1
|
Abedi Dorcheh F, Balmeh N, Hejazi SH, Allahyari Fard N. Investigation of the mutated antimicrobial peptides to inhibit ACE2, TMPRSS2 and GRP78 receptors of SARS-CoV-2 and angiotensin II type 1 receptor (AT1R) as well as controlling COVID-19 disease. J Biomol Struct Dyn 2025; 43:1641-1664. [PMID: 38109185 DOI: 10.1080/07391102.2023.2292307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/08/2022] [Accepted: 11/23/2023] [Indexed: 12/19/2023]
Abstract
SARS-CoV-2 is a global problem nowadays. Based on studies, some human receptors are involved in binding to SARS-CoV-2. Thus, the inhibition of these receptors can be effective in the treatment of Covid-19. Because of the proven benefits of antimicrobial peptides (AMPs) and the side effects of chemical drugs, they can be known as an alternative to recent medicines. RCSB PDB to obtain PDB id, StraPep and PhytAMP to acquire Bio-AMPs information and 3-D structure, and AlgPred, Toxinpred, TargetAntiAngio, IL-4pred, IL-6pred, ACPred and Hemopred databases were used to find the best score peptide features. HADDOCK 2.2 was used for molecular docking analysis, and UCSF Chimera software version 1.15, SWISS-MODEL and BIOVIA Discovery Studio Visualizer4.5 were used for mutation and structure modeling. Furthermore, MD simulation results were achieved from GROMACS 4.6.5. Based on the obtained results, the Moricin peptide was found to have the best affinity for ACE2. Moreover, Bacteriocin leucocin-A had the highest affinity for GRP78, Cathelicidin-6 had the best affinity for AT1R, and Bacteriocin PlnK had the best binding affinity for TMPRSS2. Additionally, Bacteriocin glycocin F, Bacteriocin lactococcin-G subunit beta and Cathelicidin-6 peptides were the most common compounds among the four receptors. However, these peptides also have some side effects. Consequently, the mutation eliminated the side effects, and MD simulation results indicated that the mutation proved the result of the docking analysis. The effect of AMPs on ACE2, GRP78, TMPRSS2 and AT1R receptors can be a novel treatment for Covid-19.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Fatemeh Abedi Dorcheh
- Department of Biotechnology, School of Bioscience and Biotechnology, Shahid Ashrafi Esfahani University of Isfahan, Sepahan Shahr, Iran
| | - Negar Balmeh
- Skin Diseases and Leishmaniasis Research Center, Department of Parasitology and Mycology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Seyed Hossein Hejazi
- Skin Diseases and Leishmaniasis Research Center, Department of Parasitology and Mycology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Najaf Allahyari Fard
- Department of Systems Biotechnology, National Institute of Genetic Engineering & Biotechnology (NIGEB), Tehran, Iran
| |
Collapse
|
2
|
Bekker A, Yang J, Wang J, Cotton MF, Cababasay M, Wiesner L, Moye J, Browning R, Nakwa FL, Rabie H, Violari A, Mirochnick M, Cressey TR, Capparelli EV. Safety and Pharmacokinetics of Lopinavir/Ritonavir Oral Solution in Preterm and Term Infants Starting Before 3 Months of Age. Pediatr Infect Dis J 2024; 43:355-360. [PMID: 38190642 PMCID: PMC10939833 DOI: 10.1097/inf.0000000000004243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Indexed: 01/10/2024]
Abstract
BACKGROUND Study of liquid lopinavir/ritonavir (LPV/r) in young infants has been limited by concerns for its safety in neonates. METHODS International Maternal Pediatric Adolescent AIDS Clinical Trials Network P1106 was a phase IV, prospective, trial evaluating the safety and pharmacokinetics of antiretroviral medications administered according to local guidelines to South African preterm and term infants <3 months of age. Safety evaluation through 24-week follow-up included clinical, cardiac and laboratory assessments. Pharmacokinetic data from P1106 were combined with data from International Maternal Pediatric Adolescent AIDS Clinical Trials Network studies P1030 and P1083 in a population pharmacokinetics model used to simulate LPV exposures with a weight-band dosing regimen in infants through age 6 months. RESULTS Safety and pharmacokinetics results were similar in 13/28 (46%) infants initiating LPV/r <42 weeks postmenstrual age (PMA) and in those starting ≥42 weeks PMA. LPV/r was started at a median (range) age of 47 (13-121) days. No grade 3 or higher adverse events were considered treatment related. Modeling and simulation predicted that for infants with gestational age ≥27 weeks who receive the weight-band dosing regimen, 82.6% will achieve LPV trough concentration above the target trough concentration of 1.0 µg/mL and 56.6% would exceed the observed adult lower limit of LPV exposure of 55.9 µg·h/mL through age 6 months. CONCLUSIONS LPV/r oral solution was safely initiated in a relatively small sample size of infants ≥34 weeks PMA and >2 weeks of life. No serious drug-related safety signal was observed; however, adrenal function assessments were not performed. Weight-band dosing regimen in infants with gestational age ≥27 weeks is predicted to result in LPV exposures equivalent to those observed in other pediatric studies.
Collapse
Affiliation(s)
- Adrie Bekker
- Family Centre for Research with Ubuntu, Department of Pediatrics and Child Health, Stellenbosch University, Cape Town, South Africa
| | - Jincheng Yang
- Department of Pediatrics and Skaggs School of Pharmacy and Pharmaceutical Science, University of California, San Diego, USA
- Clinical Pharmacology and Quantitative pharmacology, CPSS, AstraZeneca R&D, Waltham, MA, USA
| | - Jiajia Wang
- Center for Biostatistics in AIDS Research, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Mark F. Cotton
- Family Centre for Research with Ubuntu, Department of Pediatrics and Child Health, Stellenbosch University, Cape Town, South Africa
| | - Mae Cababasay
- Clinical Pharmacology and Quantitative pharmacology, CPSS, AstraZeneca R&D, Waltham, MA, USA
| | - Lubbe Wiesner
- Division of Clinical Pharmacology, University of Cape Town, Cape Town, South Africa
| | - Jack Moye
- Division of Extramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development
| | - Renee Browning
- Division of AIDS, National Institute of Allergy and Infectious Diseases
| | - Firdose L. Nakwa
- Department of Pediatrics and Child Health, Faculty of Health Sciences, School of Clinical Medicine, University of the Witwatersrand, Johannesburg, South Africa
| | - Helena Rabie
- Family Centre for Research with Ubuntu, Department of Pediatrics and Child Health, Stellenbosch University, Cape Town, South Africa
| | - Avy Violari
- Perinatal HIV research Unit, University of the Witwatersrand, Johannesburg, South Africa
| | - Mark Mirochnick
- Division of Neonatology, Department of Pediatrics, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
| | - Tim. R Cressey
- AMS-PHPT Research Collaboration, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
- Department of Molecular & Clinical Pharmacology, University of Liverpool, UK
| | - Edmund V. Capparelli
- Department of Pediatrics and Skaggs School of Pharmacy and Pharmaceutical Science, University of California, San Diego, USA
| |
Collapse
|
3
|
Chaivichacharn P, Avihingsanon A, Gatechompol S, Ubolyam S, Punyawudho B. Dose optimization with population pharmacokinetics of ritonavir-boosted lopinavir for Thai people living with HIV with and without active tuberculosis. Drug Metab Pharmacokinet 2022; 47:100478. [DOI: 10.1016/j.dmpk.2022.100478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/26/2022] [Revised: 09/11/2022] [Accepted: 10/11/2022] [Indexed: 11/28/2022]
|
4
|
Anesten B, Zetterberg H, Nilsson S, Brew BJ, Fuchs D, Price RW, Gisslén M, Yilmaz A. Effect of antiretroviral treatment on blood-brain barrier integrity in HIV-1 infection. BMC Neurol 2021; 21:494. [PMID: 34937542 PMCID: PMC8693475 DOI: 10.1186/s12883-021-02527-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/25/2021] [Accepted: 12/13/2021] [Indexed: 12/16/2022] Open
Abstract
Background Blood-brain barrier (BBB) injury is prevalent in patients with HIV-associated dementia (HAD) and is a frequent feature of HIV encephalitis. Signs of BBB damage are also sometimes found in neuroasymptomatic HIV-infected individuals without antiretroviral therapy (ART). The aim of this study was to investigate the integrity of the BBB before and after initiation of ART in both neuroasymptomatic HIV infection and in patients with HAD. Methods We determined BBB integrity by measuring cerebrospinal fluid (CSF)/plasma albumin ratios in archived CSF samples prior to and after initiation of ART in longitudinally-followed neuroasymptomatic HIV-1-infected individuals and patients with HAD. We also analyzed HIV RNA in blood and CSF, IgG Index, CSF WBC counts, and CSF concentrations of β2-micoglobulin, neopterin, and neurofilament light chain protein (NfL). Results We included 159 HIV-infected participants; 82 neuroasymptomatic individuals and 77 with HAD. All neuroasymptomatic individuals (82/82), and 10/77 individuals with HAD, were longitudinally followed with a median (interquartile range, IQR) follow-up of 758 (230–1752) days for the neuroasymptomatic individuals, and a median (IQR) follow-up of 241 (50–994) days for the individuals with HAD. Twelve percent (10/82) of the neuroasymptomatic individuals and 80% (8/10) of the longitudinally-followed individuals with HAD had elevated albumin ratios at baseline. At the last follow-up, 9% (7/82) of the neuroasymptomatic individuals and 20% (2/10) of the individuals with HAD had elevated albumin ratios. ART significantly decreased albumin ratios in both neuroasymptomatic individuals and in patients with HAD. Conclusion These findings indicate that ART improves and possibly normalizes BBB integrity in both neuroasymptomatic HIV-infected individuals and in patients with HAD.
Collapse
Affiliation(s)
- Birgitta Anesten
- Department of Infectious Diseases, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, SE-415 50, Gothenburg, Sweden. .,Department of Infectious Diseases, Region Västra Götaland, Sahlgrenska University Hospital, Gothenburg, Sweden.
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden.,Clinical Neurochemistry Laboratory Sahlgrenska University Hospital, Mölndal, Sweden.,Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK.,UK Dementia Research Institute at UCL, London, UK.,Hong Kong Center for Neurodegenerative Disease, Hong Kong, China
| | - Staffan Nilsson
- Mathematical Sciences, Chalmers University of Technology, Gothenburg, Sweden
| | - Bruce J Brew
- Department of Neurology, St.Vincent's Hospital, Sydney, NSW, Australia.,Department of HIV Medicine and Peter Duncan Neurosciences Unit, St Vincent's Centre for Applied Medical Research, St. Vincent's Hospital, Sydney, NSW, Australia
| | - Dietmar Fuchs
- Division of Biological Chemistry, Biocenter, Innsbruck Medical University, Innsbruck, Austria
| | - Richard W Price
- Department of Neurology, University of California San Francisco, San Francisco, California, USA
| | - Magnus Gisslén
- Department of Infectious Diseases, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, SE-415 50, Gothenburg, Sweden.,Department of Infectious Diseases, Region Västra Götaland, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Aylin Yilmaz
- Department of Infectious Diseases, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, SE-415 50, Gothenburg, Sweden.,Department of Infectious Diseases, Region Västra Götaland, Sahlgrenska University Hospital, Gothenburg, Sweden
| |
Collapse
|
5
|
Tan DHS, Chan AK, Jüni P, Tomlinson G, Daneman N, Walmsley S, Muller M, Fowler R, Murthy S, Press N, Cooper C, Lee T, Mazzulli T, McGeer A. Post-exposure prophylaxis against SARS-CoV-2 in close contacts of confirmed COVID-19 cases (CORIPREV): study protocol for a cluster-randomized trial. Trials 2021; 22:224. [PMID: 33752741 PMCID: PMC7982877 DOI: 10.1186/s13063-021-05134-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/14/2020] [Accepted: 02/15/2021] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Post-exposure prophylaxis (PEP) is a well-established strategy for the prevention of infectious diseases, in which recently exposed people take a short course of medication to prevent infection. The primary objective of the COVID-19 Ring-based Prevention Trial with lopinavir/ritonavir (CORIPREV-LR) is to evaluate the efficacy of a 14-day course of oral lopinavir/ritonavir as PEP against COVID-19 among individuals with a high-risk exposure to a confirmed case. METHODS This is an open-label, multicenter, 1:1 cluster-randomized trial of LPV/r 800/200 mg twice daily for 14 days (intervention arm) versus no intervention (control arm), using an adaptive approach to sample size calculation. Participants will be individuals aged > 6 months with a high-risk exposure to a confirmed COVID-19 case within the past 7 days. A combination of remote and in-person study visits at days 1, 7, 14, 35, and 90 includes comprehensive epidemiological, clinical, microbiologic, and serologic sampling. The primary outcome is microbiologically confirmed COVID-19 infection within 14 days after exposure, defined as a positive respiratory tract specimen for SARS-CoV-2 by polymerase chain reaction. Secondary outcomes include safety, symptomatic COVID-19, seropositivity, hospitalization, respiratory failure requiring ventilator support, mortality, psychological impact, and health-related quality of life. Additional analyses will examine the impact of LPV/r on these outcomes in the subset of participants who test positive for SARS-CoV-2 at baseline. To detect a relative risk reduction of 40% with 80% power at α = 0.05, assuming the secondary attack rate in ring members (p0) = 15%, 5 contacts per case and intra-class correlation coefficient (ICC) = 0.05, we require 110 clusters per arm, or 220 clusters overall and approximately 1220 enrollees after accounting for 10% loss-to-follow-up. We will modify the sample size target after 60 clusters, based on preliminary estimates of p0, ICC, and cluster size and consider switching to an alternative drug after interim analyses and as new data emerges. The primary analysis will be a generalized linear mixed model with logit link to estimate the effect of LPV/r on the probability of infection. Participants who test positive at baseline will be excluded from the primary analysis but will be maintained for additional analyses to examine the impact of LPV/r on early treatment. DISCUSSION Harnessing safe, existing drugs such as LPV/r as PEP could provide an important tool for control of the COVID-19 pandemic. Novel aspects of our design include the ring-based prevention approach, and the incorporation of remote strategies for conducting study visits and biospecimen collection. TRIAL REGISTRATION This trial was registered at www.ClinicalTrials.gov ( NCT04321174 ) on March 25, 2020.
Collapse
Affiliation(s)
- Darrell H S Tan
- Division of Infectious Diseases, St. Michael's Hospital, Toronto, Canada.
- MAP Centre for Urban Health Solutions, St. Michael's Hospital, Toronto, Canada.
- Department of Medicine, University of Toronto, Toronto, Canada.
- Division of Infectious Diseases, University Health Network, Toronto, Canada.
- Institute of Health Policy, Management and Evaluation, Dalla Lana School of Public Health, University of Toronto, Toronto, Canada.
| | - Adrienne K Chan
- Department of Medicine, University of Toronto, Toronto, Canada
- Institute of Health Policy, Management and Evaluation, Dalla Lana School of Public Health, University of Toronto, Toronto, Canada
- Division of Infectious Diseases, Sunnybrook Hospital, Toronto, Canada
| | - Peter Jüni
- Department of Medicine, University of Toronto, Toronto, Canada
- Institute of Health Policy, Management and Evaluation, Dalla Lana School of Public Health, University of Toronto, Toronto, Canada
- Applied Health Research Centre, St. Michael's Hospital, Toronto, Canada
| | - George Tomlinson
- Department of Medicine, University Health Network, Toronto, Canada
| | - Nick Daneman
- Department of Medicine, University of Toronto, Toronto, Canada
- Division of Infectious Diseases, Sunnybrook Hospital, Toronto, Canada
| | - Sharon Walmsley
- Department of Medicine, University of Toronto, Toronto, Canada
- Division of Infectious Diseases, University Health Network, Toronto, Canada
- Institute of Health Policy, Management and Evaluation, Dalla Lana School of Public Health, University of Toronto, Toronto, Canada
| | - Matthew Muller
- Division of Infectious Diseases, St. Michael's Hospital, Toronto, Canada
- Department of Medicine, University of Toronto, Toronto, Canada
| | - Rob Fowler
- Department of Medicine, University of Toronto, Toronto, Canada
- Institute of Health Policy, Management and Evaluation, Dalla Lana School of Public Health, University of Toronto, Toronto, Canada
- Department of Medicine, Sunnybrook Hospital, Toronto, Canada
| | - Srinivas Murthy
- Department of Pediatrics, University of British Columbia, Vancouver, Canada
| | - Natasha Press
- Division of Infectious Diseases, St. Paul's Hospital, Vancouver, Canada
| | - Curtis Cooper
- Division of Infectious Diseases, Ottawa Hospital Research Institute, Ottawa, Canada
| | - Todd Lee
- Division of Infectious Diseases, McGill University Health Centre, Montreal, Canada
| | - Tony Mazzulli
- Department of Microbiology, Mount Sinai Hospital/University Health Network, Toronto, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
| | - Allison McGeer
- Department of Medicine, University of Toronto, Toronto, Canada
- Department of Microbiology, Mount Sinai Hospital/University Health Network, Toronto, Canada
| |
Collapse
|
6
|
Mengist HM, Mekonnen D, Mohammed A, Shi R, Jin T. Potency, Safety, and Pharmacokinetic Profiles of Potential Inhibitors Targeting SARS-CoV-2 Main Protease. Front Pharmacol 2021; 11:630500. [PMID: 33597888 PMCID: PMC7883113 DOI: 10.3389/fphar.2020.630500] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/17/2020] [Accepted: 12/30/2020] [Indexed: 12/20/2022] Open
Abstract
Effective, safe, and pharmacokinetically suitable drugs are urgently needed to curb the ongoing COVID-19 pandemic. The main protease or 3C-like protease (Mpro or 3CLpro) of SARS-CoV-2 is considered an important target to formulate potent drugs corresponding to its crucial role in virus replication and maturation in addition to its relatively conserved active site. Promising baseline data on the potency and safety of drugs targeting SARS-CoV-2 Mpro are currently available. However, preclinical and clinical data on the pharmacokinetic profiles of these drugs are very limited. This review discusses the potency, safety, and pharmacokinetic profiles of potential inhibitors of SARS-CoV-2 Mpro and forward directions on the development of future studies focusing on COVID-19 therapeutics.
Collapse
Affiliation(s)
- Hylemariam Mihiretie Mengist
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Hefei National Laboratory for Physical Sciences at Microscale, Division of Life Sciences and Medicine, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, University of Science and Technology of China, Hefei, China
- Department of Medical Laboratory Science, College of Health Sciences, Debre Markos University, Debre Markos, Ethiopia
| | - Daniel Mekonnen
- Hefei National Laboratory for Physical Sciences at Microscale, Division of Life Sciences and Medicine, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, University of Science and Technology of China, Hefei, China
- Department of Medical Laboratory Science, College of Health Science and Medicine, Bahir Dar University, Bahir Dar, Ethiopia
| | - Ahmed Mohammed
- Hefei National Laboratory for Physical Sciences at Microscale, Division of Life Sciences and Medicine, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, University of Science and Technology of China, Hefei, China
| | - Ronghua Shi
- Hefei National Laboratory for Physical Sciences at Microscale, Division of Life Sciences and Medicine, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, University of Science and Technology of China, Hefei, China
| | - Tengchuan Jin
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Hefei National Laboratory for Physical Sciences at Microscale, Division of Life Sciences and Medicine, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, University of Science and Technology of China, Hefei, China
- CAS Center for Excellence in Molecular Cell Science, Chinese Academy of Science, Shanghai, China
| |
Collapse
|
7
|
Targeted delivery of lopinavir to HIV reservoirs in the mesenteric lymphatic system by lipophilic ester prodrug approach. J Control Release 2021; 329:1077-1089. [DOI: 10.1016/j.jconrel.2020.10.036] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/11/2020] [Revised: 10/16/2020] [Accepted: 10/17/2020] [Indexed: 01/03/2023]
|
8
|
Yu J, Yu D, Lane S, McConnachie L, Ho RJY. Controlled Solvent Removal from Antiviral Drugs and Excipients in Solution Enables the Formation of Novel Combination Multi-Drug-Motifs in Pharmaceutical Powders Composed of Lopinavir, Ritonavir and Tenofovir. J Pharm Sci 2020; 109:3480-3489. [PMID: 32791073 PMCID: PMC8986323 DOI: 10.1016/j.xphs.2020.08.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/05/2020] [Revised: 08/05/2020] [Accepted: 08/05/2020] [Indexed: 01/15/2023]
Abstract
Diverging physicochemical properties of HIV drug combinations are challenging to formulate as a single dosage form. We have found that 2-to-4 hydrophilic and hydrophobic HIV drugs in combination can be stabilized with lipid excipients under a controlled solvent removal process to form a novel pharmaceutical powder distinct from typical amorphous material. This discovery has enabled production of a drug combination nanoparticle (DcNP) powder composed of 3 HIV drugs-water-insoluble lopinavir (LogP = 4.7) and ritonavir (LogP = 5.6) and water-soluble tenofovir (LogP = -1.6). DcNP powder, exhibiting repeating units of multi-drug-motifs (referred to as MDM), is made by dissolving all constituents in ethanolic solution, followed by controlled solvent removal. The DcNP powder intersperses chemically diverse drug molecules with lipid excipients to form repeating MDM units. The proposed MDM structure is consistent with data collected with X-ray diffraction, differential calorimetry, and time-of-flight secondary ion mass spectrometry. The successful assembly of chemically diverse drugs in MDM structure is likely due to a novel process of making drug combination powders. The method described here has successfully extended to formulating other clinically prescribed antiviral drug combinations, and thus may serve as a platform technology for developing drug combination nanoparticles for treating a wide range of chronic diseases.
Collapse
Affiliation(s)
- Jesse Yu
- Department of Pharmaceutics, University of Washington, Seattle, WA 98195-7610, USA
| | - Danni Yu
- Department of Pharmaceutics, University of Washington, Seattle, WA 98195-7610, USA
| | - Sarah Lane
- Department of Pharmaceutics, University of Washington, Seattle, WA 98195-7610, USA
| | - Lisa McConnachie
- Department of Pharmaceutics, University of Washington, Seattle, WA 98195-7610, USA
| | - Rodney J Y Ho
- Department of Pharmaceutics, University of Washington, Seattle, WA 98195-7610, USA; Department of Bioengineering, University of Washington, Seattle, WA 98195-7610, USA.
| |
Collapse
|
9
|
Comparative Antiviral Efficacy of Viral Protease Inhibitors against the Novel SARS-CoV-2 In Vitro. Virol Sin 2020; 35:776-784. [PMID: 32910347 PMCID: PMC7481761 DOI: 10.1007/s12250-020-00288-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/04/2020] [Accepted: 08/14/2020] [Indexed: 12/13/2022] Open
Abstract
The recent outbreak of novel coronavirus pneumonia (COVID-19) caused by a new coronavirus has posed a great threat to public health. Identifying safe and effective antivirals is of urgent demand to cure the huge number of patients. Virus-encoded proteases are considered potential drug targets. The human immunodeficiency virus protease inhibitors (lopinavir/ritonavir) has been recommended in the global Solidarity Trial in March launched by World Health Organization. However, there is currently no experimental evidence to support or against its clinical use. We evaluated the antiviral efficacy of lopinavir/ritonavir along with other two viral protease inhibitors in vitro, and discussed the possible inhibitory mechanism in silico. The in vitro to in vivo extrapolation was carried out to assess whether lopinavir/ritonavir could be effective in clinical. Among the four tested compounds, lopinavir showed the best inhibitory effect against the novel coronavirus infection. However, further in vitro to in vivo extrapolation of pharmacokinetics suggested that lopinavir/ritonavir could not reach effective concentration under standard dosing regimen [marketed as Kaletra®, contained lopinavir/ritonavir (200 mg/50 mg) tablets, recommended dosage is 400 mg/10 mg (2 tablets) twice daily]. This research concluded that lopinavir/ritonavir should be stopped for clinical use due to the huge gap between in vitro IC50 and free plasma concentration. Nevertheless, the structure–activity relationship analysis of the four inhibitors provided further information for de novel design of future viral protease inhibitors of SARS-CoV-2.
Collapse
|
10
|
Charbe NB, Zacconi FC, Amnerkar N, Ramesh B, Tambuwala MM, Clementi E. Bio-analytical Assay Methods used in Therapeutic Drug Monitoring of Antiretroviral Drugs-A Review. CURRENT DRUG THERAPY 2019. [DOI: 10.2174/1574885514666181217125550] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/12/2022]
Abstract
Background: Several clinical trials, as well as observational statistics, have exhibited that the advantages of antiretroviral [ARV] treatment for humans with Human Immunodeficiency Virus / Acquired Immune Deficiency Syndrome HIV/AIDS exceed their risks. Therapeutic drug monitoring [TDM] plays a key role in optimization of ARV therapy. Determination of ARV’s in plasma, blood cells, and other biological matrices frequently requires separation techniques capable of high effectiveness, specific selectivity and high sensitivity. High-performance liquid chromatography [HPLC] coupled with ultraviolet [UV], Photodiode array detectors [PDA], Mass spectrophotometer [MS] detectors etc. are the important quantitative techniques used for the estimation of pharmaceuticals in biological samples. </P><P> Objective: This review article is aimed to give an extensive outline of different bio-analytical techniques which have been reported for direct quantitation of ARV’s. This article aimed to establish an efficient role played by the TDM in the optimum therapeutic outcome of the ARV treatment. It also focused on establishing the prominent role played by the separation techniques like HPLC and UPLC along with the detectors like UV and Mass in TDM. </P><P> Methods: TDM is based on the principle that for certain drugs, a close relationship exists between the plasma level of the drug and its clinical effect. TDM is of no value if the relationship does not exist. The analytical methodology employed in TDM should: 1) distinguish similar compounds; 2) be sensitive and precise and 3) is easy to use. </P><P> Results: This review highlights the advancement of the chromatographic techniques beginning from the HPLC-UV to the more advanced technique like UPLC-MS/MS. TDM is essential to ensure adherence, observe viral resistance and to personalize ARV dose regimens. It is observed that the analytical methods like immunoassays and liquid chromatography with detectors like UV, PDA, Florescent, MS, MS/MS and Ultra performance liquid chromatography (UPLC)-MS/MS have immensely contributed to the clinical outcome of the ARV therapy. Assay methods are not only helping physicians in limiting the side effects and drug interactions but also assisting in monitoring patient’s compliance. </P><P> Conclusion: The present review revealed that HPLC has been the most widely used system irrespective of the availability of more sensitive chromatographic technique like UPLC.
Collapse
Affiliation(s)
- Nitin B. Charbe
- Departamento de Quimica Organica, Facultad de Quimica y de Farmacia, Pontificia Universidad Catolica de Chile, Av. Vicuna McKenna 4860, Macul, Santiago 7820436, Chile
| | - Flavia C. Zacconi
- Departamento de Quimica Organica, Facultad de Quimica y de Farmacia, Pontificia Universidad Catolica de Chile, Av. Vicuna McKenna 4860, Macul, Santiago 7820436, Chile
| | - Nikhil Amnerkar
- Adv V. R. Manohar Institute of Diploma in Pharmacy, Wanadongri, Hingna Road, Nagpur, Maharashtra 441110, India
| | - B. Ramesh
- Sri Adichunchunagiri University, Sri Adichunchunagiri College of Pharmacy, BG Nagar, Karnataka 571418, India
| | - Murtaza M. Tambuwala
- School of Pharmacy and Pharmaceutical Science, University of Ulster, Coleraine, County Londonderry, Northern Ireland BT52 1SA, United Kingdom
| | - Emilio Clementi
- Clinical Pharmacology Unit, CNR Institute of Neuroscience, Department of Biomedical and Clinical Sciences, Luigi Sacco University Hospital, Universita di Milano, Milan, Italy
| |
Collapse
|
11
|
Niu WJ, Sun T, Liu L, Liu XQ, Zhang RF, Yin L, Wang JR, Jia XF, Lu HZ, Zhong MK, Jiao Z, Zhang LJ. Population pharmacokinetics and dosing regimen optimisation of lopinavir in Chinese adults infected with HIV. Basic Clin Pharmacol Toxicol 2018; 124:456-465. [PMID: 30346663 DOI: 10.1111/bcpt.13154] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/11/2018] [Accepted: 10/15/2018] [Indexed: 11/28/2022]
Abstract
Lopinavir (LPV) is a protease inhibitor (PI) for the treatment of human immunodeficiency virus (HIV) infections. Current studies on LPV are mainly focused on Caucasians, and none have investigated the population pharmacokinetics (PPK) of LPV in Chinese population. The present study aimed to develop a PPK model for oral LPV in Chinese adults who are HIV-infected. A total of 460 LPV concentrations from 174 Chinese patients who received LPV/ritonavir (LPV/r) 400/100 mg orally every 12 hours (q12h) were analysed using the non-linear mixed-effects modelling approach. Simulations of the LPV concentration profile were performed with different dosing regimens. A one-compartment model with first-order absorption and elimination process described the data. The estimated apparent clearance (CL/F) and volume of distribution (V/F) (% relative standard error [RSE]) for oral LPV were 5.9 L/h (3%) and 117 L (8%), respectively. Body-weight was identified as a covariate on CL/F. In patients who weighed between 45 and 115 kg and received the standard 400/100 mg q12h regimen, the probability of achieving target trough concentration (Ctrough ) of 1 mg/L was >98% for PI-naïve patients and the probability of achieving target Ctrough of 4 mg/L was <80% for PI-pretreated patients. This is the first population pharmacokinetic study to characterise the PK of LPV in Chinese patients with HIV infection. There were no obvious ethnic differences in the PK of LPV between the Chinese population and Caucasian population. The simulations demonstrated that the standard dosing regimen of 400/100 mg q12h (LPV/r tablets) appears to be sufficient for PI-naïve patients but suboptimal for PI-pretreated patients. Therefore, the regimen of 800/200 mg q12h was recommended for PI-pretreated patients. Further investigation of dosage recommendation could be helpful in optimising LPV therapy for HIV infections.
Collapse
Affiliation(s)
- Wan-Jie Niu
- Department of Pharmacy, Huashan, Hospital, Fudan University, Shanghai, China
| | - Tao Sun
- Shanghai Public Health Clinical Center, Shanghai, China
| | - Li Liu
- Shanghai Public Health Clinical Center, Shanghai, China
| | - Xiao-Qian Liu
- Shanghai Public Health Clinical Center, Shanghai, China
| | | | - Lin Yin
- Shanghai Public Health Clinical Center, Shanghai, China
| | | | - Xiao-Fang Jia
- Shanghai Public Health Clinical Center, Shanghai, China
| | - Hong-Zhou Lu
- Shanghai Public Health Clinical Center, Shanghai, China
| | - Ming-Kang Zhong
- Department of Pharmacy, Huashan, Hospital, Fudan University, Shanghai, China
| | - Zheng Jiao
- Department of Pharmacy, Huashan, Hospital, Fudan University, Shanghai, China
| | - Li-Jun Zhang
- Shanghai Public Health Clinical Center, Shanghai, China
| |
Collapse
|
12
|
Madgulkar AR, Bhalekar MR, Kadam AA. Improvement of Oral Bioavailability of Lopinavir Without Co-administration of Ritonavir Using Microspheres of Thiolated Xyloglucan. AAPS PharmSciTech 2018; 19:293-302. [PMID: 28717974 DOI: 10.1208/s12249-017-0834-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/18/2016] [Accepted: 06/11/2017] [Indexed: 11/30/2022] Open
Abstract
Lopinavir is a BCS Class IV drug exhibiting poor bioavailability due to P-gp efflux and limited permeation. The aim of this research was to formulate and characterize microspheres of lopinavir using thiolated xyloglucan (TH-MPs) as carrier to improve its oral bioavailability without co-administration of ritonavir. Thiomeric microspheres were prepared by ionotropic gelation between alginic acid and calcium ions. Interaction studies were performed using Fourier transform infrared spectroscopy (FT-IR). The thiomeric microspheres were characterized for its entrapment efficiency, T80, surface morphology, and mucoadhesion employing in vitro wash off test. The microspheres were optimized by 32 factorial design. The optimized thiomeric microsphere formulation revealed 93.12% entrapment efficiency, time for 80% drug release (T80) of 358.1 min, and 88% mucoadhesion after 1 h. The permeation of lopinavir from microspheres was enhanced 3.15 times as determined by ex vivo study using everted chick intestine and increased relative bioavailability over 3.22-fold over combination of lopinavir and ritonavir as determined by in vivo study in rat model.
Collapse
|
13
|
Moyle G. A review of the aetiology of dyslipidaemia and hyperlipidaemia in patients with HIV. Int J STD AIDS 2016; 16 Suppl 1:14-20; discussion 20-2, 41-3. [PMID: 16238827 DOI: 10.1258/095646205774462797] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/18/2022]
Affiliation(s)
- Graeme Moyle
- St Stephen's Centre, Chelsea & Westminster Hospital, London, UK
| |
Collapse
|
14
|
No Need for Lopinavir Dose Adjustment during Pregnancy: a Population Pharmacokinetic and Exposure-Response Analysis in Pregnant and Nonpregnant HIV-Infected Subjects. Antimicrob Agents Chemother 2015; 60:400-8. [PMID: 26525798 DOI: 10.1128/aac.01197-15] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/21/2015] [Accepted: 10/24/2015] [Indexed: 11/20/2022] Open
Abstract
Lopinavir-ritonavir is frequently prescribed to HIV-1-infected women during pregnancy. Decreased lopinavir exposure has been reported during pregnancy, but the clinical significance of this reduction is uncertain. This analysis aimed to evaluate the need for lopinavir dose adjustment during pregnancy. We conducted a population pharmacokinetic analysis of lopinavir and ritonavir concentrations collected from 84 pregnant and 595 nonpregnant treatment-naive and -experienced HIV-1-infected subjects enrolled in six clinical studies. Lopinavir-ritonavir doses in the studies ranged between 400/100 and 600/150 mg twice daily. In addition, linear mixed-effect analysis was used to compare the area under the concentration-time curve from 0 to 12 h (AUC0-12) and concentration prior to dosing (Cpredose) in pregnant women and nonpregnant subjects. The relationship between lopinavir exposure and virologic suppression in pregnant women and nonpregnant subjects was evaluated. Population pharmacokinetic analysis estimated 17% higher lopinavir clearance in pregnant women than in nonpregnant subjects. Lopinavir clearance values postpartum were 26.4% and 37.1% lower than in nonpregnant subjects and pregnant women, respectively. As the tablet formulation was estimated to be 20% more bioavailable than the capsule formulation, no statistically significant differences between lopinavir exposure in pregnant women receiving the tablet formulation and nonpregnant subjects receiving the capsule formulation were identified. In the range of lopinavir AUC0-12 or Cpredose values observed in the third trimester, there was no correlation between lopinavir exposure and viral load or proportion of subjects with virologic suppression. Similar efficacy was observed between pregnant women and nonpregnant subjects receiving lopinavir-ritonavir at 400/100 mg twice daily. The pharmacokinetic and pharmacodynamic results support the use of a lopinavir-ritonavir 400/100-mg twice-daily dose during pregnancy.
Collapse
|
15
|
Rougemont M, Nchotu Ngang P, Stoll B, Delhumeau C, Hill A, Ciaffi L, Bonnet F, Menga G, Fampou JC, Calmy A. Safety of zidovudine dose reduction in treatment-naïve HIV infected patients. A randomized controlled study (MiniZID). HIV Med 2015; 17:206-15. [PMID: 26354678 DOI: 10.1111/hiv.12303] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Accepted: 07/01/2015] [Indexed: 11/28/2022]
Abstract
BACKGROUND Since September 2014, zidovudine (ZDV)-based therapy for HIV has been the preferred second-line WHO regimen in Cameroon, but its use is limited by the risk of anaemia at standard dosage. We assessed the safety of a reduced vs. standard dose of ZDV to decrease the risk of anaemia in treatment-naïve, HIV-infected individuals. METHODS In a prospective, randomized, open-label trial in an HIV clinic in Cameroon, 142 eligible adults (CD4 count < 350 cells/μL) were randomized to receive 24 weeks of a regimen comprising lamivudine plus nevirapine with either a reduced (400 mg) or standard dose (600 mg) of ZDV. The primary endpoint was the proportion of participants with new/worsening anaemia. RESULTS Median age was 35 years; 58.5% were women; median body mass index was 23.2 kg/m(2) . At baseline, median haemoglobin was 11.6 g/dL, median CD4 cell count was 163 cells/μL, and median plasma HIV-1 RNA load was 5.4 log10 copies/mL. The proportion of participants with new/worsening anaemia was 37.5% (400 mg ZDV) and 32.9% (600 mg ZDV) (P = 0.563). Ten patients with severe anaemia required a switch from ZDV to tenofovir (11.4% in standard-dose arm vs. 2.8% in low-dose arm; P = 0.054). At 24 weeks, there was no significant difference between treatment groups, including median CD4 T-cell count increases. CONCLUSIONS No significant difference was observed in the overall rate of anaemia between HIV-infected individuals starting a ZDV-based treatment according to a standard- or reduced-dose regimen. Severe anaemia and treatment switches related to study drug, however, were more frequent with 600 mg than 400 mg ZDV.
Collapse
Affiliation(s)
- M Rougemont
- HIV Unit, Division of Infectious Diseases, Department of Internal Medical Specialties, Geneva University Hospitals, Geneva, Switzerland
| | - P Nchotu Ngang
- National Social Insurance Hospital, Approved Treatment Center, Yaounde, Cameroon
| | - B Stoll
- Institute of Global Health, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - C Delhumeau
- HIV Unit, Division of Infectious Diseases, Department of Internal Medical Specialties, Geneva University Hospitals, Geneva, Switzerland
| | - A Hill
- Pharmacology Research Laboratories, University of Liverpool, Liverpool, UK
| | - L Ciaffi
- HIV Unit, Division of Infectious Diseases, Department of Internal Medical Specialties, Geneva University Hospitals, Geneva, Switzerland
| | - F Bonnet
- CHU de Bordeaux, Service de Médecine Interne et Maladies Infectieuses & INSERM U897, Université de Bordeaux, Bordeaux, France
| | - G Menga
- National Social Insurance Hospital, Approved Treatment Center, Yaounde, Cameroon
| | - J-C Fampou
- National Social Insurance Hospital, Approved Treatment Center, Yaounde, Cameroon
| | - A Calmy
- HIV Unit, Division of Infectious Diseases, Department of Internal Medical Specialties, Geneva University Hospitals, Geneva, Switzerland
| |
Collapse
|
16
|
The Glucose Transporter PfHT1 Is an Antimalarial Target of the HIV Protease Inhibitor Lopinavir. Antimicrob Agents Chemother 2015; 59:6203-9. [PMID: 26248369 DOI: 10.1128/aac.00899-15] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/15/2015] [Accepted: 07/17/2015] [Indexed: 01/08/2023] Open
Abstract
Malaria and HIV infection are coendemic in a large portion of the world and remain a major cause of morbidity and mortality. Growing resistance of Plasmodium species to existing therapies has increased the need for new therapeutic approaches. The Plasmodium glucose transporter PfHT is known to be essential for parasite growth and survival. We have previously shown that HIV protease inhibitors (PIs) act as antagonists of mammalian glucose transporters. While the PI lopinavir is known to have antimalarial activity, the mechanism of action is unknown. We report here that lopinavir blocks glucose uptake into isolated malaria parasites at therapeutically relevant drug levels. Malaria parasites depend on a constant supply of glucose as their primary source of energy, and decreasing the available concentration of glucose leads to parasite death. We identified the malarial glucose transporter PfHT as a target for inhibition by lopinavir that leads to parasite death. This discovery provides a mechanistic basis for the antimalarial effect of lopinavir and provides a direct target for novel drug design with utility beyond the HIV-infected population.
Collapse
|
17
|
Zhu L, Hruska M, Hwang C, Shah V, Furlong M, Hanna GJ, Bertz R, Landry IS. Pharmacokinetic interactions between BMS-626529, the active moiety of the HIV-1 attachment inhibitor prodrug BMS-663068, and ritonavir or ritonavir-boosted atazanavir in healthy subjects. Antimicrob Agents Chemother 2015; 59:3816-22. [PMID: 25870057 PMCID: PMC4468697 DOI: 10.1128/aac.04914-14] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/24/2014] [Accepted: 04/05/2015] [Indexed: 11/20/2022] Open
Abstract
BMS-663068 is a prodrug of BMS-626529, a first-in-class attachment inhibitor that binds directly to HIV-1 gp120, preventing initial viral attachment and entry into host CD4(+) T cells. This open-label, multiple-dose, four-sequence, crossover study addressed potential two-way drug-drug interactions following coadministration of BMS-663068 (BMS-626529 is a CYP3A4 substrate), atazanavir (ATV), and ritonavir (RTV) (ATV and RTV are CYP3A4 inhibitors). Thirty-six healthy subjects were randomized 1:1:1:1 to receive one of four treatment sequences with three consecutive treatments: BMS-663068 at 600 mg twice daily (BID), BMS-663068 at 600 mg BID plus RTV at 100 mg once daily (QD), ATV at 300 mg QD plus RTV at 100 mg QD (RTV-boosted ATV [ATV/r]), or BMS-663068 at 600 mg BID plus ATV at 300 mg QD plus RTV at 100 mg QD. Compared with the results obtained by administration of BMS-663068 alone, coadministration of BMS-663068 with ATV/r increased the BMS-626529 maximum concentration in plasma (Cmax) and the area under the concentration-time curve in one dosing interval (AUCtau) by 68% and 54%, respectively. Similarly, coadministration of BMS-663068 with RTV increased the BMS-626529 Cmax and AUCtau by 53% and 45%, respectively. Compared with the results obtained by administration of ATV/r alone, ATV and RTV systemic exposures remained similar following coadministration of BMS-663068 with ATV/r. BMS-663068 was generally well tolerated, and there were no adverse events (AEs) leading to discontinuation, serious AEs, or deaths. Moderate increases in BMS-626529 systemic exposure were observed following coadministration of BMS-663068 with ATV/r or RTV. However, the addition of ATV to BMS-663068 plus RTV did not further increase BMS-626529 systemic exposure. ATV and RTV exposures remained similar following coadministration of BMS-663068 with either ATV/r or RTV. BMS-663068 was generally well tolerated alone or in combination with either RTV or ATV/r.
Collapse
Affiliation(s)
- Li Zhu
- Bristol-Myers Squibb, Research and Development, Princeton, New Jersey, USA
| | - Matthew Hruska
- Bristol-Myers Squibb, Research and Development, Princeton, New Jersey, USA
| | - Carey Hwang
- Bristol-Myers Squibb, Research and Development, Princeton, New Jersey, USA
| | - Vaishali Shah
- Bristol-Myers Squibb, Research and Development, Princeton, New Jersey, USA
| | - Michael Furlong
- Bristol-Myers Squibb, Research and Development, Princeton, New Jersey, USA
| | - George J Hanna
- Bristol-Myers Squibb, Research and Development, Princeton, New Jersey, USA
| | - Richard Bertz
- Bristol-Myers Squibb, Research and Development, Princeton, New Jersey, USA
| | | |
Collapse
|
18
|
Dual therapy with lopinavir and ritonavir plus lamivudine versus triple therapy with lopinavir and ritonavir plus two nucleoside reverse transcriptase inhibitors in antiretroviral-therapy-naive adults with HIV-1 infection: 48 week results of the randomised, open label, non-inferiority GARDEL trial. THE LANCET. INFECTIOUS DISEASES 2014; 14:572-80. [DOI: 10.1016/s1473-3099(14)70736-4] [Citation(s) in RCA: 128] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Academic Contribution Register] [Indexed: 11/15/2022]
|
19
|
von Bibra M, Rosenkranz B, Pretorius E, Rabie H, Edson C, Lenker U, Cotton M, Klinker H. Are lopinavir and efavirenz serum concentrations in HIV-infected children in the therapeutic range in clinical practice? Paediatr Int Child Health 2014; 34:138-41. [PMID: 24225343 DOI: 10.1179/2046905513y.0000000090] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Indexed: 01/07/2023]
Abstract
BACKGROUND In antiretroviral treatment the role of therapeutic drug monitoring via measurement of serum levels remains unclear, especially in children. AIM To quantify exposure to LPV and EFV in children receiving therapy in a routine clinical setting in order to identify risk factors associated with inadequate drug exposure. METHOD A prospective study was conducted in a routine clinical setting in Tygerberg Children's Hospital, South Africa. A total of 53 random serum levels were analyzed. Serum concentrations were determined by an established high-performance liquid chromatography method. RESULTS Of 53 HIV-infected children treated with lopinavir (n = 29, median age 1·83 y) or efavirenz (n = 24, median age 9·3 years), 12 showed serum levels outside the therapeutic range (efavirenz) or below Cmin (lopinavir). Low bodyweight, rifampicin co-treatment, and significant comorbidity were potential risk factors for inadequate drug exposure. CONCLUSION These findings, together with previous studies, indicate that therapeutic drug monitoring can improve the management of antiretroviral therapy in children at risk.
Collapse
|
20
|
Fisher M, Norton M, Saget B, Fredrick L, Wyk JV. Safety and efficacy of lopinavir/ritonavir-containing antiretroviral therapy in patients aged <50 versus ≥50 years from randomized trials. Future Virol 2014. [DOI: 10.2217/fvl.14.18] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/21/2022]
Abstract
ABSTRACT: Aim: Data describing the safety and efficacy of antiretroviral therapy (ART) in HIV-infected patients aged ≥50 years are limited. We evaluated the effect of age on safety, efficacy and tolerability in patients aged <50 and ≥50 years receiving lopinavir/ritonavir (LPV/r)-containing ART. Methods: End points from AbbVie Inc. (IL, USA) or AIDS Clinical Trials Group randomized clinical trials in adults using LPV/r 800/200 mg/day as part of a three-drug regimen (follow-up ≥48 weeks) were evaluated using a random-effects meta-analysis (virologic efficacy; intent-to-treat; noncompleter = failure) or pooled data (other end points). Results: A total of 2608 patients (2294.3 patient-years of follow-up) from ten trials were included: 2248 patients (86.2%) <50 years of age and 360 (13.8%) ≥50 years of age. Demographics and baseline characteristics were similar between age groups. At week 48, 64.9 and 67.8% of patients <50 and ≥50 years, respectively, had plasma HIV-1 RNA <50 copies/ml (random effects meta-analysis p = 0.992). Mean change from baseline in CD4+ T-cell count was +193.9 and +163.5 cells/μl (<50 and ≥50 years, respectively; p < 0.001). Smaller proportions of patients <50 years of age discontinued due to adverse events (AEs)/HIV-related events (4.9 vs 9.4%; p = 0.001) and reported moderate-to-severe treatment-related AEs (30.5 vs 36.4%; p = 0.027) compared with patients ≥50 years of age. Conclusion: This analysis suggests LPV/r-anchored three-drug therapy in patients ≥50 years of age leads to comparable rates of virologic suppression, with a smaller increase in absolute CD4+ T cells and increased AEs, including discontinuations associated with AEs compared with patients <50 years of age.
Collapse
Affiliation(s)
- Martin Fisher
- Brighton & Sussex Medical School & University Hospitals, Eastern Road, Brighton, BN2 5BE, UK
| | | | - Brad Saget
- AbbVie Inc., North Chicago, IL 60064, USA
| | | | | |
Collapse
|
21
|
Randomized clinical trial comparing the pharmacokinetics of standard- and increased-dosage lopinavir-ritonavir coformulation tablets in HIV-positive pregnant women. Antimicrob Agents Chemother 2014; 58:2884-93. [PMID: 24614377 DOI: 10.1128/aac.02599-13] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/20/2022] Open
Abstract
A lopinavir-ritonavir (LPV/r)-based regimen is recommended during pregnancy to reduce the risk of HIV mother-to-child transmission, but the appropriate dose is controversial. We compared the pharmacokinetics of standard and increased LPV/r doses during pregnancy. This randomized, open-label prospective study enrolled 60 pregnant women between gestational weeks 14 and 30. The participants received either the standard dose (400/100 mg twice a day [BID]) or increased dose (600/150 mg BID) of LPV/r tablets during pregnancy and the standard dose for 6 weeks after childbirth. Pharmacokinetics analysis was performed using a high-performance liquid chromatography-tandem mass spectrometry method. Adherent participants who received the standard dose presented minimum LPV concentrations of 4.4, 4.3, and 6.1 μg/ml in the second and third trimesters and postpartum, respectively. The increased-dose group exhibited values of 7.9, 6.9, and 9.2 μg/ml at the same three time points. Although LPV exposure was significantly higher in the increased-dose group, the standard dose produced therapeutic levels of LPV against wild-type virus in all adherent participants, except one patient in the third trimester; 50%, 37.5%, and 25%, and 0%, 15%, and 0% of the participants in the standard- and increased-dose groups failed to achieve therapeutic levels against resistant viruses during the second and third trimesters and after childbirth, respectively. After 12 weeks of treatment and after childbirth, all adherent participants achieved undetectable HIV viral loads, and their babies (49/54) were uninfected. No serious drug-related adverse events were observed. We conclude that the standard dose is appropriate for use during pregnancy and that an increased dose may be necessary for women harboring resistant HIV. (This study has been registered at ClinicalTrials.gov under registration no. NCT00605098.).
Collapse
|
22
|
Kanters S, Mills E, Thorlund K, Bucher H, Ioannidis J. Antiretroviral therapy for initial human immunodeficiency virus/AIDS treatment: critical appraisal of the evidence from over 100 randomized trials and 400 systematic reviews and meta-analyses. Clin Microbiol Infect 2014; 20:114-22. [DOI: 10.1111/1469-0691.12475] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 02/01/2023]
|
23
|
Spreen W, Min S, Ford SL, Chen S, Lou Y, Bomar M, St Clair M, Piscitelli S, Fujiwara T. Pharmacokinetics, safety, and monotherapy antiviral activity of GSK1265744, an HIV integrase strand transfer inhibitor. HIV CLINICAL TRIALS 2014; 14:192-203. [PMID: 24144896 DOI: 10.1310/hct1405-192] [Citation(s) in RCA: 90] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Academic Contribution Register] [Indexed: 11/11/2022]
Abstract
BACKGROUND GSK1265744 is an HIV integrase strand transfer inhibitor selected for clinical development. OBJECTIVE This first-time-in-human and phase IIa investigation assessed GSK1265744 antiviral activity, pharmacokinetics, safety, and tolerability in healthy and HIV-1-infected subjects. METHODS This double-blind, placebo-controlled study consisted of a dose escalation of single (part A) and multiple (part B) oral doses in 48 healthy subjects and an oral dose (part C) in 11 HIV-1-infected subjects. In part A, 2 cohorts of 9 subjects received either 5 and 25 mg or 10 and 50 mg. In part B, 3 cohorts of 10 subjects received 5, 10, or 25 mg once daily for 14 days. In part C and the phase IIa study, subjects received 5 or 30 mg once daily for 10 days. RESULTS Dose-proportional increases in drug exposure were observed in healthy and HIV-1-infected subjects. In healthy subjects, pharmacokinetic variability was low following single or repeat dosing (coefficient of variation, 13%-34% and 15%-23%, respectively). Mean plasma half-life was 31.5 hours. GSK1265744 monotherapy significantly reduced plasma HIV-1 RNA from baseline to day 11 in HIV-1-infected subjects receiving 5 or 30 mg versus placebo (P < .001); mean decrease was 2.2 to 2.3 log10 copies/mL, respectively. Study drug was generally well tolerated with no clinically relevant trends in laboratory values, vital signs, or electrocardiograms. CONCLUSIONS GSK1265744 was well tolerated in healthy and HIV-1-infected subjects. Results demonstrate once-daily doses of 5 or 30 mg exceeded minimum target therapeutic concentrations and produced a significant reduction in plasma HIV-1 RNA viral load.
Collapse
Affiliation(s)
- W Spreen
- GlaxoSmithKline, Research Triangle Park, North Carolina, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Prevalence of calcified carotid artery atheromas in panoramic radiographs of HIV-positive patients undergoing antiretroviral treatment: a retrospective study. Oral Surg Oral Med Oral Pathol Oral Radiol 2014; 117:67-74. [DOI: 10.1016/j.oooo.2013.08.022] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/05/2013] [Revised: 08/23/2013] [Accepted: 08/28/2013] [Indexed: 12/11/2022]
|
25
|
Siwak E, Kubicka J, Kowalska JD, Bąkowska E, Święcki P, Pulik P, Pietraszkiewicz E, Firląg-Burkacka E, Horban A. Safety and effectiveness of lopinavir/ritonavir twice-daily in cART naïve patients with HIV-1 infection: Data from POLCA Study Group. HIV & AIDS REVIEW 2014. [DOI: 10.1016/j.hivar.2014.06.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/16/2022] Open
|
26
|
Patterson KB, Dumond JB, Prince HA, Jenkins AJ, Scarsi KK, Wang R, Malone S, Hudgens MG, Kashuba ADM. Protein binding of lopinavir and ritonavir during 4 phases of pregnancy: implications for treatment guidelines. J Acquir Immune Defic Syndr 2013; 63:51-8. [PMID: 23221983 PMCID: PMC3625477 DOI: 10.1097/qai.0b013e31827fd47e] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 01/30/2023]
Abstract
OBJECTIVE To investigate the intraindividual pharmacokinetics (PKs) of total (protein bound plus unbound) and unbound lopinavir/ritonavir (LPV/RTV) and to assess whether the pediatric formulation (100 mg/25 mg) can overcome any pregnancy-associated changes. DESIGN Prospective longitudinal PK study. METHODS HIV-infected pregnant antiretroviral therapy-naive and experienced women receiving LPV/RTV 400 mg/100 mg tablets twice daily. Intensive PK evaluations were performed at 20-24 weeks (PK1), 30 weeks (PK2) followed by empiric dose increase using the pediatric formulation (100 mg/25 mg twice daily), 32 weeks (PK3), and 8 weeks postpartum (PK4). RESULTS Twelve women completed prespecified PK evaluations. Median (range) age was 28 (18-35) years and baseline BMI was 32 (19-41) kg/m. During pregnancy, total area under the time concentration (AUC0-12h) for LPV was significantly lower than postpartum (PK1, PK2, or PK3 vs. PK4, P = 0.005). Protein-unbound LPV AUC0-12h remained unchanged during pregnancy [PK1: 1.6 (1.3-1.9) vs. PK2: 1.6 (1.3-1.9) μg·h/mL, P = 0.4] despite a 25% dose increase [PK2 vs. PK3: 1.8 (1.3-2.1) μg·h/mL, P = 0.5]. Protein-unbound LPV predose concentrations (C12h) did not significantly change despite dose increase [PK2: 0.10 (0.08-0.15) vs. PK3: 0.12 (0.10-0.15) μg/mL, P = 0.09]. Albumin and LPV AUC0-12h fraction unbound were correlated (rs = 0.3, P = 0.03). CONCLUSIONS Total LPV exposure was significantly decreased throughout pregnancy despite the increased dose. However, the exposure of unbound LPV did not change significantly regardless of trimester or dose. Predose concentrations of unbound LPV were not affected by the additional dose and were 70-fold greater than the minimum efficacy concentration. These findings suggest dose adjustments may not be necessary in all HIV-infected pregnant women.
Collapse
Affiliation(s)
- Kristine B Patterson
- Department of Medicine, University of North Carolina, Chapel Hill, NC 27599-7215, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Katayama M, Jiamsripong P, Bukatina AE, Lombari TR, Mcmahon EM, Gades NM, Belohlavek M. Optimized administration regimen of lopinavir for a myocardial ischaemia reperfusion study in Sprague–Dawley rats. Lab Anim 2013; 47:122-6. [DOI: 10.1177/0023677213476864] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/16/2022]
Abstract
Pharmacokinetics of drugs may differ between small and large mammals (including humans); therefore, drug testing in animal models must be carefully designed. Sprague–Dawley rats were used in cardiac experiments, during which the lopinavir concentration in serum had to match human therapeutic levels (4–10 μg/mL). Lopinavir was administered as a co-formulated drug of lopinavir and ritonavir. It was found that after a single administration of a standard human peroral dose (lopinavir 13.3 mg/kg of body weight), the serum concentration of lopinavir was only one-tenth of the target level. It remained below the minimum target level even after 10-fold the standard dose was administered. After initial pilot tests, a dose escalation study was conducted with oral doses 10- and 15-fold the standard clinical dose of lopinavir (i.e. 133 and 200 mg/kg, respectively). A second administration 2 h later effectively increased and maintained higher concentrations during the experimental ischaemia and reperfusion periods. A dose-dependent increase in serum concentration of the drug was observed. Thus, the target therapeutic serum level of lopinavir in the rats was achieved by administrating 10- to 15-fold the standard human dose twice, separated by a 2 h interval.
Collapse
Affiliation(s)
- M Katayama
- Division of Cardiovascular Diseases, Mayo Clinic, 13400 E Shea Blvd, Scottsdale, AZ 85259, USA
| | - P Jiamsripong
- Department of Medicine, University of Hawaii, John A. Burns School of Medicine, Honolulu, HI, USA
| | - A E Bukatina
- Division of Cardiovascular Diseases, Mayo Clinic, 13400 E Shea Blvd, Scottsdale, AZ 85259, USA
| | - T R Lombari
- Department of Animal Care and Technologies, Arizona State University, Tempe, AZ, USA
| | - E M Mcmahon
- Division of Cardiovascular Diseases, Mayo Clinic, 13400 E Shea Blvd, Scottsdale, AZ 85259, USA
| | - N M Gades
- Department of Comparative Medicine, Mayo Clinic, Scottsdale, AZ, USA
| | - M Belohlavek
- Division of Cardiovascular Diseases, Mayo Clinic, 13400 E Shea Blvd, Scottsdale, AZ 85259, USA
| |
Collapse
|
28
|
Schwarcz L, Chen MJ, Vittinghoff E, Hsu L, Schwarcz S. Declining incidence of AIDS-defining opportunistic illnesses: results from 16 years of population-based AIDS surveillance. AIDS 2013; 27:597-605. [PMID: 23079812 DOI: 10.1097/qad.0b013e32835b0fa2] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/25/2022]
Abstract
OBJECTIVE To measure the incidence and risk factors of AIDS-defining opportunistic illnesses (AOIs) in the pre-highly active antiretroviral therapy (HAART) (1993-1995), early-HAART (1996-2000), and late-HAART (2001-2008) periods. DESIGN Prospective cohort analysis of AIDS surveillance data. METHODS Individuals living with, or diagnosed with AIDS from 1993 through 2008 were included. Poisson regression models were used to estimate annual incidence rates of the eight most frequently occurring AOIs, and to compare these rates in the pre-HAART (1993-1995), early-HAART (1996-2000), and late-HAART (2001-2008) periods. RESULTS There were 18 733 individuals with AIDS included; 5788 were diagnosed prior to 1993 and 12 945 were diagnosed between 1 January 1993 and 31 December 2008. The incidence rates of Pneumocystis jiroveci pneumonia, wasting syndrome, Kaposi's sarcoma, HIV encephalopathy, cytomegalovirus retinitis, cytomegalovirus, and esophageal candidiasis decreased during the study period, with the largest declines observed between the pre-HAART and early-HAART periods. Incidence rates also decreased between the early-HAART and late-HAART periods, though not as sharply. Incidence rate reductions between the earliest and latest period ranged from 84 to 99%. CONCLUSIONS Steep declines in incidence of AOIs were found following the introduction of HAART and continued into the late-HAART era. These declines reflect the impact of HIV diagnosis and treatment on a population level.
Collapse
|
29
|
Hermes A, Squires K, Fredrick L, Martinez M, Pasley M, Trinh R, Norton M. Meta-analysis of the safety, tolerability, and efficacy of lopinavir/ritonavir-containing antiretroviral therapy in HIV-1-infected women. HIV CLINICAL TRIALS 2013. [PMID: 23195669 DOI: 10.1310/hct1306-308] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Academic Contribution Register] [Indexed: 11/11/2022]
Abstract
PURPOSE Women comprise ≯50% of HIV-infected patients, yet safety, tolerability, and efficacy data in women taking antiretrovirals (ARVs) are limited. Lopinavir/ ritonavir (LPV/r)-anchored regimens are globally the most widely prescribed HIV-1 protease inhibitor regimens. The objective was to investigate the safety and efficacy of LPV/r-based therapy in women. METHODS A database query yielded all available data in HIV-1-infected subjects receiving LPV/r-based triple-ARV regimens from randomized clinical trials lasting ≥48 weeks from Abbott or Abbott-supported AIDS Clinical Trials Group studies. Efficacy (HIV-1 RNA levels, CD4+ T-cell counts) and safety and tolerability (treatment discontinuation, treatment-related adverse events [AE], and clinical laboratory abnormalities) at 48 weeks were assessed for total women, women by age (≥50, <50 years) and body mass index (BMI; <25, ≥25 to <30, ≥30 kg/m2), and sex. RESULTS Nine hundred ninety-two women initiated LPV/r-based therapy (of whom 79.2% were ARV-naïve), with 83.6% completing 48 weeks of treatment. There were 75.5% of women who achieved a threshold of HIV RNA <400 copies/mL by intent-to-treat, non-completer equals failure (ITT, NC = F) analysis, with a mean ± SE CD4+ T-cell count increase of 191.6 ± 4.92 cells/mm3 from baseline. Women aged ≥50 versus <50 years had higher incidence of moderate-to-severe treatment-related AEs and certain laboratory abnormalities, better virologic response (HIV RNA <400 copies/mL by ITT, NC = F), similar immunologic responses, and similar overall incidence of treatment discontinuations. Higher incidences of certain moderate-to-severe treatment-related AEs and laboratory abnormalities occurred in women with BMI ≥30 kg/m2; however, no effect of BMI on efficacy or discontinuation was observed. CONCLUSIONS LPV/r-based regimens were efficacious and well-tolerated in women without marked differences based on age and BMI categories evaluated.
Collapse
Affiliation(s)
- A Hermes
- Abbott Laboratories, Abbott Park, IL, USA.
| | | | | | | | | | | | | |
Collapse
|
30
|
Seminari E, De Silvestri A, Scudeller L, Scotti V, Tinelli C. Differences in implementation of HIV/AIDS clinical research in developed versus developing world: an evidence-based review on protease inhibitor use among women and minorities. Int J STD AIDS 2012; 23:837-42. [DOI: 10.1258/ijsa.2012.012047] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/18/2022]
Abstract
The aim of this revision is to evaluate ethnicity and gender rate of enrolment in registrative clinical trials of the protease inhibitors (Pis) from 1996 to 2009. Company-sponsored, phase II or III registrative clinical trials of PIs were evaluated. Forty-nine clinical trials were included. Clinical trials were conducted in centres in North America ( n = 39), Central-South America ( n = 22), Europe ( n = 22), Africa ( n = 8), Asia ( n = 5), Australia ( n = 10). Overall mean age was 39.6 years; median proportion of women was 16.3%. The most represented ethnic group was Caucasian. A test for trend over time (1996-2009) shows a significant increase in the proportion of women included ( P = 0.012), and a decrease in the proportion of Caucasians included, reaching borderline significance ( P = 0.061). An inverse correlation was observed between the proportion of Caucasians and that of women enrolled in each study ( r = 0.65, P < 0.0001). Women were less likely to be included in double-blind studies (11.2% versus 17%, P = 0.019). Clinical trials for treatment-naive subjects were more likely to enrol ethnicities other than Caucasian compared with Caucasian (44.7% versus 27.1 %, respectively, P = 0.04). Rates of enrolment of minorities in registrative clinical trials for PIs show a positive trend since 1996, mirroring the growing number of people of different ethnic groups accessing ART.
Collapse
Affiliation(s)
| | - A De Silvestri
- Servizio di Epidemiologia Clinica e Biometria, Fondazione IRCCS Policlinico San Matteo, Piazzale Golgi 2 27100 PAVIA, Italy
| | - L Scudeller
- Servizio di Epidemiologia Clinica e Biometria, Fondazione IRCCS Policlinico San Matteo, Piazzale Golgi 2 27100 PAVIA, Italy
| | - V Scotti
- Servizio di Epidemiologia Clinica e Biometria, Fondazione IRCCS Policlinico San Matteo, Piazzale Golgi 2 27100 PAVIA, Italy
| | - C Tinelli
- Servizio di Epidemiologia Clinica e Biometria, Fondazione IRCCS Policlinico San Matteo, Piazzale Golgi 2 27100 PAVIA, Italy
| |
Collapse
|
31
|
Vento S, Lanzafame M, Lattuada E, Cainelli F, Restelli U, Foglia E. Dose reduction of antiretrovirals: a feasible and testable approach to expand HIV treatment in developing countries. Trop Med Int Health 2012; 18:40-4. [PMID: 23094787 DOI: 10.1111/tmi.12008] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/28/2022]
|
32
|
Abstract
The efficacy of an antiretroviral (ARV) treatment regimen depends on the activity of the regimen's individual ARV drugs and the number of HIV-1 mutations required for the development of resistance to each ARV - the genetic barrier to resistance. ARV resistance impairs the response to therapy in patients with transmitted resistance, unsuccessful initial ARV therapy and multiple virological failures. Genotypic resistance testing is used to identify transmitted drug resistance, provide insight into the reasons for virological failure in treated patients, and help guide second-line and salvage therapies. In patients with transmitted drug resistance, the virological response to a regimen selected on the basis of standard genotypic testing approaches the responses observed in patients with wild-type viruses. However, because such patients are at a higher risk of harbouring minority drug-resistant variants, initial ARV therapy in this population should contain a boosted protease inhibitor (PI) - the drug class with the highest genetic barrier to resistance. In patients receiving an initial ARV regimen with a high genetic barrier to resistance, the most common reasons for virological failure are nonadherence and, potentially, pharmacokinetic factors or minority transmitted drug-resistant variants. Among patients in whom first-line ARVs have failed, the patterns of drug-resistance mutations and cross-resistance are often predictable. However, the extent of drug resistance correlates with the duration of uncontrolled virological replication. Second-line therapy should include the continued use of a dual nucleoside/nucleotide reverse transcriptase inhibitor (NRTI)-containing backbone, together with a change in the non-NRTI component, most often to an ARV belonging to a new drug class. The number of available fully active ARVs is often diminished with each successive treatment failure. Therefore, a salvage regimen is likely to be more complicated in that it may require multiple ARVs with partial residual activity and compromised genetic barriers of resistance to attain complete virological suppression. A thorough examination of the patient's ARV history and prior resistance tests should be performed because genotypic and/or phenotypic susceptibility testing is often not sufficient to identify drug-resistant variants that emerged during past therapies and may still pose a threat to a new regimen. Phenotypic testing is also often helpful in this subset of patients. ARVs used for salvage therapy can be placed into the following hierarchy: (i) ARVs belonging to a previously unused drug class; (ii) ARVs belonging to a previously used drug class that maintain significant residual antiviral activity; (iii) NRTI combinations, as these often appear to retain in vivo virological activity, even in the presence of reduced in vitro NRTI susceptibility; and rarely (iv) ARVs associated with previous virological failure and drug resistance that appear to have possibly regained their activity as a result of viral reversion to wild type. Understanding the basic principles of HIV drug resistance is helpful in guiding individual clinical decisions and the development of ARV treatment guidelines.
Collapse
Affiliation(s)
- Michele W Tang
- Stanford University, Division of Infectious Diseases, Stanford, CA 94305-5107, USA.
| | | |
Collapse
|
33
|
Abstract
PURPOSE OF REVIEW This review considers the differential diagnosis, pathophysiology and risk of hepatotoxicity of specific antiretroviral medications. RECENT FINDINGS Currently prescribed antiretroviral medications are associated with an incidence of grade 3/4 liver enzyme elevation of less than 5%. Clinically apparent hepatotoxicity rates are much lower. The risk of adverse events with combination HIV and hepatitis C virus treatments is low, assuming that several nucleosides including didanosine and stavudine are avoided. SUMMARY Irrespective of the HIV antiretroviral regimen prescribed, careful observation of liver function and enzymes is advised, especially in those with comorbid liver disease. The majority of patients do not experience treatment-limiting liver toxicities, achieve virological suppression, and realize immunological restoration.
Collapse
|
34
|
Ofotokun I, Sheth AN, Sanford SE, Easley KA, Shenvi N, White K, Eaton ME, Del Rio C, Lennox JL. A switch in therapy to a reverse transcriptase inhibitor sparing combination of lopinavir/ritonavir and raltegravir in virologically suppressed HIV-infected patients: a pilot randomized trial to assess efficacy and safety profile: the KITE study. AIDS Res Hum Retroviruses 2012; 28:1196-206. [PMID: 22364141 DOI: 10.1089/aid.2011.0336] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/13/2022] Open
Abstract
A nucleoside reverse transcriptase inhibitor (NRTI) backbone is a recommended component of standard highly active antiretroviral therapy (sHAART). However, long-term NRTI exposure can be limited by toxicities. NRTI class-sparing alternatives are warranted in select patient populations. This is a 48-week single-center, open-label pilot study in which 60 HIV-infected adults with plasma HIV-1 RNA (<50 copies/ml) on sHAART were randomized (2:1) to lopinavir/ritonavir (LPV/r) 400/100 mg BID+raltegravir (RAL) 400 mg BID switch (LPV-r/RAL arm) or to continue on sHAART. The primary endpoint was the proportion of subjects with HIV-RNA<50 copies/ml at week 48. Secondary efficacy and immunologic and safety endpoints were evaluated. Demographics and baseline lipid profile were similar across arms. Mean entry CD4 T cell count was 493 cells/mm(3). At week 48, 92% [95% confidence interval (CI): 83-100%] of the LPV-r/RAL arm and 88% (95% CI: 75-100%) of the sHAART arm had HIV-RNA<50 copies/ml (p=0.70). Lipid profile (mean ± SEM, mg/dl, LPV-r/RAL vs. sHAART) at week 24 was total-cholesterol 194 ± 5 vs. 176 ± 9 (p=0.07), triglycerides 234 ± 30 vs. 133 ± 27 (p=0.003), and LDL-cholesterol 121 ± 6 vs. 110 ± 8 (p=0.27). There were no serious adverse events (AEs) in either arm. Regimen change occurred in three LPV-r/RAL subjects (n=1, due to LPV-r/RAL-related AEs) vs. 0 in sHAART. There were no differences between arms in bone mineral density, total body fat composition, creatinine clearance, or CD4 T cell counts at week 48. In virologically suppressed patients on HAART, switching therapy to the NRTI-sparing LPV-r/RAL combination produced similar sustained virologic suppression and immunologic profile as sHAART. AEs were comparable between arms, but the LPV-r/RAL arm experienced higher triglyceridemia.
Collapse
Affiliation(s)
- Ighovwerha Ofotokun
- Emory University School of Medicine, Department of Medicine, Division of Infectious Diseases, Atlanta, Georgia
- Emory University Center for AIDS Research, Atlanta, Georgia
| | - Anandi N. Sheth
- Emory University School of Medicine, Department of Medicine, Division of Infectious Diseases, Atlanta, Georgia
| | - Sara E. Sanford
- Emory University School of Medicine, Department of Medicine, Division of Infectious Diseases, Atlanta, Georgia
| | - Kirk A. Easley
- Emory University Center for AIDS Research, Atlanta, Georgia
- Emory University School of Public Health, Biostatistics and Bioinformatics, Atlanta, Georgia
| | - Neeta Shenvi
- Emory University School of Public Health, Biostatistics and Bioinformatics, Atlanta, Georgia
| | | | - Molly E. Eaton
- Emory University School of Medicine, Department of Medicine, Division of Infectious Diseases, Atlanta, Georgia
- Emory University Center for AIDS Research, Atlanta, Georgia
| | - Carlos Del Rio
- Emory University School of Medicine, Department of Medicine, Division of Infectious Diseases, Atlanta, Georgia
- Emory University Center for AIDS Research, Atlanta, Georgia
- Emory University School of Public Health, Global Health, Atlanta, Georgia
| | - Jeffrey L. Lennox
- Emory University School of Medicine, Department of Medicine, Division of Infectious Diseases, Atlanta, Georgia
- Emory University Center for AIDS Research, Atlanta, Georgia
| |
Collapse
|
35
|
Optimising the manufacture, formulation, and dose of antiretroviral drugs for more cost-efficient delivery in resource-limited settings: a consensus statement. THE LANCET. INFECTIOUS DISEASES 2012; 12:550-60. [DOI: 10.1016/s1473-3099(12)70134-2] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Academic Contribution Register] [Indexed: 11/22/2022]
|
36
|
Neither Branded Nor Generic Lopinavir/Ritonavir Produces Adequate Lopinavir Concentrations at a Reduced Dose of 200/50 mg Twice Daily. J Acquir Immune Defic Syndr 2012; 59:55-8. [DOI: 10.1097/qai.0b013e31823ba736] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/26/2022]
|
37
|
Assessment of lopinavir pharmacokinetics with respect to developmental changes in infants and the impact on weight band-based dosing. Clin Pharmacol Ther 2011; 91:243-9. [PMID: 22190064 DOI: 10.1038/clpt.2011.218] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/08/2022]
Abstract
Improved antiretroviral therapies are needed for the treatment of HIV-infected infants, given the rapid progression of the disease and drug resistance resulting from perinatal exposure to antiretrovirals. We examined longitudinal pharmacokinetics (PK) data from a clinical trial of lopinavir/ritonavir (LPV/r) in HIV-infected infants in whom therapy was initiated at less than 6 months of age. A population PK analysis was performed using NONMEM to characterize changes in lopinavir (LPV) PK relating to maturational changes in infants, and to assess dosing requirements in this population. We also investigated the relationship between LPV PK and viral dynamic response. Age and ritonavir concentrations were the only covariates found to be significant. Population PK of LPV was characterized by high apparent clearance (CL/F) in young infants, which decreased with increasing age. Although younger infants had lower LPV concentrations, the viral dynamics did not correlate with initial LPV exposure. Monte Carlo simulations demonstrated that WHO weight band-based dosing recommendations predicted therapeutic LPV concentrations and provided drug exposure levels comparable to those resulting from US Food and Drug Administration (FDA)-suggested dosing regimens.
Collapse
|
38
|
Costiniuk CT, Angel JB. ALVAC-HIV as a prophylactic and therapeutic vaccine: highlights from over a decade of clinical trials. Future Virol 2011. [DOI: 10.2217/fvl.11.118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/21/2022]
Abstract
ALVAC-HIV vaccines (vCP125, vCP205, vCP300, vCP1433, vCP1452 and vCP1521) are preparations of a modified recombinant canarypox virus designed to induce or augment CD8+ immune responses. As the focus of several Phase I–III trials, they have been the most extensively studied live vector-based HIV vaccines. Overall, ALVAC-HIV induces modest CD8+ T-cell responses in approximately 20–50% of recipients. The addition of IL-2, recombinant glycoprotein 120 or 160, Remune or LIPO-6T to ALVAC-HIV does not appear to enhance overall CD8+ T-cell immune responses. The ability of ALVAC-HIV to induce interclade immunity and immunogenicity in newborns with perinatal exposure to HIV has important implications for the control of HIV worldwide. Experience from clinical trials in over 10,000 HIV-infected and noninfected individuals has shown that ALVAC constructs are safe, with reactogenicity profiles similar to those reported for currently licensed vaccines. Despite seemingly modest immunogenicity at the present time, studies to date have set the stage for further exploration of the potential of ALVAC-HIV vaccines. This report highlights findings from clinical trials using ALVAC-HIV, alone and in combination with other agents, as both a prophylactic and a therapeutic vaccine.
Collapse
Affiliation(s)
- Cecilia T Costiniuk
- Division of Infectious Diseases, Ottawa Hospital-General Campus, 501 Smyth Road, Ottawa, ON K1H 8L6, Canada
- Department of Biochemistry, Microbiology & Immunology, University of Ottawa, Ottawa, ON, Canada
| | | |
Collapse
|
39
|
Zhu L, Liao S, Child M, Zhang J, Persson A, Sevinsky H, Eley T, Xu X, Krystal M, Farajallah A, McGrath D, Molina JM, Bertz R. Pharmacokinetics and inhibitory quotient of atazanavir/ritonavir versus lopinavir/ritonavir in HIV-infected, treatment-naive patients who participated in the CASTLE Study. J Antimicrob Chemother 2011; 67:465-8. [DOI: 10.1093/jac/dkr490] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/12/2022] Open
|
40
|
Antiviral activity, safety, and pharmacokinetics/pharmacodynamics of dolutegravir as 10-day monotherapy in HIV-1-infected adults. AIDS 2011; 25:1737-45. [PMID: 21716073 DOI: 10.1097/qad.0b013e32834a1dd9] [Citation(s) in RCA: 178] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE To evaluate the antiviral activity, safety, pharmacokinetics, and pharmacokinetics/pharmacodynamics of dolutegravir (DTG), a next-generation HIV integrase inhibitor (INI), as short-term monotherapy. DESIGN A phase IIa, randomized, double-blind, dose-ranging study. METHODS In this study, INI-naive, HIV-1-infected adults currently off antiretroviral therapy were randomized to receive DTG (2, 10, or 50 mg) or placebo once daily for 10 days in an eight active and two placebo randomization scheme per DTG dose. Placebo patients were pooled for the purpose of analysis. RESULTS Thirty-five patients (n = 9 for DTG 2 and 10 mg, n = 10 for DTG 50 mg, and n = 7 for placebo) were enrolled. Baseline characteristics were similar across dose groups. Significant reductions in plasma HIV-1 RNA from baseline to day 11 were observed for all DTG dose groups compared with placebo (P < 0.001), with a mean decrease of 1.51-2.46 log(10) copies/ml. In addition, a well characterized dose-response relationship was observed for viral load decrease. Most patients (seven of 10, 70%) receiving DTG 50 mg achieved plasma HIV-1 RNA less than 50 copies/ml. The pharmacokinetic variability was low (coefficient of variation, range 25-50%). Plasma HIV-1 RNA reduction was best predicted by Cτ using an E(max) model. The most common adverse events were diarrhea, fatigue, and headache; the majority of adverse events were mild or moderate in severity. CONCLUSION Dolutegravir demonstrated potent antiviral activity, good short-term tolerability, low pharmacokinetic variability, and a predictable pharmacokinetics/pharmacodynamics relationship, which support once-daily dosing without a pharmacokinetic booster in integrase-naive patients in future studies.
Collapse
|
41
|
Lattuada E, Lanzafame M, Vento S. Efficacy of lopinavir-ritonavir reduced dose in HIV-infected patients. AIDS Patient Care STDS 2011; 25:455-6. [PMID: 21718150 DOI: 10.1089/apc.2011.0100] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Emanuela Lattuada
- Infectious Diseases Unit, Department of Pathology, University of Verona, “G. B. Rossi” Hospital, Verona, Italy
| | - Massimiliano Lanzafame
- Infectious Diseases Unit, Department of Pathology, University of Verona, “G. B. Rossi” Hospital, Verona, Italy
| | - Sandro Vento
- Department of Internal Medicine, Faculty of Medicine and Health Sciences, University of Botswana, Gaborone, Botswana
| |
Collapse
|
42
|
Abstract
The advent of combination antiretroviral therapy has led to significant improvement in the care of HIV-infected patients. Originally designed as a protease inhibitor (PI), ritonavir is currently exclusively used as a pharmacokinetic enhancer of other protease inhibitors, predominantly due to ritonavir's potent inhibition of the cytochrome P450 3A4 isoenzyme. Ritonavir-boosting of PIs decrease pill burden and frequency of dosing. Boosted PIs are recommended for first-line therapy in treatment and play a key role in the management of treatment-experienced patients. Potential problems associated with PIs include metabolic abnormalities (e.g. dyslipidemia), increased cardiovascular risk, and drug interactions.
Collapse
Affiliation(s)
- Mark W Hull
- Division of AIDS, Department of Medicine, University of British Columbia, Vancouver, Canada
| | | |
Collapse
|
43
|
Menezes P, Miller WC, Wohl DA, Adimora AA, Leone PA, Miller WC, Eron JJ. Does HAART efficacy translate to effectiveness? Evidence for a trial effect. PLoS One 2011; 6:e21824. [PMID: 21765918 PMCID: PMC3135599 DOI: 10.1371/journal.pone.0021824] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/31/2011] [Accepted: 06/11/2011] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Patients who participate in clinical trials may experience better clinical outcomes than patients who initiate similar therapy within clinical care (trial effect), but no published studies have evaluated a trial effect in HIV clinical trials. METHODS To examine a trial effect we compared virologic suppression (VS) among patients who initiated HAART in a clinical trial versus in routine clinical care. VS was defined as a plasma HIV RNA ≤ 400 copies/ml at six months after HAART initiation and was assessed within strata of early (1996-99) or current (2000-06) HAART periods. Risk ratios (RR) were estimated using binomial models. RESULTS Of 738 persons initiating HAART, 30.6% were women, 61.7% were black, 30% initiated therapy in a clinical trial and 67% (n = 496) had an evaluable six month HIV RNA result. HAART regimens differed between the early and current periods (p < 0.001); unboosted PI regimens (55.6%) were more common in the early and NNRTI regimens (46.4%) were more common in the current period. Overall, 78% (95%CI 74, 82%) of patients achieved VS and trial participants were 16% more likely to achieve VS (unadjusted RR 1.16, 95%CI 1.06, 1.27). Comparing trial to non-trial participants, VS differed by study period. In the early period, trial participants initiating HAART were significantly more likely to achieve VS than non-trial participants (adjusted RR 1.33; 95%CI 1.15, 1.54), but not in the current period (adjusted RR 0.98; 95%CI 0.87, 1.11). CONCLUSIONS A clear clinical trial effect on suppression of HIV replication was observed in the early HAART period but not in the current period.
Collapse
Affiliation(s)
- Prema Menezes
- Division of Infectious Diseases, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
- Department of Epidemiology, School of Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
- * E-mail:
| | - William C. Miller
- Division of Infectious Diseases, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
- Department of Epidemiology, School of Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - David A. Wohl
- Division of Infectious Diseases, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Adaora A. Adimora
- Division of Infectious Diseases, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
- Department of Epidemiology, School of Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Peter A. Leone
- Division of Infectious Diseases, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
- Department of Epidemiology, School of Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - William C. Miller
- Division of Infectious Diseases, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
- Department of Epidemiology, School of Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Joseph J. Eron
- Division of Infectious Diseases, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
- Department of Epidemiology, School of Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| |
Collapse
|
44
|
Abstract
Lopinavir/ritonavir approval for use in antiretroviral treatment 10 years ago was very important for the recognition of boosted protease inhibitor (PI)-based therapy as an attractive option for first-line therapy. Being coformulated with ritonavir and having less toxicity than former PIs it allowed for effective and durable virologic suppression with less impact on quality of life. It soon became the standard of care for salvage therapy in its class. Since then, however, its central role has been challenged by new PIs with a more favorable impact on lipid profile, better gastrointestinal tolerability or that are more active in the setting of multidrug resistance. This article summarizes the main clinical studies with lopinavir and discusses its particular characteristics as well as its possible current role in antiretroviral therapy.
Collapse
Affiliation(s)
| | - Marilia Santini de Oliveira
- Instituto de Pesquisa Clínica Evandro Chagas, Fundação Oswaldo Cruz, Av. Brasil 4365, Manguinhos, Rio de Janeiro, 21040-360, Brazil
| | - Beatriz Grinsztejn
- Instituto de Pesquisa Clínica Evandro Chagas, Fundação Oswaldo Cruz, Av. Brasil 4365, Manguinhos, Rio de Janeiro, 21040-360, Brazil
| |
Collapse
|
45
|
Jackson A, Hill A, Puls R, Else L, Amin J, Back D, Lin E, Khoo S, Emery S, Morley R, Gazzard B, Boffito M. Pharmacokinetics of plasma lopinavir/ritonavir following the administration of 400/100 mg, 200/150 mg and 200/50 mg twice daily in HIV-negative volunteers. J Antimicrob Chemother 2011; 66:635-40. [PMID: 21172791 PMCID: PMC3594886 DOI: 10.1093/jac/dkq468] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/14/2022] Open
Abstract
OBJECTIVES Data suggest that some licensed antiretroviral doses could be reduced. We assessed the safety, tolerability and pharmacokinetics of lopinavir/ritonavir at doses of 400/100, 200/150 and 200/50 mg twice daily in HIV-negative volunteers (http://clinicaltrials.gov/ct2/show/NCT00985543). METHODS Male and female volunteers were administered lopinavir/ritonavir at doses of 400/100 mg (two lopinavir/ritonavir Meltrex 200/50 mg tablets, Regimen 1), 200/150 mg (one Meltrex tablet, one 100 mg ritonavir capsule, Regimen 2) and 200/50 mg (one Meltrex tablet, Regimen 3). Each dose was given twice daily for 7 days sequentially, separated by a 7 day wash-out period. Lopinavir/ritonavir steady-state pharmacokinetics was assessed over 12 h at the end of each phase (days 7, 21 and 35). Pharmacokinetic parameters were compared using the 400/100 mg twice daily dose as reference, by determining geometric mean ratios (GMRs) and 90% confidence intervals. RESULTS Twenty-two subjects (eight females) completed the study. Lopinavir AUC(0-12) (ng h/mL), C(max) (ng/mL) and the minimum concentration (C(trough)) (ng/mL) for the 400/100, 200/150 and 200/50 mg twice daily doses, respectively, were as follows: 99,599, 73,603 and 45,146; 11,965, 8939 and 6404; and 5776, 4293 and 1749. Lopinavir pharmacokinetic parameters were significantly lower for Regimens 2 and 3: GMR (90% CI) AUC(0-12), 0.74 (0.65-0.84) and 0.45 (0.40-0.51); C(max), 0.75 (0.66-0.85) and 0.54 (0.40-0.60); and C(trough), 0.74 (0.62-0.89) and 0.30 (0.25-0.36), respectively. All subjects taking the 400/100 and 200/150 mg twice daily doses, and 19 (86%) subjects taking 200/50 mg twice daily had lopinavir concentrations above the suggested minimum effective concentration of 1000 ng/mL. CONCLUSIONS These pharmacokinetic data show that therapeutic plasma concentrations of lopinavir can be achieved with 200/150 mg of lopinavir/ritonavir twice daily (one Meltrex tablet and one 100 mg ritonavir capsule twice daily).
Collapse
Affiliation(s)
- Akil Jackson
- St Stephen's Centre, Chelsea and Westminster Hospital, London, UK.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Abstract
Lopinavir/ritonavir (Kaletra®) is an orally administered coformulated ritonavir-boosted protease inhibitor (PI) comprising lopinavir and low-dose ritonavir. It is indicated, in combination with other antiretroviral agents, for the treatment of HIV-1 infection in adults, adolescents and children. Lopinavir/ritonavir is available as a tablet, soft-gel capsule and an oral solution for patients with difficulty swallowing. In well designed, randomized clinical trials, lopinavir/ritonavir, in combination with other antiretroviral therapies (ART), provided durable virological suppression and improved immunological outcomes in both ART-naive and -experienced adult patients with virological failure. Furthermore, lopinavir/ritonavir demonstrated a high barrier to the development of resistance in ART-naive patients. More limited data indicate that it is effective in reducing plasma HIV-1 RNA levels in paediatric patients. Lopinavir/ritonavir has served as a well established benchmark comparator for the noninferiority of other ritonavir-boosted PI regimens. Although generally well tolerated, lopinavir/ritonavir is associated with generally manageable adverse gastrointestinal side effects and hypertriglyceridaemia and hypercholesterolaemia, which may require coadministration of lipid-lowering agents to reduce the risk of coronary heart disease. Lopinavir/ritonavir, in combination with other ART agents, is a well established and cost-effective treatment for both ART-naive and -experienced patients with HIV-1 infection and, with successful management of adverse events, continues to have a role as an effective component of ART regimens for the control of HIV-1 infection.
Collapse
|
47
|
Cressey TR, Jourdain G, Rawangban B, Varadisai S, Kongpanichkul R, Sabsanong P, Yuthavisuthi P, Chirayus S, Ngo-Giang-Huong N, Voramongkol N, Pattarakulwanich S, Lallemant M. Pharmacokinetics and virologic response of zidovudine/lopinavir/ritonavir initiated during the third trimester of pregnancy. AIDS 2010; 24:2193-200. [PMID: 20625263 PMCID: PMC3070207 DOI: 10.1097/qad.0b013e32833ce57d] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE To evaluate the pharmacokinetics and HIV viral load response following initiation during the third trimester of pregnancy of zidovudine plus standard-dose lopinavir boosted with ritonavir (LPV/r), twice daily, until delivery for the prevention of mother-to-child transmission of HIV. DESIGN Prospective study nested within a multicenter, three-arm, randomized, phase III prevention of mother-to-child transmission of HIV trial in Thailand (PHPT-5, ClinicalTrials.gov Identifier: NCT00409591). METHODS Women randomized to receive 300 mg zidovudine and 400/100 mg LPV/r twice daily from 28 weeks' gestation, or as soon as possible thereafter, until delivery had intensive steady-state 12-h blood sampling performed. LPV/r pharmacokinetic parameters were calculated using noncompartmental analysis. Rules were defined a priori for a LPV/r dose escalation based on the proportion of women with an LPV area under the concentration-time curve (AUC) below 52 microg h/ml (10th percentile for LPV AUC in nonpregnant adults). HIV-1 RNA response was assessed during the third trimester. RESULTS Thirty-eight women were evaluable; at entry, median (range) gestational age was 29 (28-36) weeks, weight 59.5 (45.0-91.6) kg, CD4 cells count 442 (260-1327) cells/microl and HIV-1 RNA viral load 7818 (<40-402 015) copies/ml. Geometric mean (90% confidence interval) LPV AUC, Cmax and Cmin were 64.6 (59.7-69.8) microg h/ml, 8.1 (7.5-8.7) microg/ml and 2.7 (2.4-3.0) microg/ml, respectively. Thirty-one of 38 (81%) women had an LPV AUC above the AUC target. All women had a HIV-1 viral load less than 400 copies/ml at the time of delivery. CONCLUSION A short course of zidovudine plus standard-dose LPV/r initiated during the third trimester of pregnancy achieved adequate LPV exposure and virologic response.
Collapse
Affiliation(s)
- Tim R Cressey
- Department of Medical Technology, Chiang Mai University, Thailand.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Abstract
OBJECTIVE Reduced lopinavir concentrations have been demonstrated with use of the capsule formulation during the third trimester of pregnancy. This study determined lopinavir exposure with an increased dose of the new tablet formulation during the third trimester. DESIGN International Maternal Pediatric Adolescent AIDS Clinical Trials 1026s is a prospective nonblinded pharmacokinetic study in HIV-infected pregnant women, including a cohort receiving 2 lopinavir/ritonavir tablets (400 mg/100 mg) twice daily during the second trimester, 3 tablets (600 mg/150 mg) twice daily during the third trimester, and 2 tablets (400 mg/100 mg) twice daily post delivery through 2 weeks postpartum. METHODS Steady-state 12-hour pharmacokinetic profiles were performed during pregnancy and at 2 weeks postpartum. Lopinavir and ritonavir were measured by reverse-phase high-performance liquid chromatography (detection limit, 0.09 mcg/mL). RESULTS Thirty-three women were studied. Median lopinavir AUC for the second trimester (n = 11), third trimester (n = 33), and postpartum (n = 27) were 72, 96, and 133 mcg x hr/mL, respectively. Median minimum lopinavir concentrations were 3.4, 4.9, and 6.9 mcg/mL. CONCLUSIONS The higher lopinavir/ritonavir tablet dose (600 mg/150 mg) provided exposure during the third trimester similar to the average AUC (98 mcg x hr x mL(-1) in nonpregnant adults taking 400 mg/100 mg twice daily. The higher dose should be used during the second and third trimesters of pregnancy. Postpartum dosing can be reduced to standard dosing before 2 weeks postpartum.
Collapse
|
49
|
Katritzky AR, Hall CD, El-Gendy BEDM, Draghici B. Tautomerism in drug discovery. J Comput Aided Mol Des 2010; 24:475-84. [PMID: 20490619 DOI: 10.1007/s10822-010-9359-z] [Citation(s) in RCA: 111] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/01/2010] [Accepted: 04/17/2010] [Indexed: 12/26/2022]
|
50
|
Kinetics and determining factors of the virologic response to antiretrovirals during pregnancy. Infect Dis Obstet Gynecol 2010; 2009:621780. [PMID: 20130816 PMCID: PMC2814231 DOI: 10.1155/2009/621780] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/23/2009] [Accepted: 10/15/2009] [Indexed: 12/03/2022] Open
Abstract
HIV-infected pregnant women with undetectable plasma HIV RNA concentrations at delivery pose a minimal risk of vertical transmission. We studied the kinetics and the determinants of the virologic response to antiretroviral therapy in 117 consecutive pregnancies. Patients who initiated therapy during pregnancy had a VL decrease of 2 and 2.5 log10 after 4 and 24 weeks, respectively. Therapeutic drug monitoring (TDM) of the protease inhibitors administered in doses recommended for nonpregnant adults resulted in below-target concentrations in 29%, 35%, and 44% of 1st, 2nd, and 3rd trimester measurements, respectively, but low drug concentrations did not correlate with virologic failure. Demographic characteristics, antiretroviral experience prior to pregnancy, baseline VL, or use of specific antiretrovirals did not affect the virologic response. Adherence to ≥95% of prescribed doses and utilization of psychosocial services were associated with undetectable plasma HIV RNA at delivery. In conclusion, the virologic responses of pregnant and nonpregnant adults share similar charactersitics.
Collapse
|