Network


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

Hotspot


Dive into the research topics where Katherine Kay is active.

Publication


Featured researches published by Katherine Kay.


Malaria Journal | 2014

Optimizing the programmatic deployment of the anti-malarials artemether-lumefantrine and dihydroartemisinin-piperaquine using pharmacological modelling

Eva Maria Hodel; Katherine Kay; Daniel Hayes; Dianne J Terlouw; Ian M. Hastings

BackgroundSuccessful programmatic use of anti-malarials faces challenges that are not covered by standard drug development processes. The development of appropriate pragmatic dosing regimens for low-resource settings or community-based use is not formally regulated, even though these may alter factors which can substantially affect individual patient and population level outcome, such as drug exposure, patient adherence and the spread of drug resistance and can affect a drug’s reputation and its eventual therapeutic lifespan.MethodsAn in silico pharmacological model of anti-malarial drug treatment with the pharmacokinetic/pharmacodynamic profiles of artemether-lumefantrine (AM-LF, Coartem®) and dihydroartemisinin-piperaquine (DHA-PPQ, Eurartesim®) was constructed to assess the potential impact of programmatic factors, including regionally optimized, age-based dosing regimens, poor patient adherence, food effects and drug resistance on treatment outcome at population level, and compared both drugs’ susceptibility to these factors.ResultsCompared with DHA-PPQ, therapeutic effectiveness of AM-LF seems more robust to factors affecting drug exposure, such as age- instead of weight-based dosing or poor adherence. The model highlights the sub-optimally low ratio of DHA:PPQ which, in combination with the narrow therapeutic dose range of PPQ compared to DHA that drives the weight or age cut-offs, leaves DHA at a high risk of under-dosing.ConclusionPharmacological modelling of real-life scenarios can provide valuable supportive data and highlight modifiable determinants of therapeutic effectiveness that can help optimize the deployment of anti-malarials in control programmes.


Malaria Journal | 2015

Age-shifting in malaria incidence as a result of induced immunological deficit: a simulation study.

Peter Pemberton-Ross; Thomas Smith; Eva Maria Hodel; Katherine Kay; Melissa A. Penny

Effective population-level interventions against Plasmodium falciparum malaria lead to age-shifts, delayed morbidity or rebounds in morbidity and mortality whenever they are deployed in ways that do not permanently interrupt transmission. When long-term intervention programmes target specific age-groups of human hosts, the age-specific morbidity rates ultimately adjust to new steady-states, but it is very difficult to study these rates and the temporal dynamics leading up to them empirically because the changes occur over very long time periods. This study investigates the age and magnitude of age- and time- shifting of incidence induced by either pre-erythrocytic vaccination (PEV) programmes or seasonal malaria chemo-prevention (SMC), using an ensemble of individual-based stochastic simulation models of P. falciparum dynamics. The models made various assumptions about immunity decay, transmission heterogeneity and were parameterized with data on both age-specific infection and disease incidence at different levels of exposure, on the durations of different stages of the parasite life-cycle and on human demography. Effects of transmission intensity, and of levels of access to malaria treatment were considered. While both PEV and SMC programmes are predicted to have overall strongly positive health effects, a shift of morbidity into older children is predicted to be induced by either programme if transmission levels remain static and not reduced by other interventions. Predicted shifting of burden continue into the second decade of the programme. Even if long-term surveillance is maintained it will be difficult to avoid mis-attribution of such long-term changes in age-specific morbidity patterns to other factors. Conversely, short-lived transient changes in incidence measured soon after introduction of a new intervention may give over-positive views of future impacts. Complementary intervention strategies could be designed to specifically protect those age-groups at risk from burden shift.


Antimicrobial Agents and Chemotherapy | 2015

Altering antimalarial drug regimens may dramatically enhance and restore drug effectiveness.

Katherine Kay; Eva Maria Hodel; Ian M. Hastings

ABSTRACT There is considerable concern that malaria parasites are starting to evolve resistance to the current generation of antimalarial drugs, the artemisinin-based combination therapies (ACTs). We use pharmacological modeling to investigate changes in ACT effectiveness likely to occur if current regimens are extended from 3 to 5 days or, alternatively, given twice daily over 3 days. We show that the pharmacology of artemisinins allows both regimen changes to substantially increase the artemisinin killing rate. Malaria patients rarely contain more than 1012 parasites, while the standard dosing regimens allow approximately 1 in 1010 parasites to survive artemisinin treatment. Parasite survival falls dramatically, to around 1 in 1017 parasites if the dose is extended or split; theoretically, this increase in drug killing appears to be more than sufficient to restore failing ACT efficacy. One of the most widely used dosing regimens, artemether-lumefantrine, already successfully employs a twice-daily dosing regimen, and we argue that twice-daily dosing should be incorporated into all ACT regimen design considerations as a simple and effective way of ensuring the continued long-term effectiveness of ACTs.


Antimicrobial Agents and Chemotherapy | 2016

Incorporating Stage-Specific Drug Action into Pharmacological Modeling of Antimalarial Drug Treatment

Eva Maria Hodel; Katherine Kay; Ian M. Hastings

ABSTRACT Pharmacological modeling of antiparasitic treatment based on a drugs pharmacokinetic and pharmacodynamic properties plays an increasingly important role in identifying optimal drug dosing regimens and predicting their potential impact on control and elimination programs. Conventional modeling of treatment relies on methods that do not distinguish between parasites at different developmental stages. This is problematic for malaria parasites, as their sensitivity to drugs varies substantially during their 48-h developmental cycle. We investigated four drug types (short or long half-lives with or without stage-specific killing) to quantify the accuracy of the standard methodology. The treatment dynamics of three drug types were well characterized with standard modeling. The exception were short-half-life drugs with stage-specific killing (i.e., artemisinins) because, depending on time of treatment, parasites might be in highly drug-sensitive stages or in much less sensitive stages. We describe how to bring such drugs into pharmacological modeling by including additional variation into the drugs maximal killing rate. Finally, we show that artemisinin kill rates may have been substantially overestimated in previous modeling studies because (i) the parasite reduction ratio (PRR) (generally estimated to be 104) is based on observed changes in circulating parasite numbers, which generally overestimate the “true” PRR, which should include both circulating and sequestered parasites, and (ii) the third dose of artemisinin at 48 h targets exactly those stages initially hit at time zero, so it is incorrect to extrapolate the PRR measured over 48 h to predict the impact of doses at 48 h and later.


Scientific Reports | 2016

Quantifying the pharmacology of antimalarial drug combination therapy

Ian M. Hastings; Eva Maria Hodel; Katherine Kay

Most current antimalarial drugs are combinations of an artemisinin plus a ‘partner’ drug from another class, and are known as artemisinin-based combination therapies (ACTs). They are the frontline drugs in treating human malaria infections. They also have a public-health role as an essential component of recent, comprehensive scale-ups of malaria interventions and containment efforts conceived as part of longer term malaria elimination efforts. Recent reports that resistance has arisen to artemisinins has caused considerable concern. We investigate the likely impact of artemisinin resistance by quantifying the contribution artemisinins make to the overall therapeutic capacity of ACTs. We achieve this using a simple, easily understood, algebraic approach and by more sophisticated pharmacokinetic/pharmacodynamic analyses of drug action; the two approaches gave consistent results. Surprisingly, the artemisinin component typically makes a negligible contribution (≪0.0001%) to the therapeutic capacity of the most widely used ACTs and only starts to make a significant contribution to therapeutic outcome once resistance has started to evolve to the partner drugs. The main threat to antimalarial drug effectiveness and control comes from resistance evolving to the partner drugs. We therefore argue that public health policies be re-focussed to maximise the likely long-term effectiveness of the partner drugs.


Antimicrobial Agents and Chemotherapy | 2014

Improving the Role and Contribution of Pharmacokinetic Analyses in Antimalarial Drug Clinical Trials

Katherine Kay; Eva Maria Hodel; Ian M. Hastings

ABSTRACT It is now World Health Organization (WHO) policy that drug concentrations on day 7 be measured as part of routine assessment in antimalarial drug efficacy trials. The rationale is that this single pharmacological measure serves as a simple and practical predictor of treatment outcome for antimalarial drugs with long half-lives. Herein we review theoretical data and field studies and conclude that the day 7 drug concentration (d7c) actually appears to be a poor predictor of therapeutic outcome. This poor predictive capability combined with the fact that many routine antimalarial trials will have few or no failures means that there appears to be little justification for this WHO recommendation. Pharmacological studies have a huge potential to improve antimalarial dosing, and we propose study designs that use more-focused, sophisticated, and cost-effective ways of generating these data than the mass collection of single d7c concentrations.


Malaria Journal | 2016

OptiMal-PK: an internet-based, user-friendly interface for the mathematical-based design of optimized anti-malarial treatment regimens

Ghaith Aljayyoussi; Katherine Kay; Stephen A. Ward; Giancarlo A. Biagini

BackgroundThe search for highly effective anti-malarial therapies has gathered pace and recent years have seen a number of promising single and combined therapies reach the late stages of development. A key drug development challenge is the need for early assessment of the clinical utility of new drug leads as it is often unclear for developers whether efforts should be focused on efficacy or metabolic stability/exposure or indeed whether the continuation of iterative QSAR (quantitative structure–activity and relationships) cycles of medicinal chemistry and biological testing will translate to improved clinical efficacy. Pharmacokinetic and pharmacodynamic (PK/PD)-based measurements available from in vitro studies can be used for such clinical predictions. However, these predictions often require bespoke mathematical PK/PD modelling expertise and are normally performed after candidate development and, therefore, not during the pre-clinical development phase when such decisions need to be made.MethodsAn internet-based tool has been developed using STELLA® software. The tool simulates multiple differential equations that describe anti-malarial PK/PD relationships where the user can easily input PK/PD parameters. The tool utilizes a simple stop-light system to indicate the efficacy of each combination of parameters. This tool, called OptiMal-PK, additionally allows for the investigation of the effect of drug combinations with known or custom compounds.ResultsThe results of simulations obtained from OptiMal-PK were compared to a previously published and validated mathematical model on which this tool is based. The tool has also been used to simulate the PK/PD relationship for a number of existing anti-malarial drugs in single or combined treatment. Simulations were predictive of the published clinical parasitological clearance activities for these existing therapies.ConclusionsOptiMal-PK is designed to be implemented by medicinal chemists and pharmacologists during the pre-clinical anti-malarial drug development phase to explore the impact of different PK/PD parameters upon the predicted clinical activity of any new compound. It can help investigators to identify which pharmacological features of a compound are most important to the clinical performance of a new chemical entity and how partner drugs could potentially improve the activity of existing therapies.


Malaria Journal | 2012

An in silico drug treatment model to assess the robustness of regional age-based dosing regimens for artemisinin-based combination therapies

Eva Maria Hodel; Katherine Kay; Daniel Hayes; Anja Terlouw; Ian M. Hastings

The standard drug development process for antimalarials and other drugs uses weight-based dosing (mg/kg) to predict blood concentrations of the drug, and hence their effect. Consequently, the current World Health Organization Guidelines for the treatment of malaria [1] provide target doses and therapeutic dose ranges in mg/ kg/day. However, in resource-poor settings, age-based dosing is often employed instead of weight-based dosing because of the scarcity of correctly functioning weighing scales outside of clinical settings. Due to the wide variation in weight by age this approach inevitably results in over- and under-dosing of a proportion of the population. We have recently developed a modelling method to create statistically robust global and regional malaria-specific weight-for-age references representative of the malaria-endemic countries [2] and employed it to predict optimized age-based regimens for artemisinin-based combination therapies (ACTs) for case management of uncomplicated malaria (unpublished). The presented work now assesses the robustness of these age-based regimens using an in silico model of antimalarial drug treatment to predict treatment outcome based on individual infection parameters such as parasite numbers, variation in patient pharmacokinetics, and parasite variation in their drug sensitivity [3]. This extended pharmacokinetic/pharmakodynamic model for ACTs allowed us to investigate extreme treatment scenarios in a large number of patients over long follow-up periods that for ethical reasons could not be applied in clinical trials: typical examples include poor adherence (e.g. delayed, reduced or missed doses) or administration of doses above or below recommended therapeutic dose ranges and particularly in most vulnerable individuals such as infants and young children. Pharmacological modelling of antimalarial treatment cannot replace the gold standard of clinical trials, but the model outputs can identify patient groups that are at higher risk of treatment failure due to under-dosing or adverse events due to over-dosing. We acknowledge the Medical Research Council for funding of this work.


Parasites & Vectors | 2018

Modelling the impact of insecticide-based control interventions on the evolution of insecticide resistance and disease transmission

Susana Barbosa; Katherine Kay; Nakul Chitnis; Ian M. Hastings

BackgroundCurrent strategies to control mosquito-transmitted infections use insecticides targeted at various stages of the mosquito life-cycle. Control is increasingly compromised by the evolution of insecticide resistance but there is little quantitative understanding of its impact on control effectiveness. We developed a computational approach that incorporates the stage-structured mosquito life-cycle and allows tracking of insecticide resistant genotypes. This approach makes it possible to simultaneously investigate: (i) the population dynamics of mosquitoes throughout their whole life-cycle; (ii) the impact of common vector control interventions on disease transmission; (iii) how these interventions drive the spread of insecticide resistance; and (iv) the impact of resistance once it has arisen and, in particular, whether it is sufficient for malaria transmission to resume. The model consists of a system of difference equations that tracks the immature (eggs, larvae and pupae) and adult stages, for males and females separately, and incorporates density-dependent regulation of mosquito larvae in breeding sites.ResultsWe determined a threshold level of mosquitoes below which transmission of malaria is interrupted. It is based on a classic Ross-Macdonald derivation of the malaria basic reproductive number (R0) and may be used to assess the effectiveness of different control strategies in terms of whether they are likely to interrupt disease transmission. We simulated different scenarios of insecticide deployment by changing key parameters in the model to explore the comparative impact of insecticide treated nets, indoor residual spraying and larvicides.ConclusionsOur simulated results suggest that relatively low degrees of resistance (in terms of reduced mortality following insecticide contact) can induce failure of interventions, and the rate of spread of resistance is faster when insecticides target the larval stages. The optimal disease control strategy depends on vector species demography and local environmental conditions but, in our illustrative parametrisation, targeting larval stages achieved the greatest reduction of the adult population, followed by targeting of non-host-seeking females, as provided by indoor residual spraying. Our approach is designed to be flexible and easily generalizable to many scenarios using different calibrations and to diseases other than malaria.


British Journal of Clinical Pharmacology | 2018

Physiologically‐based pharmacokinetic model of vaginally administered dapivirine ring and film formulations

Katherine Kay; Dhaval K. Shah; Lisa C. Rohan; Robert R. Bies

A physiologically‐based pharmacokinetic (PBPK) model of the vaginal space was developed with the aim of predicting concentrations in the vaginal and cervical space. These predictions can be used to optimize the probability of success of vaginally administered dapivirine (DPV) for HIV prevention. We focus on vaginal delivery using either a ring or film.

Collaboration


Dive into the Katherine Kay's collaboration.

Top Co-Authors

Avatar

Ian M. Hastings

Liverpool School of Tropical Medicine

View shared research outputs
Top Co-Authors

Avatar

Eva Maria Hodel

Liverpool School of Tropical Medicine

View shared research outputs
Top Co-Authors

Avatar

Daniel Hayes

Liverpool School of Tropical Medicine

View shared research outputs
Top Co-Authors

Avatar

Anja Terlouw

Liverpool School of Tropical Medicine

View shared research outputs
Top Co-Authors

Avatar

Dianne J Terlouw

Liverpool School of Tropical Medicine

View shared research outputs
Top Co-Authors

Avatar

Ghaith Aljayyoussi

Liverpool School of Tropical Medicine

View shared research outputs
Top Co-Authors

Avatar

Giancarlo A. Biagini

Liverpool School of Tropical Medicine

View shared research outputs
Top Co-Authors

Avatar

Stephen A. Ward

Liverpool School of Tropical Medicine

View shared research outputs
Top Co-Authors

Avatar

Susana Barbosa

Liverpool School of Tropical Medicine

View shared research outputs
Top Co-Authors

Avatar

Melissa A. Penny

Swiss Tropical and Public Health Institute

View shared research outputs
Researchain Logo
Decentralizing Knowledge