Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Sara Gibbons is active.

Publication


Featured researches published by Sara Gibbons.


Clinical Pharmacokinectics | 1999

Pharmacokinetics and Potential Interactions Amongst Antiretroviral Agents Used To Treat Patients with HIV Infection

Michael J. Barry; Fiona Mulcahy; Concepta Merry; Sara Gibbons; David Back

There are 3 groups of drugs available for the treatment of patients with HIV disease. These are the nucleoside reverse transcriptase inhibitors (‘nucleoside analogues’) [zidovudine, didanosine, zalcitabine, lamivudine and abacavir]; the non-nucleoside reverse transcriptase inhibitors (nevirapine, delavirdine and efavirenz); and the protease inhibitors (saquinavir, ritonavir, indinavir, nelfinavir and amprenavir).The preferred initial regimen should reduce and maintain plasma HIV RNA below the level of detection. Presently, the regimen of choice consists of 2 nucleoside analogues plus a protease inhibitor with high in vivo efficacy. An alternative combination consists of 2 nucleoside analogues plus a non-nucleoside reverse transcriptase inhibitor. Drug interactions are one of the major problems associated with these multidrug regimens.Changes in plasma concentrations of the nucleoside analogues are unlikely to be of clinical relevance as drug effect is mainly dependent on the rate and extent of intracellular phosphorylation. Combinations of zidovudine plus stavudine, and probably zalcitabine plus lamivudine, should be avoided as competition for phosphorylating enzymes may occur. The antiviral efficacy of some nucleoside analogues, e.g. stavudine, may be compromised by prior treatment with other nucleosides (e.g. zidovudine). However, these data need to be clarified in further studies. It is unlikely that administration of other antiretrovirals will influence the activity of nucleoside analogues.Protease inhibitors are metabolised by hepatic cytochrome P450 (CYP) 3A4. Combination protease inhibitor therapy can result in drug interactions mediated by enzyme inhibition. Ritonavir is the most potent inhibitor, saquinavir the least. The protease inhibitors also interact with the non-nucleoside reverse transcriptase inhibitors. Nevirapine and efavirenz induce drug metabolising enzymes and may reduce plasma concentrations of protease inhibitors. A study in healthy volunteers showed that nelfinavir concentrations are increased by combination with efavirenz. Delavirdine inhibits drug metabolising enzymes and increases the plasma concentration of coadministered protease inhibitors. The nucleoside analogues would not be expected to interact with the protease inhibitors.Apart from the ability of didanosine to reduce the area under the concentration-time curve of delavirdine, there are no reports of clinically significant interactions of other antiretrovirals with the non-nucleoside reverse transcriptase inhibitors.Triple therapy is the current standard of care for patients with HIV disease. However, studies of quadruple therapy are already under way. Drug interactions are likely to remain one of the major considerations when selecting a therapeutic regimen for patients with HIV.


AIDS | 1997

Saquinavir pharmacokinetics alone and in combination with nelfinavir in HIV-infected patients.

Concepta Merry; Michael J. Barry; Fiona Mulcahy; Mairin Ryan; Jane Heavey; John Tjia; Sara Gibbons; Alasdair Breckenridge; David Back

Objective:To investigate the pharmacokinetics of saquinavir (SQV) hard gel when administered alone and in combination with nelfinavir (NLF) to HIV-positive patients. Design:Six patients receiving triple therapy (dual nucleoside plus SQV 600 mg three times daily) were studied. On the first study day blood samples were drawn for assay of SQV. Prior to the second study day, patients received their usual medication plus NLF 750 mg three times daily for 2 days. Methods:Blood samples were obtained at times 0, 1, 2, 4, 6 and 8 h after dosing on study days 1 and 2. Following centrifugation, separated plasma was heated at 58°C for at least 30 min to inactivate HIV and stored at −80°C until analysis using high performance liquid chromatography. Results:The geometric mean Cmax and AUC0–8h on the first study day were 253 ng/ml (range, < 25–1200 ng/ml) and 1106 ng/ml•h (range, < 100–3479 ng/ml•h), respectively, and on the second study day were 1204 ng/ml (range, 379–2755 ng/ml) and 5472 ng/ml•h (range, 1434–12538 ng/ml•h), respectively. The geometric mean ratio for Cmax was 4.75 and for AUC0–8h was 4.94. Conclusions:NLF increases the oral bioavailability of SQV (hard gel) approximately fivefold. For some patients the addition of NLF to SQV will increase the drug levels from subtherapeutic to the therapeutic range. In one of our patients the addition of NLF resulted in SQV levels that were much higher than previous work suggests are necessary for maximum antiviral effect. The variability in SQV concentrations both at baseline and following addition of NLF suggest that dosing may best be adjusted by individual therapeutic drug monitoring.


Clinical Pharmacokinectics | 1997

Protease inhibitors in patients with HIV disease. Clinically important pharmacokinetic considerations

M. Barry; Sara Gibbons; David Back; Fiona Mulcahy

SummarySince its introduction in 1987, zidovudine monotherapy has been the treatment of choice for patients with HIV infection. Unfortunately it has been established that the beneficial effects of zidovudine are not sustained due to the development of resistant viral strains. This has led to the strategy of combination therapy, and in 1995 treatment with zidovudine plus didanosine, or zidovudine plus zalcitabine, was demonstrated to be more effective than zidovudine monotherapy in preventing disease progression and reducing mortality in patients with HIV disease.Recent work demonstrates an even greater antiviral effect from triple therapy with 2 nucleosides, zidovudine plus zalcitabine with the addition of saquinavir, a new protease inhibitor drug. The HIV protease enzyme is responsible for the post-translational processing of gag and gagpol polyprotein precursors, and its inhibition by drugs such as saquinavir, ritonavir, indinavir and VX-478 results in the production of non-infectious virions. As resistance may also develop to the protease inhibitors they may be used in combination, and future strategies may well include quadruple therapy with 2 nucleoside analogues plus 2 protease inhibitors. Administration of protease inhibitors alone or in combination with other drugs does raise a number of important pharmacokinetic issues for patients with HIV disease.Some protease inhibitors (e.g. saquinavir) have kinetic profiles characterised by reduced absorption and a high first pass effect, resulting in poor bioavailability which may be improved by administrating with food. Physiological factors including achlorhydria, malabsorption and hepatic dysfunction may influence the bioavailability of protease inhibitors in HIV disease. Protease inhibitors are very highly bound to plasma proteins (>98%), predominantly to αn1-acid glycoprotein. This may influence their antiviral activity in vitro and may also predispose to plasma protein displacement interactions. Such interactions are usually only of clinical relevance if the metabolism of the displaced drug is also inhibited. This is precisely the situation likely to pertain to the protease inhibitors, as ritonavir may displace other protease inhibitor drugs, such as saquinavir, from plasma proteins and inhibit their metabolism.Protease inhibitors are extensively metabolised by the cytochrome P450 (CYP) enzymes present in the liver and small intestine. In vitro studies suggest that the most influential CYP isoenzyme involved in the metabolism of the protease inhibitors is CYP3A, with the isoforms CYP2C9 and CYP2D6 also contributing. Ritonavir has an elimination half-life (t½β) of 3 hours, indinavir 2 hours and saquinavir between 7 and 12 hours. Renal elimination is not significant, with less than 5% of ritonavir and saquinavir excreted in the unchanged form. As patients with HIV disease are likely to be taking multiple prolonged drug regimens this may lead to drug interactions as a result of enzyme induction or inhibition. Recognised enzyme inducers of CYP3A, which are likely to be prescribed for patients with HIV disease, include rifampicin (rifampin) [treatment of pulmonary tuberculosis], rifabutin (treatment and prophylaxis of Mycobacterium avium complex), phenobarbital (phenobarbitone), phenytoin and carbamazepine (treatment of seizures secondary to cerebral toxoplasmosis or cerebral lymphoma). These drugs may reduce the plasma concentrations of the proteaseinhibitors and reduce their antiviral efficacy.If coadministered drugs are substrates for a common CYP enzyme, the elimination of one or both drugs may be impaired. Drugs which are metabolised by CYP3A and are likely to be used in the treatment of patients with HIV disease include the azole antifungals, macrolide antibiotics and dapsone; therefore, protease inhibitors may interact with these drugs. Although dapsone and cotrimoxazole (trimethoprim/sulfamethoxazole) are used in the treatment and prophylaxis of Pneumocystis carinii pneumonia, the incidence of adverse effects is very high, and has been attributed to the formation of a hydroxylamine metabolite mediated by CYP3A and CYP2C9 isoenzymes. The protease inhibitors, particularly ritonavir, may inhibit hydroxylamine production and reduce the incidence of adverse effects. This potential interaction may be beneficial and should be studied in the clinical setting.Protease inhibitors will be prescribed in combination with nucleoside analogues. It is unlikely that a pharmacokinetic drug interaction will result as the metabolic pathways differ. The effect of protease inhibitors on the intracellular phosphorylation of nucleoside analogues is unknown.


Pharmacogenetics and Genomics | 2010

HIV protease inhibitors are substrates for OATP1A2, OATP1B1 and OATP1B3 and lopinavir plasma concentrations are influenced by SLCO1B1 polymorphisms

Ruben C. Hartkoorn; Wai San Kwan; Victoria Shallcross; Ammara Chaikan; Neill J. Liptrott; Deirdre Egan; J Enrique Salcedo Sora; Chloe E. James; Sara Gibbons; Pat G Bray; David Back; Saye Khoo; Andrew Owen

Objective OATP1B1 and OATP1B3 are major hepatic drug transporters whilst OATP1A2 is mainly located in the brain but is also located in liver and several other organs. These transporters affect the distribution and clearance of many endobiotics and xenobiotics and have been reported to have functional single nucleotide polymorphisms (SNPs). We have assessed the substrate specificities of these transporters for a panel of antiretrovirals and investigated the effects of SNPs within these transporters on the pharmacokinetics of lopinavir. Methods SLCO1A2, SLCO1B1 and SLCO1B3 were cloned, verified and used to generate cRNA for use in the Xenopuslaevis oocyte transport system. Using the oocyte system, antiretrovirals were tested for their substrate specificities. Plasma samples (n=349) from the Liverpool therapeutic drug monitoring registry were genotyped for SNPs in SLCO1A2, SLCO1B1 and SLCO1B3 and associations between SNPs and lopinavir plasma concentrations were analysed. Result Antiretroviral protease inhibitors, but not non-nucleoside reverse transcriptase inhibitors, are substrates for OATP1A2, OATP1B1 and OATP1B3. Furthermore, ritonavir was not an inhibitor of OATP1B1. The 521T>C polymorphism in SLCO1B1 was significantly associated with higher lopinavir plasma concentrations. No associations were observed with functional variants of SLCO1A2 and SLCO1B3. Conclusion These data add to our understanding of the factors that contribute to variability in plasma concentrations of protease inhibitors. Further studies are now required to confirm the association of SLCO1B1 521T>C with lopinavir plasma concentrations and to assess the influence of other polymorphisms in the SLCO family.


Antiviral Therapy | 2010

Prevalence of comedications and effect of potential drug-drug interactions in the Swiss HIV Cohort Study.

Catia Marzolini; Luigia Elzi; Sara Gibbons; Rainer Weber; Christoph A. Fux; Hansjakob Furrer; Jean-Philippe Chave; Matthias Cavassini; Enos Bernasconi; Alexandra Calmy; Pietro Vernazza; Saye Khoo; Bruno Ledergerber; David Back; Manuel Battegay

BACKGROUND Potential drug-drug interactions (PDDIs) might expand with new combination antiretroviral therapies (ART) and polypharmacy related to increasing age and comorbidities. We investigated the prevalence of comedications and PDDIs within a large HIV cohort, and their effect on ART efficacy and tolerability. METHODS All medications were prospectively recorded in 1,497 ART-treated patients and screened for PDDIs using a customized version of the Liverpool drug interactions database. RESULTS Overall, 68% (1,013/1,497) of patients had a comedication and 40% (599/1,497) had > or = 1 PDDI. Among patients with comedication, 2% (21/1,013) had red-flag interactions (contraindicated) and 59% (597/1,013) had orange-flag interactions (potential dose adjustment and/or close monitoring required). The latter involved mainly central nervous system drugs (49%), cardiovascular drugs (34%) and methadone (19%). In the multivariate analysis, factors associated with having a comedication were advanced age, female gender, obesity and HCV infection. Independent risk factors for PDDIs were regimens combining protease inhibitors and non-nucleoside reverse transcriptase inhibitors (odds ratio [OR] 3.06, 95% confidence interval [CI] 1.44-6.48), > or = 2 comedications (OR 1.89, 95% CI 1.32-2.70), current illicit drug use (OR 2.00, 95% CI 1.29-3.10) and patients with HCV infection (OR 1.74, 95% CI 1.19-2.56). Viral response was similar in patients with and without PDDIs (84.5% versus 86.4%; P=0.386). During follow-up, ART was modified in 134 patients with comedication regardless of the presence of PDDIs (P=0.524). CONCLUSIONS PDDIs increase with complex ART and comorbidities. No adverse effect was noted on ART efficacy or tolerability; however, most PDDIs affected comedication but were manageable through dose adjustment or monitoring.


AIDS | 1996

The effect of zidovudine dose on the formation of intracellular phosphorylated metabolites

Michael J. Barry; Saye Khoo; Gareth J. Veal; Patrick G. Hoggard; Sara Gibbons; E Wilkins; Olwen Williams; Alasdair Breckenridge; David Back

Objectives:Zidovudine (ZDV) requires intracellular phosphorylation to ZDV triphosphate (ZDV-TP) prior to the inhibition of HIV replication. The effect of ZDV dose on the formation of intracellular phosphorylated metabolites may help define the optimum daily dose of ZDV, which is still unknown. Design and methods:The plasma and intracellular phosphorylated metabolite concentrations of ZDV were determined over a 12 h period following oral administration of 100 and 300 mg ZDV to 10 HIV-seropositive patients at steady state during two dosing regimens (i.e., 100 mg three times daily and 300 mg twice daily). The intracellular ZDV phosphates, ZDV monophosphate (ZDV-MP), ZDV diphosphate (ZDV-DP) and ZDV-TP were measured in peripheral blood mononuclear cells using a combination of high-performance liquid chromatography and radioimmunoassay. Results:There was a greater than threefold increase in maximum plasma concentration (Cmax) following 300 mg ZDV when compared with 100 mg ZDV (mean ± SD, 2.59 ± 0.52 versus 0.70 ± 0.14 μmol/l). The area under the concentration time curve (AUC0–12 h) was also significantly increased (4.59 ± 0.79 versus 1.42 ± 0.51 μmol/l×h) following 300 mg ZDV dose. For total intracellular ZDV phosphate metabolites the AUC0–12 h was doubled (7.64 ± 3.67 versus 3.71 ± 1.83 pmol/106 cells×h) in patients taking 300 mg ZDV compared with 100 mg. The AUC0–12 h for ZDV-MP was significantly increased at the higher dose (6.47 ± 3.14 versus 2.77 ± 1.70 pmol/106 cells×h), whereas the active moiety ZDV-TP was variable and not significantly different (0.42 ± 0.42 versus 0.61 ± 0.81 pmol/106 cells×h) following 100 and 300 mg ZDV. Conclusions:Administration of 100 mg ZDV orally produces significantly less of the potentially toxic metabolite, ZDV-MP, and comparative, although variable, concentrations of the active metabolite ZDV-TP when compared with 300 mg ZDV orally. This finding supports clinical data indicating the efficacy of low-dose (300 mg daily) ZDV. The measurement of intracellular phosphorylated metabolites advances our understanding of the clinical pharmacology of ZDV.


Clinical Infectious Diseases | 2001

Pharmacokinetic Interactions of Nevirapine and Methadone and Guidelines for Use of Nevirapine to Treat Injection Drug Users

Susan Clarke; Fiona Mulcahy; John Tjia; Helen E. Reynolds; Sara Gibbons; Michael J. Barry; David Back

Administration of nevirapine to HIV-infected injection drug users who also receive methadone results in a significant reduction in methadone exposure after 7-10 days of therapy. Many patients require an increase in methadone dose to counteract this effect.


Therapeutic Drug Monitoring | 2006

An Update on Therapeutic Drug Monitoring for Antiretroviral Drugs

David Back; Sara Gibbons; Saye Khoo

There is an increasing uptake of TDM of antiretroviral drugs, particularly in Europe. There is consensus that current antiretroviral drugs meet most of the criteria of drugs that can be considered as candidates for a TDM strategy. This review examines the pharmacokinetic-pharmacodynamic relationship for protease inibitors and non nucleoside reverse transcriptase inhibitor, give an overview of the published randomised clinical trials and then summarises the scenarios for use of TDM. Finally the development of the inhibitory quotient (IQ) concept is discussed.


Clinical Infectious Diseases | 2008

Association of a single-nucleotide polymorphism in the pregnane X receptor (PXR 63396C-->T) with reduced concentrations of unboosted atazanavir.

Marco Siccardi; Antonio D'Avolio; Lorena Baietto; Sara Gibbons; Mauro Sciandra; Daniela Colucci; Stefano Bonora; Saye Khoo; David Back; Giovanni Di Perri; Andrew Owen

This study investigated pregnane X receptor polymorphisms in relation to unboosted atazanavir plasma concentrations in 2 cohorts of patients. The polymorphism 63396T-->C predicted concentrations below the minimum effective concentration (150 ng/mL) with odds ratios of 18 (P = .008) and 5.13 (P = .02). Prospective studies determining potential clinical usefulness are now warranted.


Clinical Infectious Diseases | 2010

Recognition of Risk for Clinically Significant Drug Interactions among HIV-Infected Patients Receiving Antiretroviral Therapy

John G. Evans-Jones; Lucy E. Cottle; David Back; Sara Gibbons; Nicholas J. Beeching; P. Carey; Saye Khoo

We assessed the risk of clinically significant drug interactions in patients receiving antiretrovirals, and their recognition by physicians. Clinically significant drug interactions were recorded in 27% of 159 patients, with 15% of interactions potentially lowering antiretroviral concentrations. Risk of clinically significant drug interactions was significantly related to receipt of protease inhibitors. Only 36% of clinically significant drug interactions were correctly identified by physicians.

Collaboration


Dive into the Sara Gibbons's collaboration.

Top Co-Authors

Avatar

David Back

University of Liverpool

View shared research outputs
Top Co-Authors

Avatar

Saye Khoo

University of Liverpool

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Andrew Owen

University of Liverpool

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Deenan Pillay

University of Birmingham

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

John Tjia

University of Liverpool

View shared research outputs
Top Co-Authors

Avatar

John S. Lambert

Mater Misericordiae University Hospital

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge