Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Clin Pharmacol Ther ; 113(3): 660-669, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36260349

RESUMEN

Artemisinin-based combination therapies (ACTs) are the primary treatment for malaria. It is essential to characterize the pharmacokinetics (PKs) and pharmacodynamics (PDs) of ACTs in vulnerable populations at risk of suboptimal dosing. We developed a population PK/PD model using data from our previous study of artemether-lumefantrine in HIV-uninfected and HIV-infected children living in a high-transmission region of Uganda. HIV-infected children were on efavirenz-, nevirapine-, or lopinavir-ritonavir-based antiretroviral regimens, with daily trimethoprim-sulfamethoxazole prophylaxis. We assessed selection for resistance in two key parasite transporters, pfcrt and pfmdr1, over 42-day follow-up and incorporated genotyping into a time-to-event model to ascertain how resistance genotype in relation to drug exposure impacts recurrence risk. Two hundred seventy-seven children contributed 364 episodes to the model (186 HIV-uninfected and 178 HIV-infected), with recurrent microscopy-detectable parasitemia detected in 176 episodes by day 42. The final model was a two-compartment model with first-order absorption and an estimated age effect on bioavailability. Systemic lumefantrine exposure was highest with lopinavir-ritonavir, lowest with efavirenz, and equivalent with nevirapine and HIV-uninfected children. HIV status and lumefantrine concentration were significant factors associated with recurrence risk. Significant selection was demonstrated for pfmdr1 N86 and pfcrt K76 in recurrent infections, with no evidence of selection for pfmdr1 Y184F. Less sensitive parasites were able to tolerate lumefantrine concentrations ~ 3.5-fold higher than more sensitive parasites. This is the first population PK model of lumefantrine in HIV-infected children and demonstrates selection for reduced lumefantrine susceptibility, a concern as we confront the threat to ACTs posed by emerging artemisinin resistance in Africa.


Asunto(s)
Antimaláricos , Artemisininas , Infecciones por VIH , Malaria Falciparum , Malaria , Niño , Humanos , Antimaláricos/uso terapéutico , Antimaláricos/farmacocinética , Lopinavir/uso terapéutico , Ritonavir/uso terapéutico , Arteméter/uso terapéutico , Nevirapina/uso terapéutico , Uganda , Fluorenos/uso terapéutico , Fluorenos/farmacocinética , Combinación Arteméter y Lumefantrina/uso terapéutico , Malaria/tratamiento farmacológico , Artemisininas/farmacocinética , Lumefantrina , Combinación de Medicamentos , Infecciones por VIH/tratamiento farmacológico , Malaria Falciparum/tratamiento farmacológico
2.
Antimicrob Agents Chemother ; 65(10): e0043721, 2021 09 17.
Artículo en Inglés | MEDLINE | ID: mdl-34252299

RESUMEN

Regulatory clinical trials are required to ensure the continued supply and deployment of effective antimalarial drugs. Patient follow-up in such trials typically lasts several weeks, as the drugs have long half-lives and new infections often occur during this period. "Molecular correction" is therefore used to distinguish drug failures from new infections. The current WHO-recommended method for molecular correction uses length-polymorphic alleles at highly diverse loci but is inherently poor at detecting low-density clones in polyclonal infections. This likely leads to substantial underestimates of failure rates, delaying the replacement of failing drugs with potentially lethal consequences. Deep-sequenced amplicons (AmpSeq) substantially increase the detectability of low-density clones and may offer a new "gold standard" for molecular correction. Pharmacological simulation of clinical trials was used to evaluate the suitability of AmpSeq for molecular correction. We investigated the impact of factors such as the number of amplicon loci analyzed, the informatics criteria used to distinguish genotyping "noise" from real low-density signals, the local epidemiology of malaria transmission, and the potential impact of genetic signals from gametocytes. AmpSeq greatly improved molecular correction and provided accurate drug failure rate estimates. The use of 3 to 5 amplicons was sufficient, and simple, nonstatistical criteria could be used to classify recurrent infections as drug failures or new infections. These results suggest AmpSeq is strongly placed to become the new standard for molecular correction in regulatory trials, with potential extension into routine surveillance once the requisite technical support becomes established.


Asunto(s)
Antimaláricos , Malaria Falciparum , Malaria , Preparaciones Farmacéuticas , Antimaláricos/uso terapéutico , Humanos , Malaria/tratamiento farmacológico , Malaria Falciparum/tratamiento farmacológico , Plasmodium falciparum/genética
3.
Evol Appl ; 13(10): 2723-2739, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-33294019

RESUMEN

INTRODUCTION: Control strategies for human infections are often investigated using individual-based models (IBMs) to quantify their impact in terms of mortality, morbidity and impact on transmission. Genetic selection can be incorporated into the IBMs to track the spread of mutations whose origin and spread are driven by the intervention and which subsequently undermine the control strategy; typical examples are mutations which encode drug resistance or diagnosis- or vaccine-escape phenotypes. METHODS AND RESULTS: We simulated the spread of malaria drug resistance using the IBM OpenMalaria to investigate how the finite sizes of IBMs require strategies to optimally incorporate genetic selection. We make four recommendations. Firstly, calculate and report the selection coefficients, s, of the advantageous allele as the key genetic parameter. Secondly, use these values of "s" to calculate the wait time until a mutation successfully establishes itself in the pathogen population. Thirdly, identify the inherent limits of the IBM to robustly estimate small selection coefficients. Fourthly, optimize computational efficacy: when "s" is small, fewer replicates of larger IBMs may be more efficient than a larger number of replicates of smaller size. DISCUSSION: The OpenMalaria IBM of malaria was an exemplar and the same principles apply to IBMs of other diseases.

4.
Artículo en Inglés | MEDLINE | ID: mdl-31932376

RESUMEN

Antimalarial drugs have long half-lives, so clinical trials to monitor their efficacy require long periods of follow-up to capture drug failure that may become patent only weeks after treatment. Reinfections often occur during follow-up, so robust methods of distinguishing drug failures (recrudescence) from emerging new infections are needed to produce accurate failure rate estimates. Molecular correction aims to achieve this by comparing the genotype of a patient's pretreatment (initial) blood sample with that of any infection that occurs during follow-up, with matching genotypes indicating drug failure. We use an in silico approach to show that the widely used match-counting method of molecular correction with microsatellite markers is likely to be highly unreliable and may lead to gross under- or overestimates of the true failure rates, depending on the choice of matching criterion. A Bayesian algorithm for molecular correction was previously developed and utilized for analysis of in vivo efficacy trials. We validated this algorithm using in silico data and showed it had high specificity and generated accurate failure rate estimates. This conclusion was robust for multiple drugs, different levels of drug failure rates, different levels of transmission intensity in the study sites, and microsatellite genetic diversity. The Bayesian algorithm was inherently unable to accurately identify low-density recrudescence that occurred in a small number of patients, but this did not appear to compromise its utility as a highly effective molecular correction method for analyzing microsatellite genotypes. Strong consideration should be given to using Bayesian methodology to obtain accurate failure rate estimates during routine monitoring trials of antimalarial efficacy that use microsatellite markers.


Asunto(s)
Antimaláricos/uso terapéutico , Biología Computacional/métodos , Malaria Falciparum/tratamiento farmacológico , Repeticiones de Microsatélite/genética , Plasmodium falciparum/efectos de los fármacos , Algoritmos , Combinación Arteméter y Lumefantrina/uso terapéutico , Artesunato/uso terapéutico , Simulación por Computador , Interpretación Estadística de Datos , Femenino , Humanos , Masculino , Mefloquina/uso terapéutico , Plasmodium falciparum/genética , Reinfección/genética , Reinfección/parasitología , Insuficiencia del Tratamiento
5.
AAPS J ; 21(2): 27, 2019 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-30737615

RESUMEN

Tumor doubling time can significantly affect the outcome of anticancer therapy, but it is very challenging to determine. Here, we present a statistical approach that extracts doubling times from progression-free survival (PFS) plots, which inherently contains information regarding the growth of solid tumors. Twelve cancers were investigated and multiple PFS plots were evaluated for each type. The PFS plot showing fastest tumor growth was deemed to best represent the inherent growth kinetics of the solid tumor, and selected for further analysis. The exponential tumor growth rates were extracted from each PFS plot, along with associated variabilities, which ultimately allowed for the estimation of solid tumor doubling times. The mean simulated doubling times for pancreatic cancer, melanoma, hepatocellular carcinoma (HCC), renal cell carcinoma, triple negative breast cancer, non-small cell lung cancer, hormone receptor positive (HR+) breast cancer, human epidermal growth factor receptor-2 positive (HER-2+) breast cancer, gastric cancer, glioblastoma multiforme, colorectal cancer, and prostate cancer were 5.06, 3.78, 3.06, 2.67, 2.38, 2.40, 4.31, 4.12, and 3.84 months, respectively. For all cancers, clinically reported doubling times were within the estimated ranges. For all cancers, except HCC, the growth rates were best characterized by a log-normal distribution. For HCC, the gamma distribution best described the data. The statistical approach presented here provides a qualified method for extracting tumor growth rates and doubling times from PFS plots. It also allows estimation of the distributional characteristics for tumor growth rates and doubling times in a given patient population.


Asunto(s)
Antineoplásicos/farmacología , Modelos Biológicos , Neoplasias/mortalidad , Distribuciones Estadísticas , Antineoplásicos/uso terapéutico , Ensayos Clínicos como Asunto/estadística & datos numéricos , Interpretación Estadística de Datos , Conjuntos de Datos como Asunto , Femenino , Humanos , Estimación de Kaplan-Meier , Masculino , Neoplasias/tratamiento farmacológico , Supervivencia sin Progresión , Factores de Tiempo
6.
Front Pharmacol ; 10: 1514, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-32082142

RESUMEN

Pre-exposure prophylaxis (PrEP) has emerged as a promising strategy for preventing the transmission of HIV. Although only one formulation is currently approved for PrEP, research into both new compounds and new delivery systems for PrEP regimens offer intriguing challenges from the perspective of pharmacokinetic and pharmacodynamic modeling. This review aims to provide an overview the current modeling landscape for HIV PrEP, focused on PK/PD and QSP models relating to antiretroviral agents. Both current PrEP treatments and new compounds that show promise as PrEP agents are highlighted, as well as models of uncommon administration routes, predictions based on models of mechanism of action and viral dynamics, and issues related to adherence to therapy. The spread of human immunodeficiency virus (HIV) remains one of the foremost global health concerns. In the absence of a vaccine, other prophylactic strategies have been developed to prevent HIV transmission. One approach, known as pre-exposure prophylaxis (PrEP), allows HIV-negative individuals who are at high risk of exposure to the virus, be it through an HIV-positive sexual partner or through the shared use of drug injection equipment, to substantially reduce the risk of developing an HIV infection. PrEP is a relatively recent approach to combating the HIV epidemic, with the only currently approved treatment being Truvada, a daily oral antiretroviral (ARV) therapy initially indicated in the treatment of active HIV-1 infections, but approved for HIV PrEP in 2012. Although PrEP therapy has consistently demonstrated high efficacy in preventing HIV infection, this efficacy is dependent on patient adherence to the prescribed treatment regimen. This can present a significant problem in low- and middle-income countries, which may lack the infrastructure to provide sufficient access to PrEP medication to maintain daily dosing regimens. Furthermore, while the conventional approach has generally been to advocate for continuous administration akin to regimens used for viral suppression in infected patients, there has been some discussion of whether a better treatment paradigm might be to push for PrEP therapy primarily during those known periods of heightened exposure risk, while relying on post-exposure prophylaxis regimens to prevent infection after unanticipated exposures during low-risk periods. These considerations have led to a push for the development of long-duration and on-demand PrEP formulations, including subdermal and subcutaneous implants, slow-release intramuscular depot injections, vaginal and rectal antimicrobial gels, and intravaginal rings and dissolving films. PrEP therapy is a quickly evolving field, with a variety of antiretroviral compounds and formulations under investigation. This review aims to report on notable drugs and formulations from a pharmacokinetic/pharmacodynamic (PK/PD) modeling perspective. Given the nature of PrEP as a preventive therapy designed for long-term use, clinical trials for PrEP therapies can last for months or even years, particularly in the case of long-duration formulations. Furthermore, in contrast to antiretroviral trials in infected patients, pharmacodynamic endpoints in PrEP therapies are difficult to quantify, as the primary endpoint for efficacy is generally the rate of seroconversion. Computational modeling approaches offer flexible and powerful tools to provide insight into drug behavior in clinical settings, and can ultimately reduce the time, expense, and patient burden incurred in the development of PrEP therapies.

7.
J Infect Dis ; 219(8): 1243-1253, 2019 04 08.
Artículo en Inglés | MEDLINE | ID: mdl-30517708

RESUMEN

BACKGROUND: Standard treatment for severe malaria is with artesunate; patient survival in the 24 hours immediately posttreatment is the key objective. Clinical trials use clearance rates of circulating parasites as their clinical outcome, but the pathology of severe malaria is attributed primarily to noncirculating, sequestered, parasites, so there is a disconnect between existing clinical metrics and objectives. METHODS: We extend existing pharmacokinetic/pharmacodynamic modeling methods to simulate the treatment of 10000 patients with severe malaria and track the pathology caused by sequestered parasites. RESULTS: Our model recovered the clinical outcomes of existing studies (based on circulating parasites) and showed a "simplified" artesunate regimen was noninferior to the existing World Health Organization regimen across the patient population but resulted in worse outcomes in a subgroup of patients with infections clustered in early stages of the parasite life cycle. This same group of patients were extremely vulnerable to resistance emerging in parasite early ring stages. CONCLUSIONS: We quantify patient outcomes in a manner appropriate for severe malaria with a flexible framework that allows future researchers to implement their beliefs about underlying pathology. We highlight with some urgency the threat posed to treatment of severe malaria by artemisinin resistance in parasite early ring stages.


Asunto(s)
Antimaláricos/uso terapéutico , Artemisininas/uso terapéutico , Malaria Falciparum/tratamiento farmacológico , Enfermedad Aguda , Antimaláricos/administración & dosificación , Artesunato/administración & dosificación , Artesunato/uso terapéutico , Resistencia a Medicamentos , Humanos , Malaria Falciparum/parasitología , Malaria Falciparum/patología , Modelos Biológicos , Parasitemia/tratamiento farmacológico , Parasitemia/parasitología , Plasmodium falciparum/efectos de los fármacos , Resultado del Tratamiento
8.
Parasit Vectors ; 11(1): 482, 2018 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-30153869

RESUMEN

BACKGROUND: Current 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. RESULTS: We 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. CONCLUSIONS: Our 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.


Asunto(s)
Simulación por Computador , Resistencia a los Insecticidas , Insecticidas/farmacología , Malaria/transmisión , Animales , Anopheles/efectos de los fármacos , Anopheles/genética , Femenino , Humanos , Mosquiteros Tratados con Insecticida/efectos adversos , Larva/efectos de los fármacos , Estadios del Ciclo de Vida/efectos de los fármacos , Malaria/epidemiología , Malaria/prevención & control , Masculino , Control de Mosquitos/métodos , Mosquitos Vectores/efectos de los fármacos , Dinámica Poblacional , Piretrinas/farmacología , Reproducción
9.
Br J Clin Pharmacol ; 84(9): 1950-1969, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29714824

RESUMEN

AIMS: 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. METHODS: A PBPK model describing the physiological structure of the vaginal tissue and fluid was defined mathematically and implemented in MATLAB. Literature reviews provided estimates for relevant physiological and physiochemical parameters. Drug concentration-time profiles were simulated in luminal fluids, vaginal tissue and plasma after administration of ring or film. Patient data were extracted from published clinical trials and used to test model predictions. RESULTS: The DPV ring simulations tested the two dosing regimens and predicted PK profiles and area under the curve of luminal fluids (29 079 and 33 067 mg h l-1 in groups A and B, respectively) and plasma (0.177 and 0.211 mg h l-1 ) closely matched those reported (within one standard deviation). While the DPV film study reported drug concentration at only one time point per patient, our simulated profiles pass through reported concentration range. CONCLUSIONS: HIV is a major public health issue and vaginal microbicides have the potential to provide a crucial, female-controlled option for protection. The PBPK model successfully simulated realistic representations of drug PK. It provides a reliable, inexpensive and accessible platform where potential effectiveness of new compounds and the robustness of treatment modalities for pre-exposure prophylaxis can be evaluated.


Asunto(s)
Fármacos Anti-VIH/farmacocinética , Infecciones por VIH/tratamiento farmacológico , Modelos Biológicos , Pirimidinas/farmacocinética , Administración Intravaginal , Fármacos Anti-VIH/administración & dosificación , Femenino , Humanos , Pirimidinas/administración & dosificación , Distribución Tisular , Vagina/metabolismo
10.
Sci Rep ; 6: 32762, 2016 09 08.
Artículo en Inglés | MEDLINE | ID: mdl-27604175

RESUMEN

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.


Asunto(s)
Antimaláricos/farmacología , Antimaláricos/uso terapéutico , Artemisininas/farmacología , Antimaláricos/farmacocinética , Artemisininas/farmacocinética , Artemisininas/uso terapéutico , Farmacorresistencia Microbiana/efectos de los fármacos , Quimioterapia Combinada , Humanos , Modelos Biológicos
12.
Malar J ; 15(1): 344, 2016 07 07.
Artículo en Inglés | MEDLINE | ID: mdl-27388207

RESUMEN

BACKGROUND: The 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. METHODS: An 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. RESULTS: The 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. CONCLUSIONS: OptiMal-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.


Asunto(s)
Antimaláricos/farmacología , Antimaláricos/farmacocinética , Evaluación Preclínica de Medicamentos/métodos , Internet , Malaria/tratamiento farmacológico , Programas Informáticos , Antimaláricos/aislamiento & purificación , Humanos , Modelos Teóricos
13.
Antimicrob Agents Chemother ; 60(5): 2747-56, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-26902760

RESUMEN

Pharmacological modeling of antiparasitic treatment based on a drug's 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 drug's 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 10(4)) 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.


Asunto(s)
Antimaláricos/farmacocinética , Antimaláricos/uso terapéutico , Relación Dosis-Respuesta a Droga , Malaria/tratamiento farmacológico , Modelos Teóricos
15.
Antimicrob Agents Chemother ; 59(10): 6428-36, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26239987

RESUMEN

Artemisinin-based combination therapies (ACTs) are currently the first-line drugs for treating uncomplicated falciparum malaria, the most deadly of the human malarias. Malaria parasite clearance rates estimated from patients' blood following ACT treatment have been widely adopted as a measure of drug effectiveness and as surveillance tools for detecting the presence of potential artemisinin resistance. This metric has not been investigated in detail, nor have its properties or potential shortcomings been identified. Herein, the pharmacology of drug treatment, parasite biology, and human immunity are combined to investigate the dynamics of parasite clearance following ACT. This approach parsimoniously recovers the principal clinical features and dynamics of clearance. Human immunity is the primary determinant of clearance rates, unless or until artemisinin killing has fallen to near-ineffective levels. Clearance rates are therefore highly insensitive metrics for surveillance that may lead to overconfidence, as even quite substantial reductions in drug sensitivity may not be detected as lower clearance rates. Equally serious is the use of clearance rates to quantify the impact of ACT regimen changes, as this strategy will plausibly miss even very substantial increases in drug effectiveness. In particular, the malaria community may be missing the opportunity to dramatically increase ACT effectiveness through regimen changes, particularly through a switch to twice-daily regimens and/or increases in artemisinin dosing levels. The malaria community therefore appears overreliant on a single metric of drug effectiveness, the parasite clearance rate, that has significant and serious shortcomings.


Asunto(s)
Antimaláricos/farmacología , Artemisininas/farmacología , Malaria Falciparum/tratamiento farmacológico , Modelos Estadísticos , Parasitemia/tratamiento farmacológico , Plasmodium falciparum/efectos de los fármacos , Adulto , Antimaláricos/farmacocinética , Artemisininas/farmacocinética , Artesunato , Niño , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Combinación de Medicamentos , Cálculo de Dosificación de Drogas , Resistencia a Medicamentos , Humanos , Inmunidad Innata , Malaria Falciparum/diagnóstico , Malaria Falciparum/inmunología , Malaria Falciparum/parasitología , Mefloquina/farmacocinética , Mefloquina/farmacología , Parasitemia/diagnóstico , Parasitemia/inmunología , Pruebas de Sensibilidad Parasitaria , Plasmodium falciparum/fisiología , Quinolinas/farmacocinética , Quinolinas/farmacología , Resultado del Tratamiento
16.
Antimicrob Agents Chemother ; 59(10): 6419-27, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26239993

RESUMEN

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 10(12) parasites, while the standard dosing regimens allow approximately 1 in 10(10) parasites to survive artemisinin treatment. Parasite survival falls dramatically, to around 1 in 10(17) 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.


Asunto(s)
Antimaláricos/farmacocinética , Artemisininas/farmacocinética , Etanolaminas/farmacocinética , Fluorenos/farmacocinética , Malaria Falciparum/tratamiento farmacológico , Modelos Estadísticos , Plasmodium falciparum/efectos de los fármacos , Adulto , Antimaláricos/farmacología , Combinación Arteméter y Lumefantrina , Artemisininas/farmacología , Niño , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Combinación de Medicamentos , Cálculo de Dosificación de Drogas , Etanolaminas/farmacología , Femenino , Fluorenos/farmacología , Humanos , Malaria Falciparum/parasitología , Masculino , Pruebas de Sensibilidad Parasitaria , Plasmodium falciparum/fisiología , Embarazo , Resultado del Tratamiento
17.
Malar J ; 14: 292, 2015 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-26228915

RESUMEN

BACKGROUND: The long half-lives of malaria 'partner' drugs are a potent force selecting for drug resistance. Clinical trials can quantify this effect by estimating a window of selection (WoS), defined as the amount of time post-treatment when drug levels are sufficiently high that resistant parasites can re-establish an infection while preventing drug-sensitive parasites from establishing viable infections. METHODS: The ability of clinical data to accurately estimate the true WoS was investigated using standard pharmacokinetic-pharmacodynamic models for three widely used malaria drugs: artemether-lumefantrine (AR-LF), artesunate-mefloquine (AS-MQ) and dihydroartemisinin-piperaquine (DHA-PPQ). Estimates of the clinical WoS either (1) ignored all new infections occurring after the 63-day follow-up period, as is currently done in clinical trials, or, (2) recognized that all individuals would eventually be re-infected and arbitrarily assigned them a new infection day. RESULTS: The results suggest current methods of estimating the clinical WoS underestimate the true WoS by as much as 9 days for AR-LF, 33 days for AS-MQ and 7 days for DHA-PPQ. The new method of estimating clinical WoS (i.e., retaining all individuals in the analysis) was significantly better at estimating the true WoS for AR-LF and AS-MQ. CONCLUSIONS: Previous studies, based on clinically observed WoS, have probably underestimated the 'true' WoS and hence the role of drugs with long half-lives in driving resistance. This has important policy implications: high levels of drug use are inevitable in mass drug administration programmes and intermittent preventative treatment programmes and the analysis herein suggests these policies will be far more potent drivers of resistance than previously thought.


Asunto(s)
Antimaláricos , Malaria Falciparum , Plasmodium falciparum/efectos de los fármacos , Antimaláricos/administración & dosificación , Antimaláricos/farmacocinética , Antimaláricos/farmacología , Antimaláricos/uso terapéutico , Combinación Arteméter y Lumefantrina , Artemisininas/administración & dosificación , Artemisininas/farmacocinética , Artemisininas/farmacología , Artemisininas/uso terapéutico , Combinación de Medicamentos , Etanolaminas/administración & dosificación , Etanolaminas/farmacocinética , Etanolaminas/farmacología , Etanolaminas/uso terapéutico , Fluorenos/administración & dosificación , Fluorenos/farmacocinética , Fluorenos/farmacología , Fluorenos/uso terapéutico , Humanos , Malaria Falciparum/tratamiento farmacológico , Malaria Falciparum/parasitología , Modelos Biológicos , Quinolinas/administración & dosificación , Quinolinas/farmacocinética , Quinolinas/farmacología , Quinolinas/uso terapéutico
18.
Malar J ; 14: 287, 2015 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-26206255

RESUMEN

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.


Asunto(s)
Quimioprevención , Malaria Falciparum/epidemiología , Malaria Falciparum/inmunología , Vacunación Masiva , Adolescente , Adulto , Factores de Edad , Anciano , Anciano de 80 o más Años , Niño , Preescolar , Simulación por Computador , Humanos , Incidencia , Lactante , Recién Nacido , Malaria Falciparum/parasitología , Malaria Falciparum/prevención & control , Persona de Mediana Edad , Modelos Teóricos , Estaciones del Año , Adulto Joven
19.
Antimicrob Agents Chemother ; 58(10): 5643-9, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24982091

RESUMEN

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.


Asunto(s)
Antimaláricos/farmacocinética , Antimaláricos/uso terapéutico , Artemisininas/farmacocinética , Artemisininas/uso terapéutico , Ensayos Clínicos como Asunto , Evaluación de Medicamentos , Humanos , Quinolinas/farmacocinética , Quinolinas/uso terapéutico
20.
Malar J ; 13: 138, 2014 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-24708571

RESUMEN

BACKGROUND: Successful 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. METHODS: An 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. RESULTS: Compared 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. CONCLUSION: Pharmacological 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.


Asunto(s)
Antimaláricos/administración & dosificación , Artemisininas/administración & dosificación , Etanolaminas/administración & dosificación , Fluorenos/administración & dosificación , Malaria/tratamiento farmacológico , Quinolinas/administración & dosificación , Adolescente , Adulto , Antimaláricos/farmacocinética , Combinación Arteméter y Lumefantrina , Artemisininas/farmacocinética , Niño , Preescolar , Simulación por Computador , Combinación de Medicamentos , Etanolaminas/farmacocinética , Femenino , Fluorenos/farmacocinética , Humanos , Lactante , Masculino , Quinolinas/farmacocinética , Resultado del Tratamiento , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...