified regimen with a lower dose lead-in phase may reduce potential saquinavir-/ritonavir-induced QT prolongations. Objective To explore the effect of the modified saquinavir/ritonavir regimen on QT interval, pharmacokinetics, antiviral activity, and safety in treatment-naïve HIV-1-infected patients. Methods Twenty-three HIV-1-infected treatment-naïve patients received saquinavir/ritonavir 500/100 mg BID on days 1–7 and 1000/100 mg BID on days 8–14 in combination with two nucleoside reverse transcriptase inhibitors. The primary endpoint was mean maximum change from dense predose baseline in QT values corrected using Fridericia’s formula (∆QTcFdense) across study days. Secondary endpoints included maximum change from time-matched baseline in QTcF, antiviral activity, pharmacokinetics, and safety over the 14 days. Results The mean maximum ∆QTcFdense was 3, 1, 7, 12, and 7 ms on days 3, 4, 7, 10, and 14, respectively. Across all study days, 2/21 patients had a maximum ∆QTcFdense ≥30 ms (on day 10); the highest mean ∆QTcFdense was <10 ms. During week 1, saquinavir exposure was highest on day 3 and lowest on day 7. All patients showed continuous declines in HIV-RNA; none experienced virologic breakthrough/rebound. The modified regimen was generally well tolerated. Conclusion Treatment initiation with the modified saquinavir/ritonavir regimen in treatment-naïve HIV-1-infected patients reduced saquinavir exposure during week 1, potentially mitigating/reducing QT liability while suppressing HIV-RNA during the course of treatment.
Collapse
Affiliation(s)
- Marta Boffito
- Chelsea and Westminster Hospital NHS Foundation Trust, London, UK
| | | | | | | | | | | | | | | |
Collapse
|
3
|
Luber AD, Condoluci DV, Slowinski PD, Andrews M, Olson K, Peloquin CA, Pappa KA, Pakes GE. Steady-state amprenavir and tenofovir pharmacokinetics after coadministration of unboosted or ritonavir-boosted fosamprenavir with tenofovir disoproxil fumarate in healthy volunteers. HIV Med 2009; 11:193-9. [PMID: 19863619 DOI: 10.1111/j.1468-1293.2009.00765.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Objective An open-label, three-period pharmacokinetic study was conducted to investigate the drug interaction potential between fosamprenavir (FPV) and tenofovir disoproxil fumarate (TDF). Methods Thirty-six healthy subjects received TDF 300 mg once daily (qd) for 7 days (period 1), and then were randomized to 14 days of either FPV 1400 mg twice daily (bid) or FPV/ritonavir (RTV) 700/100 mg bid alone or with TDF (period 2). Subjects continued their randomized dose of FPV for 14 more days, adding or removing TDF based upon its receipt in period 2 (period 3). Twenty-four-hour pharmacokinetic sampling was carried out on day 7 of period 1 and on day 14 of periods 2 and 3. Steady-state plasma amprenavir (APV) and tenofovir (TFV) pharmacokinetics were assessed by noncompartmental analysis and parameter values observed with each regimen were compared using geometric mean ratios with 90% confidence intervals. Results After TDF coadministration, APV geometric mean minimum concentration (C(min)), maximum concentration (C(max)), and area under the plasma concentration-time curve (AUC) increased by 31, 3 and 7% above values observed with unboosted FPV alone; they also increased by 31, 4 and 16% above values observed with FPV/RTV alone. TFV C(min), C(max) and AUC decreased by 12, 25 and 15% after FPV coadministration and by 9, 18 and 7% after FPV/RTV coadministration. No significant changes in RTV pharmacokinetics were observed. No differences were noted in adverse events among dosing periods. Conclusions In this evaluation of the interaction between FPV and TDF, increases in APV exposures and modest decreases in TFV exposures were observed. These were unlikely to be clinically significant.
Collapse
|
4
|
Cytochrome P450 3A inhibition by atazanavir and ritonavir, but not demography or drug formulation, influences saquinavir population pharmacokinetics in human immunodeficiency virus type 1-infected adults. Antimicrob Agents Chemother 2009; 53:3524-7. [PMID: 19528289 DOI: 10.1128/aac.00025-09] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Inadequate concentrations of the human immunodeficiency virus (HIV) protease inhibitor saquinavir jeopardize individual therapy success or produce side effects despite treatment according to the current guidelines. We performed a population pharmacokinetic analysis with NONMEM and determined that the steady-state pharmacokinetics of saquinavir in 136 HIV type 1-infected adults was modulated by a decrease in saquinavir CL following coadministration of the cytochrome P450 3A inhibitors ritonavir and atazanavir. In contrast, age, sex, weight, pregnancy, and the pharmaceutical formulation exerted only minor, nonsignificant effects.
Collapse
|
5
|
Dickinson L, Boffito M, Back DJ, Khoo SH, Pozniak AL, Mugyenyi P, Merry C, Autar RS, Burger DM, Aarons LJ. Population pharmacokinetics of ritonavir-boosted saquinavir regimens in HIV-infected individuals. J Antimicrob Chemother 2008; 62:1344-55. [PMID: 18824460 PMCID: PMC3597129 DOI: 10.1093/jac/dkn399] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
OBJECTIVES The aim of this study was to develop and validate a population pharmacokinetic model in order to describe ritonavir-boosted saquinavir concentrations dosed twice and once daily in human immunodeficiency virus (HIV)-infected patients from the UK, Uganda and Thailand and to identify factors that may influence saquinavir pharmacokinetics. METHODS Pharmacokinetic data from 10 clinical studies were combined. Non-linear mixed effects modelling (NONMEM version V) was applied to determine the saquinavir pharmacokinetic parameters, interindividual/interoccasion variability (IIV/IOV) and residual error. Various covariates potentially related to saquinavir pharmacokinetics were explored, and the final model was validated by means of 95% prediction interval and testing the predictive performance of the model with data not included in the model-building process. RESULTS Ninety-seven patients were included from the UK (n = 52), Uganda (n = 18) and Thailand (n = 27), contributing 347 saquinavir profiles (1-14 profiles per patient). A one-compartment model with zero-order absorption and lag-time best described the data with IIV/IOV on apparent oral clearance (CL/F) and volume of distribution (V/F) and with IIV on duration and absorption lag-time. The ritonavir area under the curve over the dosing interval was significantly associated with saquinavir CL/F and V/F. A typical patient from the UK had approximately 1.5- and 3-fold higher saquinavir CL/F compared with patients from Uganda (89.0 versus 49.8 L/h) and Thailand (89.0 versus 26.7 L/h), respectively. CONCLUSIONS A model to characterize ritonavir-boosted saquinavir pharmacokinetics in HIV-infected adults has been developed and validated. The model could be used for dosage adaptation following therapeutic drug monitoring and to assess patients' suitability for once-daily boosted saquinavir therapy.
Collapse
Affiliation(s)
- Laura Dickinson
- NIHR National Biomedical Research Centre, Royal Liverpool and Broadgreen University Hospital Trust, Liverpool, UK
- Department of Pharmacology and Therapeutics, University of Liverpool, Liverpool, UK
| | - Marta Boffito
- St Stephen’s Centre, Chelsea and Westminster Foundation Trust, London, UK
| | - David J. Back
- Department of Pharmacology and Therapeutics, University of Liverpool, Liverpool, UK
| | - Saye H. Khoo
- Department of Pharmacology and Therapeutics, University of Liverpool, Liverpool, UK
| | - Anton L. Pozniak
- St Stephen’s Centre, Chelsea and Westminster Foundation Trust, London, UK
| | | | - Concepta Merry
- Department of Pharmacology, Trinity College Dublin, Dublin, Ireland
| | | | - David M. Burger
- Department of Clinical Pharmacy, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | - Leon J. Aarons
- School of Pharmacy and Pharmaceutical Sciences, University of Manchester, Manchester, UK
| |
Collapse
|
6
|
Dickinson L, Boffito M, Khoo SH, Schutz M, Aarons LJ, Pozniak AL, Back DJ. Pharmacokinetic analysis to assess forgiveness of boosted saquinavir regimens for missed or late dosing. J Antimicrob Chemother 2008; 62:161-7. [PMID: 18467305 PMCID: PMC3672987 DOI: 10.1093/jac/dkn187] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
OBJECTIVES One potential concern of once-daily protease inhibitor administration is low trough concentrations and ultimately the 'forgiveness' or robustness in comparison with the originally licensed twice-daily dose. To give an estimation of 'forgiveness', we determined the length of time plasma drug concentrations were below target in HIV-infected patients receiving saquinavir/ritonavir regimens. METHODS Seventy-seven pharmacokinetic profiles (saquinavir/ritonavir 1000/100 mg twice daily, n = 34; 1600/100 mg once daily, n = 26; 2000/100 mg once daily, n = 17) from five studies were combined, presented as twice- and once-daily percentiles (P10-P90) and compared. At percentiles where trough concentrations fell below the alleged minimum effective concentration (MEC; 100 ng/mL), the length of time below MEC was determined. RESULTS Saquinavir concentrations were below MEC at P10 for 0.7 h for twice-daily saquinavir/ritonavir when compared with 8.6 and 6.6 h for 1600/100 and 2000/100 mg once daily, respectively. At P25, 1600/100 mg once daily produced suboptimal concentrations for 5.5 h in contrast to 0.5 h for 2000/100 mg once daily. CONCLUSIONS Here, we provide substantive data that indicate once-daily saquinavir, in particular 1600/100 mg, is not as robust as the twice-daily regimen based on a population of UK patients; this raises concern over late or missed doses. However, pharmacokinetic data can only ever be a guide to the impact on long-term efficacy.
Collapse
Affiliation(s)
- Laura Dickinson
- Department of Pharmacology, University of Liverpool, Liverpool, UK.
| | | | | | | | | | | | | |
Collapse
|
7
|
Pulido F, Fiorante S. Datos clínicos II. Experiencia clínica de tenofovir en combinaciones con inhibidores de proteasa. Enferm Infecc Microbiol Clin 2008; 26 Suppl 8:13-8. [DOI: 10.1157/13126267] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
8
|
Differences in the pharmacokinetics of protease inhibitors between healthy volunteers and HIV-infected persons. Curr Opin HIV AIDS 2008; 3:296-305. [DOI: 10.1097/coh.0b013e3282f82bf1] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
9
|
Dickinson L, Back D, Pozniak A, Khoo S, Boffito M. Limited-sampling strategy for the prediction of boosted hard-gel saquinavir exposure at a dosage of 1000/100 mg twice daily in human immunodeficiency virus-infected individuals. Ther Drug Monit 2007; 29:361-7. [PMID: 17529895 DOI: 10.1097/ftd.0b013e3180683b25] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Area under the concentration time curve (AUC) over a dosing interval is considered to be the best estimate of drug exposure in a patient. However, determination of this parameter is costly and often impractical, requiring multiple samples and a great deal of time and resources. A limited-sampling strategy (LSS) may overcome some of these issues, making pharmacokinetic studies easier to perform, particularly in a limited-resource setting. The aim of this work was to develop and validate a pragmatic LSS for the accurate and precise prediction of boosted saquinavir AUC0-12 (AUC over the 12-hour dosing interval) at a dosage of 1000/100 mg twice daily. Pharmacokinetic data were obtained from 34 human immunodeficiency virus (HIV)-infected individuals stable on saquinavir/ritonavir-containing therapy, randomly split into two sets (n = 17 per set). One set was used to construct prediction models using univariate and multivariate analysis (development set), and the second was used to determine the predictive performance of the models (validation set). For single samples, 6- and 10-hour concentrations correlated best with saquinavir AUC0-12 (r2: 0.913 and 0.911, respectively), yet all single samples failed to produce precise and unbiased predictions. However, combinations at 2, 6; 0, 2, 6; 0, 4, 10; 0, 4, 12; and 2, 4, 6 hours achieved good predictive performances, and both precise [root mean squared relative prediction error (%RMSE): 6.4% to 11.9%] and unbiased [mean relative prediction error (%MPE), 95% CI: -2.7%, (-0.8)-2.7 to 1.6%, (-1.8)-4.7] estimations of saquinavir AUC0-12. Of these models, concentrations obtained at 0, 2, 6 and 2, 4, 6 hours are more practical in a clinical setting and are therefore the LSS with most potential. Provided that the technique is validated in specific patient populations, an LSS approach is a potentially useful tool to evaluate the AUC0-12 of saquinavir in resource-limited settings, reducing both costs and volumes of blood taken. It may also aid the choice of sampling times for population analysis.
Collapse
Affiliation(s)
- Laura Dickinson
- Department of Pharmacology, University of Liverpool, Liverpool, UK.
| | | | | | | | | |
Collapse
|
10
|
Hoetelmans RMW, Mariën K, De Pauw M, Hill A, Peeters M, Sekar V, De Doncker P, Woodfall B, Lefebvre E. Pharmacokinetic interaction between TMC114/ritonavir and tenofovir disoproxil fumarate in healthy volunteers. Br J Clin Pharmacol 2007; 64:655-61. [PMID: 17610528 PMCID: PMC2203266 DOI: 10.1111/j.1365-2125.2007.02957.x] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
AIM TMC114 is a new HIV protease inhibitor, used in combination with low-dose ritonavir (TMC114/r) as a pharmacokinetic enhancer. Tenofovir disoproxil fumarate (TDF) is a nucleotide reverse transcriptase inhibitor. Both antiretrovirals show activity against wild-type and resistant HIV. An open-label crossover study was conducted in HIV - healthy volunteers to investigate the potential for a pharmacokinetic interaction between TMC114/r and tenofovir. METHODS Two groups, each of six volunteers, were evaluated in two consecutive sessions. In session 1, volunteers received TMC114/r (300/100 mg bid) for 7 days, followed by a wash-out period of at least 6 days. In session 2, volunteers received TMC114/r (300/100 mg bid) plus TDF (300 mg qd). RESULTS When TMC114/r and TDF were coadministered, tenofovir plasma concentrations (C(min) and C(max)), and area under the curve (AUC(24 h)) increased by 37%, 24% and 22%, respectively. When TDF and ritonavir were coadministered, TMC114 plasma C(min), C(max) and AUC(12 h) increased by 24%, 16% and 21%, respectively. There were no changes in the urinary excretion of unchanged tenofovir or TMC114 during coadministration. Administration of TMC114/r in HIV- healthy volunteers with or without TDF was well tolerated. CONCLUSIONS The interaction between TMC114/r and tenofovir is not clinically relevant and no dose adjustments are required when these drugs are coadministered.
Collapse
|
11
|
Marin-Niebla A, Lopez-Cortes LF, Ruiz-Valderas R, Viciana P, Mata R, Gutierrez A, Pascual R, Rodriguez M. Clinical and pharmacokinetic data support once-daily low-dose boosted saquinavir (1,200 milligrams saquinavir with 100 milligrams ritonavir) in treatment-naive or limited protease inhibitor-experienced human immunodeficiency virus-infected patients. Antimicrob Agents Chemother 2007; 51:2035-42. [PMID: 17371813 PMCID: PMC1891384 DOI: 10.1128/aac.01136-06] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
We evaluated the plasma and intracellular pharmacokinetics, clinical efficacy, and safety of once-daily low-dose boosted saquinavir (SQVr; 1,200 of saquinavir [SQV] with 100 mg of ritonavir) plus two nucleotide reverse transcriptase inhibitors in treatment-naive or limited protease inhibitor (PI)-experienced human immunodeficiency virus (HIV)-infected patients. A prospective study without entry restrictions on the plasma HIV-RNA (VL) or CD4 cell count was carried out. Plasma and intracellular SQV levels were measured by high-performance liquid chromatography. Efficacy was evaluated by an intention-to-treat analysis; treatment failure was defined as virological failure (a VL of >50 copies/ml after 24 weeks or a confirmed rebound to >50 copies/ml) or interruption for any reason. A total of 151 patients were included in the study (106 of them either had never received PI or had no previous virological failure on PIs) and could be characterized as follows: previous C3 stage, 28.9%; injection-drug users, 69.1%; subjects with chronic viral hepatitis, 53%; and subjects with cirrhosis, 10%. The median baseline CD4 level was 184/mul, and the median VL was 4.8 log(10) copies/ml. Median C(max), area under the concentration-time curve from 0 to 24 h, and C(min) plasma and intracellular SQV levels were 3,672 and 10,105 ng/ml, 34,283 and 99,535 ng.h/ml, and 359 and 1,062 ng/ml, respectively. The efficacy as determined by intention to treat at 52 weeks was 69.7% (96% in the on-treatment analysis), with similar results regardless of the baseline VL and CD4 counts. Only five patients had virological failure despite adequate C(min) levels, but with a poor adherence (the only variable related to virological failure). Adverse events caused the withdrawal of the treatment in four patients (2.6%). In conclusion, given the pharmacokinetic profile, efficacy, and tolerability of this regimen, once-daily low-dose SQVr may be considered a treatment option in treatment-naive or limited PI-experienced HIV-infected patients, with the additional benefit of being currently the least-expensive PI-based regimen available.
Collapse
Affiliation(s)
- Ana Marin-Niebla
- Servicio de Hematología, Hospital Universitario Virgen del Rocío, Avda. Manuel Siurot s/n, 41013 Seville, Spain.
| | | | | | | | | | | | | | | |
Collapse
|
12
|
Gazzard BG. Use of tenofovir disoproxil fumarate and emtricitabine combination in HIV-infected patients. Expert Opin Pharmacother 2006; 7:793-802. [PMID: 16556093 DOI: 10.1517/14656566.7.6.793] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
With the continuing spread of HIV infection, particularly in developing countries, cost-effective treatment for its management is a high priority. Truvada (Gilead Sciences) is a single combination pill of the nucleotide reverse transcriptase inhibitors tenofovir disoproxil fumarate and emtricitabine, which is used once daily. It is anticipated to be a clinically potent combination that is free of short-term irritating toxicity. The drug has recently been licensed but there are currently little clinical efficacy data regarding its use. The limited published data have indicated that emtricitabine and lamivudine have equivalent potency, and randomised controlled trials have produced evidence of the efficacy of lamivudine combined with tenofovir disoproxil fumarate in a regimen containing either the non-nucleoside reverse transcriptase inhibitor efavirenz or a protease inhibitor lopinavir/ritonavir. In these trials, long-term durability data are available for < or = 96 weeks.
Collapse
|
13
|
Chittick GE, Zong J, Blum MR, Sorbel JJ, Begley JA, Adda N, Kearney BP. Pharmacokinetics of tenofovir disoproxil fumarate and ritonavir-boosted saquinavir mesylate administered alone or in combination at steady state. Antimicrob Agents Chemother 2006; 50:1304-10. [PMID: 16569845 PMCID: PMC1426992 DOI: 10.1128/aac.50.4.1304-1310.2006] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
A phase I study was conducted to formally evaluate the steady-state pharmacokinetics (PK) of tenofovir disoproxil fumarate (TDF) and ritonavir (RTV)-boosted saquinavir mesylate (SQV) when coadministered in healthy volunteers. Forty subjects received multiple doses of TDF (300 mg, once daily) and SQV/RTV (1,000 mg/100 mg, twice daily) alone and together under steady-state conditions in an open-label, fixed sequence design. Blood samples for tenofovir (TFV) and SQV/RTV PK were drawn over respective 24- and 12-h dosing intervals, and drug concentrations were measured by liquid chromatography-tandem mass spectrometry. Safety was assessed periodically by clinical and laboratory monitoring. Thirty-two subjects completed the study and were fully evaluable; three subjects discontinued participation in the study due to adverse events, three subjects withdrew for personal reasons, and two subjects withdrew because of inadequate venous access for blood sampling. Steady-state TFV PK were not significantly altered upon coadministration with SQV/RTV. Steady-state SQV (administered as SQV/RTV) AUCtau, Cmax, and Ctau increased 29, 22, and 47%, respectively, upon coadministration with TDF, and all subjects achieved a Ctau of >100 ng/ml. These modestly increased SQV exposures are not clinically meaningful given its clinical use with RTV already results in >10-fold-higher SQV levels. Steady-state RTV AUCtau and Cmax levels were not significantly altered, whereas Ctau was 23% higher upon coadministration of SQV/RTV and TDF. Thus, no clinically relevant interactions between TDF and RTV-boosted SQV were observed under conditions simulating clinical practice.
Collapse
Affiliation(s)
- Gregory E Chittick
- Gilead Sciences, Inc., Department of Clinical Pharmacology and Pharmacokinetics, 4 University Place, 4611 University Dr., Durham, NC 27707, USA
| | | | | | | | | | | | | |
Collapse
|
14
|
Andrews KT, Fairlie DP, Madala PK, Ray J, Wyatt DM, Hilton PM, Melville LA, Beattie L, Gardiner DL, Reid RC, Stoermer MJ, Skinner-Adams T, Berry C, McCarthy JS. Potencies of human immunodeficiency virus protease inhibitors in vitro against Plasmodium falciparum and in vivo against murine malaria. Antimicrob Agents Chemother 2006; 50:639-48. [PMID: 16436721 PMCID: PMC1366900 DOI: 10.1128/aac.50.2.639-648.2006] [Citation(s) in RCA: 104] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Parasite resistance to antimalarial drugs is a serious threat to human health, and novel agents that act on enzymes essential for parasite metabolism, such as proteases, are attractive targets for drug development. Recent studies have shown that clinically utilized human immunodeficiency virus (HIV) protease inhibitors can inhibit the in vitro growth of Plasmodium falciparum at or below concentrations found in human plasma after oral drug administration. The most potent in vitro antimalarial effects have been obtained for parasites treated with saquinavir, ritonavir, or lopinavir, findings confirmed in this study for a genetically distinct P. falciparum line (3D7). To investigate the potential in vivo activity of antiretroviral protease inhibitors (ARPIs) against malaria, we examined the effect of ARPI combinations in a murine model of malaria. In mice infected with Plasmodium chabaudi AS and treated orally with ritonavir-saquinavir or ritonavir-lopinavir, a delay in patency and a significant attenuation of parasitemia were observed. Using modeling and ligand docking studies we examined putative ligand binding sites of ARPIs in aspartyl proteases of P. falciparum (plasmepsins II and IV) and P. chabaudi (plasmepsin) and found that these in silico analyses support the antimalarial activity hypothesized to be mediated through inhibition of these enzymes. In addition, in vitro enzyme assays demonstrated that P. falciparum plasmepsins II and IV are both inhibited by the ARPIs saquinavir, ritonavir, and lopinavir. The combined results suggest that ARPIs have useful antimalarial activity that may be especially relevant in geographical regions where HIV and P. falciparum infections are both endemic.
Collapse
Affiliation(s)
- Katherine T Andrews
- Queensland Institute of Medical Research and Australian Centre for International and Tropical Health and Nutrition, 300 Herston Road, Herston, Queensland 4029, Australia.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Piketty C, Gérard L, Chazallon C, Marcelin AG, Clavel F, Taburet AM, Calvez V, Madelaine-Chambrin I, Molina JM, Aboulker JP, Girard PM. Salvage Therapy with Atazanavir/Ritonavir Combined to Tenofovir in HIV-Infected Patients with Multiple Treatment Failures: Randomized Anrs 107 Trial. Antivir Ther 2006. [DOI: 10.1177/135965350601100213] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Background Ritonavir (RTV)-boosted atazanavir (ATV) and tenofovir disoproxil fumarate (TDF-DF) are promising in highly experienced patients because of their pharmacokinetic profile, activity, safety and resistance properties. Methods A 26-week study of the safety and efficacy of RTV-boosted ATV plus TDF-DF was conducted in 53 HIV-infected patients who were failing their current highly active antiretroviral therapy (HAART) regimen. Patients with history of failure to at least two protease inhibitors (PIs) and one non-nucleoside reverse transcriptase inhibitor (NNRTI) were randomized to either continue their current regimen (group 1) or replace the PI by ATV (300 mg once daily) boosted by RTV (100 mg; group 2) for 2 weeks. Then, all patients received the same combination of ATV, RTV and TDF-DF (300 mg) plus optimized NRTIs regimen. Results At baseline, median CD4+ T-cell count was 206/mm3, median viral load (VL) 5.0 log10/ml and median numbers of NRTI, NNRTI and PI resistance mutations were 7, 1 and 8, respectively. At week 2, median VL remained unchanged from baseline in group 2 as compared with group 1 (-0.1 vs -0.1 log10/ml). At week 26, a mild decrease in median VL from baseline of 0.2 log10/ml was observed, with 16 (31%) and 9 (17%) patients exhibiting a decrease in viral load of at least 0.5 and 1.0 log10/ml, respectively. Baseline phenotypic and genotypic resistance to ATV were the most predictive independent factors of virological response. The regimen was well tolerated. Conclusion In these very advanced patients failing highly HAART, the combination of boosted ATV plus TDF-DF yielded low antiretroviral activity.
Collapse
Affiliation(s)
- Christophe Piketty
- Department of Immunology, Hôpital Georges Pompidou, Assistance Publique-Hôpitaux de Paris, Paris, France
| | | | | | - Anne-Geneviève Marcelin
- Department of Virology, Hôpital Pitié Salpetrière, Assistance Publique-Hôpitaux de Paris, Paris, Francis
| | - François Clavel
- Department of Virology, Hôpital Bichat, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Anne-Marie Taburet
- Clinical Pharmacy Department, Hôpital Bicêtre, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Vincent Calvez
- Department of Virology, Hôpital Pitié Salpetrière, Assistance Publique-Hôpitaux de Paris, Paris, Francis
| | | | - Jean-Michel Molina
- Department of Infectious Diseases Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris, Paris
| | | | - Pierre-Marie Girard
- Department of Infectious Diseases Hôpital Saint Antoine, Assistance Publique-Hôpitaux de Paris, Paris, France
| |
Collapse
|
16
|
Boffito M, Pozniak A, Kearney BP, Higgs C, Mathias A, Zhong L, Shah J. Lack of pharmacokinetic drug interaction between tenofovir disoproxil fumarate and nelfinavir mesylate. Antimicrob Agents Chemother 2006; 49:4386-9. [PMID: 16189129 PMCID: PMC1251521 DOI: 10.1128/aac.49.10.4386-4389.2005] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
A study explored the pharmacokinetics of tenofovir (300 mg administered once daily) and nelfinavir (1,250 mg twice daily) when coadministered in 29 healthy volunteers. Tenofovir, nelfinavir, and M8 pharmacokinetics was unaltered when tenofovir and nelfinavir were coadministered, and tenofovir administration did not affect the M8/nelfinavir area under the concentration-versus-time curve over the dosing interval (AUC(tau)) ratio. No interaction between tenofovir and nelfinavir was observed.
Collapse
Affiliation(s)
- Marta Boffito
- PK Research, St. Stephen's Centre, Chelsea and Westminster Hospital, London, UK.
| | | | | | | | | | | | | |
Collapse
|
17
|
Dickinson L, Robinson L, Tjia J, Khoo S, Back D. Simultaneous determination of HIV protease inhibitors amprenavir, atazanavir, indinavir, lopinavir, nelfinavir, ritonavir and saquinavir in human plasma by high-performance liquid chromatography-tandem mass spectrometry. J Chromatogr B Analyt Technol Biomed Life Sci 2005; 829:82-90. [PMID: 16226495 DOI: 10.1016/j.jchromb.2005.09.032] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2005] [Revised: 09/20/2005] [Accepted: 09/23/2005] [Indexed: 10/25/2022]
Abstract
We report a precise and accurate method for simultaneous quantification of protease inhibitors (PIs) amprenavir, atazanavir, indinavir, lopinavir, nelfinavir, ritonavir and saquinavir in plasma. An internal standard was added to samples prior to protein precipitation with acetonitrile followed by addition of ammonium formate buffer. Analysis was by HPLC-MS/MS. Calibration curves were validated over concentration ranges encompassing both subtherapeutic and potentially 'toxic' drug concentrations. Inter- and intra-assay variation were below 11% and PI recovery was above 87%. The bioanalytical method described is successfully applied to measure PI concentrations obtained from clinical pharmacokinetic studies and routine therapeutic drug monitoring (TDM).
Collapse
Affiliation(s)
- Laura Dickinson
- Department of Pharmacology, University of Liverpool, Pharmacology Research Laboratories, Block H, First Floor, 70 Pembroke Place, Liverpool L69 3GF, UK.
| | | | | | | | | |
Collapse
|